Linking bacterial enterotoxins and alpha defensin 5 expansion in the Crohn's colitis: A new insight into the etiopathogenetic and differentiation triggers driving colonic inflammatory bowel disease

Tanu Rana, Olga Y Korolkova, Girish Rachakonda, Amanda D Williams, Alexander T Hawkins, Samuel D James, Amos M Sakwe, Nian Hui, Li Wang, Chang Yu, Jeffrey S Goodwin, Michael G Izban, Regina S Offodile, Mary K Washington, Billy R Ballard, Duane T Smoot, Xuan-Zheng Shi, Digna S Forbes, Anil Shanker, Amosy E M'Koma, Tanu Rana, Olga Y Korolkova, Girish Rachakonda, Amanda D Williams, Alexander T Hawkins, Samuel D James, Amos M Sakwe, Nian Hui, Li Wang, Chang Yu, Jeffrey S Goodwin, Michael G Izban, Regina S Offodile, Mary K Washington, Billy R Ballard, Duane T Smoot, Xuan-Zheng Shi, Digna S Forbes, Anil Shanker, Amosy E M'Koma

Abstract

Evidence link bacterial enterotoxins to apparent crypt-cell like cells (CCLCs), and Alpha Defensin 5 (DEFA5) expansion in the colonic mucosa of Crohn's colitis disease (CC) patients. These areas of ectopic ileal metaplasia, positive for Paneth cell (PC) markers are consistent with diagnosis of CC. Retrospectively, we: 1. Identified 21 patients with indeterminate colitis (IC) between 2000-2007 and were reevaluation their final clinical diagnosis in 2014 after a followed-up for mean 8.7±3.7 (range, 4-14) years. Their initial biopsies were analyzed by DEFA5 bioassay. 2. Differentiated ulcer-associated cell lineage (UACL) analysis by immunohistochemistry (IHC) of the CC patients, stained for Mucin 6 (MUC6) and DEFA5. 3. Treated human immortalized colonic epithelial cells (NCM460) and colonoids with pure DEFA5 on the secretion of signatures after 24hr. The control colonoids were not treated. 4. Treated colonoids with/without enterotoxins for 14 days and the spent medium were collected and determined by quantitative expression of DEFA5, CCLCs and other biologic signatures. The experiments were repeated twice. Three statistical methods were used: (i) Univariate analysis; (ii) LASSO; and (iii) Elastic net. DEFA5 bioassay discriminated CC and ulcerative colitis (UC) in a cohort of IC patients with accuracy. A fit logistic model with group CC and UC as the outcome and the DEFA5 as independent variable differentiator with a positive predictive value of 96 percent. IHC staining of CC for MUC6 and DEFA5 stained in different locations indicating that DEFA5 is not co-expressed in UACL and is therefore NOT the genesis of CC, rather a secretagogue for specific signature(s) that underlie the distinct crypt pathobiology of CC. Notably, we observed expansion of signatures after DEFA5 treatment on NCM460 and colonoids cells expressed at different times, intervals, and intensity. These factors are key stem cell niche regulators leading to DEFA5 secreting CCLCs differentiation 'the colonic ectopy ileal metaplasia formation' conspicuously of pathogenic importance in CC.

