Gut Antibody Deficiency in a Mouse Model of CVID Results in Spontaneous Development of a Gluten-Sensitive Enteropathy.
Journal: 2019/November - Frontiers in Immunology
ISSN: 1664-3224
Abstract:
Primary immunodeficiencies are heritable disorders of immune function. CD19 is a B cell co-receptor important for B cell development, and CD19 deficiency is a known genetic risk factor for a rare form of primary immunodeficiency known as "common variable immunodeficiency" (CVID); an antibody deficiency resulting in low levels of serum IgG and IgA. Enteropathies are commonly observed in CVID patients but the underlying reason for this is undefined. Here, we utilize CD19-/- mice as a model of CVID to test the hypothesis that antibody deficiency negatively impacts gut physiology under steady-state conditions. As anticipated, immune phenotyping experiments demonstrate that CD19-/- mice develop a severe B cell deficiency in gut-associated lymphoid tissues that result in significant reductions to antibody concentrations in the gut lumen. Antibody deficiency was associated with defective anti-commensal IgA responses and the outgrowth of anaerobic bacteria in the gut. Expansion of anaerobic bacteria coincides with the development of a chronic inflammatory condition in the gut of CD19-/- mice that results in an intestinal malabsorption characterized by defects in lipid metabolism and transport. Administration of the antibiotic metronidazole to target anaerobic members of the microbiota rescues mice from disease indicating that intestinal malabsorption is a microbiota-dependent phenomenon. Finally, intestinal malabsorption in CD19-/- mice is a gluten-sensitive enteropathy as exposure to a gluten-free diet also significantly reduces disease severity in CD19-/- mice. Collectively, these results support an effect of antibody deficiency on steady-state gut physiology that compliment emerging data from human studies linking IgA deficiency with non-infectious complications associated with CVID. They also demonstrate that CD19-/- mice are a useful model for studying the role of B cell deficiency and gut dysbiosis on gluten-sensitive enteropathies; a rapidly emerging group of diseases in humans with an unknown etiology.
Relations:
Content
Citations
(5)
References
(77)
Diseases
(2)
Conditions
(2)
Drugs
(2)
Organisms
(3)
Processes
(2)
Anatomy
(2)
Affiliates
(2)
Similar articles
Articles by the same authors
Discussion board
Front Immunol 10: 2484

Gut Antibody Deficiency in a Mouse Model of CVID Results in Spontaneous Development of a Gluten-Sensitive Enteropathy

+7 authors

Supplementary Table 1

List of antibodies used.

Click here for additional data file.(32K, pdf)

Supplementary Table 2

Differentially enriched fecal OTUs.

Click here for additional data file.(17K, xlsx)

Supplementary Table 3

DE Ileal OTUs.

Click here for additional data file.(11K, xlsx)

Supplementary Table 4

Differentially enriched ileal transcripts.

Click here for additional data file.(23K, xlsx)

Supplementary Table 5

Differentially enriched OTUs in metronidazole-treated mice.

Click here for additional data file.(18K, xlsx)
Click here for additional data file.(8.3M, pdf)
Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
School of Veterinary Medicine, University of Baghdad, Baghdad, Iraq
Biology Department, University of Texas at Arlington, Arlington, TX, United States
Department of Biomedical Sciences, College of Medicine, University of Houston, Houston, TX, United States
College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, United States
Edited by: Cecil Czerkinsky, Institut National de la Santé et de la Recherche Médicale (INSERM), France
Reviewed by: Karol Sestak, PreCliniTria, LLC., United States; Katri Lindfors, University of Tampere, Finland; Natalia Shulzhenko, Oregon State University, United States
*Correspondence: Jason L. Kubinak ude.cs.demcsu@kanibuK.nosaJ
This article was submitted to Mucosal Immunity, a section of the journal Frontiers in Immunology
Edited by: Cecil Czerkinsky, Institut National de la Santé et de la Recherche Médicale (INSERM), France
Reviewed by: Karol Sestak, PreCliniTria, LLC., United States; Katri Lindfors, University of Tampere, Finland; Natalia Shulzhenko, Oregon State University, United States
Received 2019 May 28; Accepted 2019 Oct 4.
This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

Abstract

Primary immunodeficiencies are heritable disorders of immune function. CD19 is a B cell co-receptor important for B cell development, and CD19 deficiency is a known genetic risk factor for a rare form of primary immunodeficiency known as “common variable immunodeficiency” (CVID); an antibody deficiency resulting in low levels of serum IgG and IgA. Enteropathies are commonly observed in CVID patients but the underlying reason for this is undefined. Here, we utilize CD19 mice as a model of CVID to test the hypothesis that antibody deficiency negatively impacts gut physiology under steady-state conditions. As anticipated, immune phenotyping experiments demonstrate that CD19 mice develop a severe B cell deficiency in gut-associated lymphoid tissues that result in significant reductions to antibody concentrations in the gut lumen. Antibody deficiency was associated with defective anti-commensal IgA responses and the outgrowth of anaerobic bacteria in the gut. Expansion of anaerobic bacteria coincides with the development of a chronic inflammatory condition in the gut of CD19 mice that results in an intestinal malabsorption characterized by defects in lipid metabolism and transport. Administration of the antibiotic metronidazole to target anaerobic members of the microbiota rescues mice from disease indicating that intestinal malabsorption is a microbiota-dependent phenomenon. Finally, intestinal malabsorption in CD19 mice is a gluten-sensitive enteropathy as exposure to a gluten-free diet also significantly reduces disease severity in CD19 mice. Collectively, these results support an effect of antibody deficiency on steady-state gut physiology that compliment emerging data from human studies linking IgA deficiency with non-infectious complications associated with CVID. They also demonstrate that CD19 mice are a useful model for studying the role of B cell deficiency and gut dysbiosis on gluten-sensitive enteropathies; a rapidly emerging group of diseases in humans with an unknown etiology.

Keywords: CVID, primary immunodeficiency, dysbiosis, CVID enteropathy, gluten sensitivity
Abstract

Acknowledgments

We would like to thank Dr. June L. Round for her useful comments during manuscript preparation.

Acknowledgments

Footnotes

Funding. JK was supported by a NIAID K22 Award, NIH grant R21AI142409, NIH grant P20GM103641 (awarded to Prakash and Mitzi Nagarkatti), a Jeffrey Modell Foundation Specific Diseases Research Award, and a University of South Carolina ASPIRE-I Award. ADM was supported by the Iraqi Government through participation in the MOHESR Program. CO was supported by NIH/NIAMS R21AR067996.

Footnotes
Click here for additional data file.(32K, pdf)Click here for additional data file.(17K, xlsx)Click here for additional data file.(11K, xlsx)Click here for additional data file.(23K, xlsx)Click here for additional data file.(18K, xlsx)Click here for additional data file.(8.3M, pdf)