Conflict of interest statement

The authors declare conflicts of interests to disclose, A.E. M’Koma has received Honoraria fees for Educational Presentation from Lipscomb University Health Sciences. Further, he is an inventor of two Patents: (i) Assay methods for diagnosing and treating inflammatory bowel disease with human alpha-defensin 5 (US16/571,034, 2020) and (ii) A.E. M’Koma and A.M. Sakwe - Targeted DEFA5 antibody and assay methods for diagnosing and treating inflammatory bowel disease (US62/522,652, 2020). This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Representative H & E staining…
Fig 1. Representative H & E staining of the colon.
A, Normalnormal crypt; B, UC–sporadic to no CCLCs even in severe disease activity; and C, CC–abundant CCLCs in the crypt base (arrow) seen even in mild disease activity.
Fig 2. Immunohistochemical detection of CCLCs marker…
Fig 2. Immunohistochemical detection of CCLCs marker (positive for Paneth cell) DEFA5 and UACL marker MUC6 in the intestinal mucosa of colonic CC.
Seral adjacent tissue sections were stained with MUC6 (left column) and DEFA5 (right column) on diseased colon from Crohn’s patients. Full tissue section (row A) and enlarged regions associated with MUC6 (box B) or DEFA5 (box C) demonstrate that DEFA5 expression within the apparent CCLCs is not localized within the UACL region of the diseased colon.
Fig 3. Multivariable logistic regression model for…
Fig 3. Multivariable logistic regression model for probability of being CC.
A, twenty-one (#21) IC patients were followed for 8.7±3.7 (range, 4–14) years. Although most IC resolved to UC or CC, but the mean diagnostic delay was 7 (range, 4–14) years. 28.5% of the patients could still not be diagnosed into authentic UC or CC. The mean area fraction of DEFA5 count (%) by NEARAS IHC staining agrees with final diagnostic outcome of every IC patient samples tested. B, Clinical data of these 21 IC patients who correlate with 3A data could have been diagnosed using DEFA5 immunoreactivity during the first endoscopy biopsy to avoid diagnosis delay. C, Bioinformatic data depicting a fit logistic model with group CC vs. UC (in 3A) as the outcome and DEFA5 as independent variable discriminator. The R2 of the model fit is 1 and area under the ROC curve is 1. The area under the ROC curve was not plotted since it will look similar as the orange curve in ROC plot. Three statistical methods were used: (1) Univariate analysis: Mann Whitney U test between two groups for each peak, adjusting p-values by FDR; (2) LASSO; and (3) Elastic net. As the scale of all measures were small, we multiplied all values by 10,000.
Fig 4. Treatment of colonoids with bacterial…
Fig 4. Treatment of colonoids with bacterial enterotoxins.
A–B, Equal numbers of colonoids were treated with/without enterotoxins (LPS, LTA, SAE) following a time course of up to 14 days. At each time point, we collected both the colonoids and the spent medium and determined the expression and/or secretion of DEFA5 by ELISA and IHC staining for lysozymes. We also seeded the colonoids at low density in triplicate, allowed to attach overnight, and then treated one set with enterotoxins and the other set without. We allowed the colonoids to form colonies for 14 days. Colonies were stained with anti-DEFA5 to determine whether the treatment led to differentiation into apparent CCLCs. The expression and/or secretion of DEFA5 and proinflammatory cytokines, etc. was determined by ELISA. C, Lysozyme assay: The assay was performed using lysozyme ELISA kit firm abcam (catalog no. ab108880 as per manufacturer’s instructions.
Fig 5. Treatment of immortalized colonic epithelial…
Fig 5. Treatment of immortalized colonic epithelial cells and colonoids with DEFA5.
We graphically present the data on the signatures for immortalized colonic epithelial cells and colonoids separately. We did not perform statistical tests on these data. Immortalized colonic epithelial cells (NCM 460) were received by a Material Transfer Agreement from INCELL Corporation (San Antonio, Texas, USA) [8]. NCM 460 cells (1x106 cells) were plated in a 100 mm culture dish prior to starting the experiment. At the end of the treatment period, the cell culture supernatant was collected and frozen at –80°C until supernatants from all the treatment time-points were collected. Abcam Cytokine Antibody Array (catalog no. ab133998) was used for the simultaneous detection of multiple cytokines following the manufacturer’s protocol. Colonoids were purchased from Cellesce Limited (Medicentre, Cardiff, UK). Organoid culture was performed as per instructions provided by the manufacturer (Cellesce). Two thousand colonoids per well were plated in a 12-well plate. After 2 days of growth in complete medium, the colonoids were treated with purified Human DEFA5 (5 μg/ml) for 6hr. and 24hr. The control colonoids were not treated. At the end of the treatment period, the colonoids culture supernatant was collected and frozen at –80°C until supernatants from all the treatment time-points were collected. The experiments were repeated twice. At each time point, we collected both the colonoids and the spent medium and determined the expression and/or secretion of DEFA5 by ELISA and IHC staining for lysozymes. We also seeded the colonoids at low density in triplicate, allowed to attach overnight, and then treated one set with enterotoxins and the other set without and allowed the colonoids to form colonies for up to 14 days. Cytokines, chemokines, and growth factor measurements (signatures) are depicted: A, 24hr measurement; B, (24hr measurement) minus (6hr measurement) and C, (24hr measurement) minus (Control 24hr measurement) respectively.