References

  • 1. Durandy A, Kracker S, Fischer A. Primary antibody deficiencies. Nat Rev Immunol. (2013) 13:519–33. 10.1038/nri3466 [] [[PubMed]
  • 2. Bonilla FA, Khan DA, Ballas ZK, Chinen J, Frank MM, Hsu JT, et al. . Practice parameter for the diagnosis and management of primary immunodeficiency. J Allergy Clin Immunol. (2015) 136:1186–205. e1181–78. 10.1016/j.jaci.2015.04.049 [] [[PubMed]
  • 3. Lilic D, Sewell WA. IgA deficiency: what we should-or should not-be doing. J Clin Pathol. (2001) 54:337–8. 10.1136/jcp.54.5.337 ] [
  • 4. Cunningham-Rundles C. Physiology of IgA and IgA deficiency. J Clin Immunol. (2001) 21:303–9. 10.1023/A:1012241117984 [] [[PubMed]
  • 5. Cunningham-Rundles C. The many faces of common variable immunodeficiency. Hematol Am Soc Hematol Educ Program. (2012) 2012:301–5. 10.1182/asheducation-2012.1.301 ] [
  • 6. Azizi G, Bagheri Y, Tavakol M, Askarimoghaddam F, Porrostami K, Rafiemanesh H, et al. . The clinical and immunological features of patients with primary antibody deficiencies. Endocr Metab Immune Disord Drug Targets. (2018) 18:537–45. 10.2174/1871530318666180413110216 [] [[PubMed]
  • 7. Uzzan M, Ko HM, Mehandru S, Cunningham-Rundles C. Gastrointestinal Disorders Associated with Common Variable Immune Deficiency (CVID) and Chronic Granulomatous Disease (CGD). Curr Gastroenterol Rep. (2016) 18:17. 10.1007/s11894-016-0491-3 ] [
  • 8. Kaetzel CS, Blanch VJ, Hempen PM, Phillips KM, Piskurich JF, Youngman KR. The polymeric immunoglobulin receptor: structure and synthesis. Biochem Soc Trans. (1997) 25:475–80. 10.1042/bst0250475 [] [[PubMed]
  • 9. Kaetzel CS. The polymeric immunoglobulin receptor: bridging innate and adaptive immune responses at mucosal surfaces. Immunol Rev. (2005) 206:83–99. 10.1111/j.0105-2896.2005.00278.x [] [[PubMed]
  • 10. Phalipon A, Cardona A, Kraehenbuhl JP, Edelman L, Sansonetti PJ, Corthésy B. Secretory component: a new role in secretory IgA-mediated immune exclusion in vivo. Immunity. (2002) 17:107–15. 10.1016/S1074-7613(02)00341-2 [] [[PubMed]
  • 11. Mantis NJ, Forbes SJ. Secretory IgA: arresting microbial pathogens at epithelial borders. Immunol Invest. (2010) 39:383–406. 10.3109/08820131003622635 ] [
  • 12. Mantis NJ, Rol N, Corthésy B. Secretory IgA's complex roles in immunity and mucosal homeostasis in the gut. Mucosal Immunol. (2011) 4:603–11. 10.1038/mi.2011.41 ] [
  • 13. Kubinak JL, Round JL. Do antibodies select a healthy microbiota?Nat Rev Immunol. (2016) 16:767–74. 10.1038/nri.2016.114 [] [[PubMed]
  • 14. Jørgensen SF, Trøseid M, Kummen M, Anmarkrud JA, Michelsen AE, Osnes LT, et al. . Altered gut microbiota profile in common variable immunodeficiency associates with levels of lipopolysaccharide and markers of systemic immune activation. Mucosal Immunol. (2016) 9:1455–65. 10.1038/mi.2016.18 [] [[PubMed]
  • 15. Keller MD, Jyonouchi S. Chipping away at a mountain: genomic studies in common variable immunodeficiency. Autoimmun Rev. (2013) 12:687–9. 10.1016/j.autrev.2012.10.017 ] [
  • 16. Yong PF, Thaventhiran JE, Grimbacher B. “A rose is a rose is a rose,” but CVID is Not CVID common variable immune deficiency (CVID), what do we know in 2011?Adv Immunol. (2011) 111:47–107. 10.1016/B978-0-12-385991-4.00002-7 [] [[PubMed]
  • 17. Orange JS, Glessner JT, Resnick E, Sullivan KE, Lucas M, Ferry B, et al. . Genome-wide association identifies diverse causes of common variable immunodeficiency. J Allergy Clin Immunol. (2011) 127:1360–7.e1366. 10.1016/j.jaci.2011.02.039 ] [
  • 18. Rickert RC, Rajewsky K, Roes J. Impairment of T-cell-dependent B-cell responses and B-1 cell development in CD19-deficient mice. Nature. (1995) 376:352–5. 10.1038/376352a0 [] [[PubMed]
  • 19. Morsy DE, Sanyal R, Zaiss AK, Deo R, Muruve DA, Deans JP. Reduced T-dependent humoral immunity in CD20-deficient mice. J Immunol. (2013) 191:3112–8. 10.4049/jimmunol.1202098 [] [[PubMed]
  • 20. Kubinak JL, Petersen C, Stephens WZ, Soto R, Bake E, O'Connell RM, et al. . MyD88 signaling in T cells directs IgA-mediated control of the microbiota to promote health. Cell Host Microbe. (2015) 17:153–63. 10.1016/j.chom.2014.12.009 ] [
  • 21. Zhao G, Nyman M, Jönsson JA. Rapid determination of short-chain fatty acids in colonic contents and faeces of humans and rats by acidified water-extraction and direct-injection gas chromatography. Biomed Chromatogr. (2006) 20:674–82. 10.1002/bmc.580 [] [[PubMed]
  • 22. Barman M, Unold D, Shifley K, Amir E, Hung K, Bos N, et al. . Enteric salmonellosis disrupts the microbial ecology of the murine gastrointestinal tract. Infect Immun. (2008) 76:907–15. 10.1128/IAI.01432-07 ] [
  • 23. Basso M, Berlin J, Xia L, Sleiman SF, Ko B, Haskew-Layton R, et al. . Transglutaminase inhibition protects against oxidative stress-induced neuronal death downstream of pathological ERK activation. J Neurosci. (2012) 32:6561–9. 10.1523/JNEUROSCI.3353-11.2012 ] [
  • 24. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al. . QIIME allows analysis of high-throughput community sequencing data. Nat Methods. (2010) 7:335–6. 10.1038/nmeth.f.303 ] [
  • 25. Erben U, Loddenkemper C, Doerfel K, Spieckermann S, Haller D, Heimesaat MM, et al. . A guide to histomorphological evaluation of intestinal inflammation in mouse models. Int J Clin Exp Pathol. (2014) 7:4557–76.
  • 26. Wirtz S, Neufert C, Weigmann B, Neurath MF. Chemically induced mouse models of intestinal inflammation. Nat Protoc. (2007) 2:541–6. 10.1038/nprot.2007.41 [] [[PubMed]
  • 27. Fagarasan S, Kawamoto S, Kanagawa O, Suzuki K. Adaptive immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis. Annu Rev Immunol. (2010) 28:243–73. 10.1146/annurev-immunol-030409-101314 [] [[PubMed]
  • 28. Ahmad R, Sorrell MF, Batra SK, Dhawan P, Singh AB. Gut permeability and mucosal inflammation: bad, good or context dependent. Mucosal Immunol. (2017) 10:307–17. 10.1038/mi.2016.128 ] [
  • 29. Föger N, Jenckel A, Orinska Z, Lee KH, Chan AC, Bulfone-Paus S. Differential regulation of mast cell degranulation versus cytokine secretion by the actin regulatory proteins Coronin1a and Coronin1b. J Exp Med. (2011) 208:1777–87. 10.1084/jem.20101757 ] [
  • 30. Hasegawa M, Fujimoto M, Poe JC, Steeber DA, Tedder TF. CD19 can regulate B lymphocyte signal transduction independent of complement activation. J Immunol. (2001) 167:3190–200. 10.4049/jimmunol.167.6.3190 [] [[PubMed]
  • 31. Del Nagro CJ, Kolla RV, Rickert RC. A critical role for complement C3d and the B cell coreceptor (CD19/CD21) complex in the initiation of inflammatory arthritis. J Immunol. (2005) 175:5379–89. 10.4049/jimmunol.175.8.5379 [] [[PubMed]
  • 32. Cherukuri A, Cheng PC, Pierce SK. The role of the CD19/CD21 complex in B cell processing and presentation of complement-tagged antigens. J Immunol. (2001) 167:163–72. 10.4049/jimmunol.167.1.163 [] [[PubMed]
  • 33. Sato S, Ono N, Steeber DA, Pisetsky DS, Tedder TF. CD19 regulates B lymphocyte signaling thresholds critical for the development of B-1 lineage cells and autoimmunity. J Immunol. (1996) 157:4371–8. [[PubMed]
  • 34. Fujimoto M, Poe JC, Hasegawa M, Tedder TF. CD19 regulates intrinsic B lymphocyte signal transduction and activation through a novel mechanism of processive amplification. Immunol Res. (2000) 22:281–98. 10.1385/IR:22:2-3:281 [] [[PubMed]
  • 35. Haas KM, Poe JC, Steeber DA, Tedder TF. B-1a and B-1b cells exhibit distinct developmental requirements and have unique functional roles in innate and adaptive immunity to S. pneumonia. Immunity. (2005) 23:7–18. 10.1016/j.immuni.2005.04.011 [] [[PubMed]
  • 36. Matsushita T, Fujimoto M, Hasegawa M, Komura K, Takehara K, Tedder TF, et al. . Inhibitory role of CD19 in the progression of experimental autoimmune encephalomyelitis by regulating cytokine response. Am J Pathol. (2006) 168:812–21. 10.2353/ajpath.2006.050923 ] [
  • 37. Hasegawa M, Fujimoto M, Poe JC, Steeber DA, Lowell CA, Tedder TF. A CD19-dependent signaling pathway regulates autoimmunity in Lyn-deficient mice. J Immunol. (2001) 167:2469–78. 10.4049/jimmunol.167.5.2469 [] [[PubMed]
  • 38. Yanaba K, Yoshizaki A, Asano Y, Kadono T, Tedder TF, Sato S. IL-10-producing regulatory B10 cells inhibit intestinal injury in a mouse model. Am J Pathol. (2011) 178:735–43. 10.1016/j.ajpath.2010.10.022 ] [
  • 39. Yoshizaki A, Iwata Y, Komura K, Ogawa F, Hara T, Muroi E, et al. . CD19 regulates skin and lung fibrosis via Toll-like receptor signaling in a model of bleomycin-induced scleroderma. Am J Pathol. (2008) 172:1650–63. 10.2353/ajpath.2008.071049 ] [
  • 40. Wentink MW, Lambeck AJ, van Zelm MC, Simons E, van Dongen JJ, IJspeert H, et al. . CD21 and CD19 deficiency: Two defects in the same complex leading to different disease modalities. Clin Immunol. (2015) 161:120–7. 10.1016/j.clim.2015.08.010 [] [[PubMed]
  • 41. van Zelm MC, Reisli I, van der Burg M, Castaño D, van Noesel CJ, van Tol MJ, et al. . An antibody-deficiency syndrome due to mutations in the CD19 gene. N Engl J Med. (2006) 354:1901–12. 10.1056/NEJMoa051568 [] [[PubMed]
  • 42. Ahn S, Cunningham-Rundles C. Role of B cells in common variable immune deficiency. Expert Rev Clin Immunol. (2009) 5:557–64. 10.1586/eci.09.43 ] [
  • 43. Maseda D, Candando KM, Smith SH, Kalampokis I, Weaver CT, Plevy SE, et al. . Peritoneal cavity regulatory B cells (B10 cells) modulate IFN-gamma+CD4+ T cell numbers during colitis development in mice. J Immunol. (2013) 191:2780–95. 10.4049/jimmunol.1300649 ] [
  • 44. Schmidt EG, Larsen HL, Kristensen NN, Poulsen SS, Claesson MH, Pedersen AE. B cells exposed to enterobacterial components suppress development of experimental colitis. Inflamm Bowel Dis. (2012) 18:284–93. 10.1002/ibd.21769 [] [[PubMed]
  • 45. Fagarasan S, Muramatsu M, Suzuki K, Nagaoka H, Hiai H, Honjo T. Critical roles of activation-induced cytidine deaminase in the homeostasis of gut flora. Science. (2002) 298:1424–7. 10.1126/science.1077336 [] [[PubMed]
  • 46. Muramatsu M, Kinoshita K, Fagarasan S, Yamada S, Shinkai Y, Honjo T. Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell. (2000) 102:553–63. 10.1016/S0092-8674(00)00078-7 [] [[PubMed]
  • 47. Cazac BB, Roes J. TGF-beta receptor controls B cell responsiveness and induction of IgA in vivo. Immunity. (2000) 13:443–51. 10.1016/S1074-7613(00)00044-3 [] [[PubMed]
  • 48. Borsutzky S, Cazac BB, Roes J, Guzmán CA. TGF-beta receptor signaling is critical for mucosal IgA responses. J Immunol. (2004) 173:3305–9. 10.4049/jimmunol.173.5.3305 [] [[PubMed]
  • 49. Harriman GR, Bogue M, Rogers P, Finegold M, Pacheco S, Bradley A, et al. . Targeted deletion of the IgA constant region in mice leads to IgA deficiency with alterations in expression of other Ig isotypes. J Immunol. (1999) 162:2521–9. [[PubMed]
  • 50. Arnaboldi PM, Behr MJ, Metzger DW. Mucosal B cell deficiency in IgA-/- mice abrogates the development of allergic lung inflammation. J Immunol. (2005) 175:1276–85. 10.4049/jimmunol.175.2.1276 [] [[PubMed]
  • 51. Davies EG, Thrasher AJ. Update on the hyper immunoglobulin M syndromes. Br J Haematol. (2010) 149:167–80. 10.1111/j.1365-2141.2010.08077.x ] [
  • 52. Shulzhenko N, Morgun A, Hsiao W, Battle M, Yao M, Gavrilova O, et al. . Crosstalk between B lymphocytes, microbiota and the intestinal epithelium governs immunity versus metabolism in the gut. Nat Med. (2011) 17:1585–93. 10.1038/nm.2505 ] [
  • 53. Shulzhenko N, Dong X, Vyshenska D, Greer RL, Gurung M, Vasquez-Perez S, et al. . CVID enteropathy is characterized by exceeding low mucosal IgA levels and interferon-driven inflammation possibly related to the presence of a pathobiont. Clin Immunol. (2018) 197:139–53. 10.1016/j.clim.2018.09.008 ] [
  • 54. Resnick ES, Moshier EL, Godbold JH, Cunningham-Rundles C. Morbidity and mortality in common variable immune deficiency over 4 decades. Blood. (2012) 119:1650–7. 10.1182/blood-2011-09-377945 ] [
  • 55. Rogier EW, Frantz AL, Bruno ME, Kaetzel CS. Secretory IgA is concentrated in the outer layer of colonic mucus along with gut bacteria. Pathogens. (2014) 3:390–403. 10.3390/pathogens3020390 ] [
  • 56. Perreau M, Vigano S, Bellanger F, Pellaton C, Buss G, Comte D, et al. . Exhaustion of bacteria-specific CD4 T cells and microbial translocation in common variable immunodeficiency disorders. J Exp Med. (2014) 211:2033–45. 10.1084/jem.20140039 ] [
  • 57. Romberg N, Le Coz C, Glauzy S, Schickel JN, Trofa M, Nolan BE, et al. . Patients with common variable immunodeficiency with autoimmune cytopenias exhibit hyperplastic yet inefficient germinal center responses. J Allergy Clin Immunol. (2019) 143:258–65. 10.1016/j.jaci.2018.06.012 ] [
  • 58. Sachdev AH, Pimentel M. Gastrointestinal bacterial overgrowth: pathogenesis and clinical significance. Ther Adv Chronic Dis. (2013) 4:223–31. 10.1177/2040622313496126 ] [
  • 59. Dukowicz AC, Lacy BE, Levine GM. Small intestinal bacterial overgrowth: a comprehensive review. Gastroenterol Hepatol. (2007) 3:112–22.
  • 60. Haboubi NY, Lee GS, Montgomery RD. Duodenal mucosal morphometry of elderly patients with small intestinal bacterial overgrowth: response to antibiotic treatment. Age Ageing. (1991) 20:29–32. 10.1093/ageing/20.1.29 [] [[PubMed]
  • 61. Di Stefano M, Malservisi S, Veneto G, Ferrieri A, Corazza GR. Rifaximin versus chlortetracycline in the short-term treatment of small intestinal bacterial overgrowth. Aliment Pharmacol Ther. (2000) 14:551–6. 10.1046/j.1365-2036.2000.00751.x [] [[PubMed]
  • 62. Di Stefano M, Miceli E, Missanelli A, Mazzocchi S, Corazza GR. Absorbable vs. non-absorbable antibiotics in the treatment of small intestine bacterial overgrowth in patients with blind-loop syndrome. Aliment Pharmacol Ther. (2005) 21:985–92. 10.1111/j.1365-2036.2005.02397.x [] [[PubMed]
  • 63. Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS. Bile acids and the gut microbiome. Curr Opin Gastroenterol. (2014) 30:332–8. 10.1097/MOG.0000000000000057 ] [
  • 64. Verdu EF, Armstrong D, Murray JA. Between celiac disease and irritable bowel syndrome: the “No Man's Land” of Gluten Sensitivity”. Am J Gastroenterol. (2009) 104:1587–94. 10.1038/ajg.2009.188 ] [
  • 65. Fasano A, Sapone A, Zevallos V, Schuppan D. Nonceliac gluten and wheat sensitivity. Gastroenterology. (2015) 148:1195–204. 10.1053/j.gastro.2014.12.049 [] [[PubMed]
  • 66. Dickey W, Hughes DF. Histology of the terminal ileum in coeliac disease. Scand J Gastroenterol. (2004) 39:665–7. 10.1080/00365520410004901 [] [[PubMed]
  • 67. Barton SH, Kelly DG, Murray JA. Nutritional deficiencies in celiac disease. Gastroenterol Clin North Am. (2007) 36:93–108. 10.1016/j.gtc.2007.01.006 [] [[PubMed]
  • 68. Hopper AD, Hurlstone DP, Leeds JS, McAlindon ME, Dube AK, Stephenson TJ, et al. . The occurrence of terminal ileal histological abnormalities in patients with coeliac disease. Dig Liver Dis. (2006) 38:815–9. 10.1016/j.dld.2006.04.003 [] [[PubMed]
  • 69. Fasano A. Clinical presentation of celiac disease in the pediatric population. Gastroenterology. (2005) 128:S68–73. 10.1053/j.gastro.2005.02.015 [] [[PubMed]
  • 70. Rubio-Tapia A, Murray JA. Classification and management of refractory coeliac disease. Gut. (2010) 59:547–57. 10.1136/gut.2009.195131 ] [
  • 71. Taylor AK, Lebwohl B, Snyder CL, Green PHR. Celiac disease. In: Adam MP, Ardinger HH, Pagon RA, et al., editors. editors. GeneReviews® [Internet]. Seattle, WA: University of Washington; (2008). [PubMed]
  • 72. Marasco G, Di Biase AR, Schiumerini R, Eusebi LH, Iughetti L, Ravaioli F, et al. . Gut microbiota and celiac disease. Digest Dis Sci. (2016) 61:1461–72. 10.1007/s10620-015-4020-2 [] [[PubMed]
  • 73. Kumar V, Jarzabek-Chorzelska M, Sulej J, Karnewska K, Farrell T, Jablonska S. Celiac disease and immunoglobulin a deficiency: how effective are the serological methods of diagnosis?Clin Diagn Lab Immunol. (2002) 9:1295–300. 10.1128/CDLI.9.6.1295-1300.2002 ] [
  • 74. Chow MA, Lebwohl B, Reilly NR, Green PH. Immunoglobulin A deficiency in celiac disease. J Clin Gastroenterol. (2012) 46:850–4. 10.1097/MCG.0b013e31824b2277 [] [[PubMed]
  • 75. Licinio R, Principi M, Amoruso A, Piscitelli D, Ierardi E, Di Leo A. Celiac disease and common variable immunodeficiency: a familial inheritance?J Gastrointestin Liver Dis. (2013) 22:473. [[PubMed]
  • 76. Malamut G, Verkarre V, Suarez F, Viallard JF, Lascaux AS, Cosnes J, et al. . The enteropathy associated with common variable immunodeficiency: the delineated frontiers with celiac disease. Am J Gastroenterol. (2010) 105:2262–75. 10.1038/ajg.2010.214 [] [[PubMed]
  • 77. Giorgio F, Principi M, Losurdo G, Piscitelli D, Iannone A, Barone M, et al. . Seronegative celiac disease and immunoglobulin deficiency: where to look in the submerged iceberg?Nutrients. (2015) 7:7486–504. 10.3390/nu7095350 ] [
  • 78. Torsten M, Aaron L. Microbial transglutaminase is immunogenic and potentially pathogenic in pediatric celiac disease. Front Pediatric. (2018) 6:389. 10.3389/fped.2018.00389 ] [
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.