References

    1. M’Koma AE. The Multifactorial Etiopathogeneses Interplay of Inflammatory Bowel Disease: An Overview. Gastrointest Disord 2018;1:75–105.
    1. Liu JZ, van Sommeren S, Huang H, et al.. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet 2015;47:979–86. 10.1038/ng.3359
    1. Satsangi J, Silverberg MS, Vermeire S, Colombel JF. The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut 2006;55:749–53. 10.1136/gut.2005.082909
    1. Spekhorst LM, Visschedijk MC, Alberts R, et al.. Performance of the Montreal classification for inflammatory bowel diseases. World J Gastroenterol 2014;20:15374–81. 10.3748/wjg.v20.i41.15374
    1. Williams AD, Korolkova OY, Sakwe AM, et al.. Human alpha defensin 5 is a candidate biomarker to delineate inflammatory bowel disease. PLoS One 2017;12:e0179710. 10.1371/journal.pone.0179710
    1. Korolkova OY, Myers JN, Pellom S. T., Wang L., M’Koma A. E. Characterization of Serum Cytokine Profile in Predominantly Colonic Inflammatory Bowel Disease to Delineate Ulcerative and Crohn’s Colitides. Clin Med Insingts Gastroenterol 2015:29–44. 10.4137/CGast.S20612
    1. Jarde T, Lloyd-Lewis B, Thomas M, et al.. Wnt and Neuregulin1/ErbB signalling extends 3D culture of hormone responsive mammary organoids. Nat Commun 2016;7:13207. 10.1038/ncomms13207
    1. Moyer MP, Manzano LA, Merriman RL, Stauffer JS, Tanzer LR. NCM460, a normal human colon mucosal epithelial cell line. In Vitro Cell Dev Biol Anim 1996;32:315–7. 10.1007/BF02722955
    1. Myers JN, Schaffer MW, Korolkova OY, Williams AD, Gangula PR, M’Koma AE. Implications of the Colonic Deposition of Free Hemoglobin-alpha Chain: A Previously Unknown Tissue By-product in Inflammatory Bowel Disease. Inflamm Bowel Dis 2014;20:1530–47. 10.1097/MIB.0000000000000144
    1. Gmyr V, Bonner C, Lukowiak B, et al.. Automated digital image analysis of islet cell mass using Nikon’s inverted eclipse Ti microscope and software to improve engraftment may help to advance the therapeutic efficacy and accessibility of islet transplantation across centers. Cell Transplant 2015;24:1–9. 10.3727/096368913X667493
    1. M’Koma AESE, Washington MK, Schwartz DA, Muldoon RL, Herline AJ, Wise PE, et al.. Proteomic profiling of mucosal and submucosal colonic tissues yields protein signatures that differentiate the inflammatory colitides. Inflamm Bowel Dis 2011;17:875–83. 10.1002/ibd.21442
    1. Seeley EH, Washington MK, Caprioli RM, M’Koma AE. Proteomic patterns of colonic mucosal tissues delineate Crohn’s colitis and ulcerative colitis. Proteomics Clin Appl 2013;7:541–9. 10.1002/prca.201200107
    1. Targan SR, Karp LC. Inflammatory bowel disease diagnosis, evaluation and classification: state-of-the art approach. Curr Opin Gastroenterol 2007;23:390–4. 10.1097/MOG.0b013e3281722271
    1. James SD, Wise PE, Zuluaga-Toro T, Schwartz DA, Washington MK, Shi C. Identification of pathologic features associated with "ulcerative colitis-like" Crohn’s disease. World J Gastroenterol 2014;20:13139–45. 10.3748/wjg.v20.i36.13139
    1. M’Koma AE. Inflammatory Bowel Disease: An Expanding Global Health Problem. Clinical Medicine Insights Gastroenterology 2013:33–47. 10.4137/CGast.S12731
    1. Dahlhamer JM, Zammitti EP, Ward BW, Wheaton AG, Croft JB. Prevalence of Inflammatory Bowel Disease Among Adults Aged >/ = 18 Years—United States, 2015. MMWR Morb Mortal Wkly Rep 2016;65:1166–9. 10.15585/mmwr.mm6542a3
    1. Kappelman MD, Moore KR, Allen JK, Cook SF. Recent trends in the prevalence of Crohn’s disease and ulcerative colitis in a commercially insured US population. Dig Dis Sci 2013;58:519–25. 10.1007/s10620-012-2371-5
    1. Abramson O, Durant M, Mow W, et al.. Incidence, prevalence, and time trends of pediatric inflammatory bowel disease in Northern California, 1996 to 2006. J Pediatr 2010;157:233–9 e1. 10.1016/j.jpeds.2010.02.024
    1. Adamiak T, Walkiewicz-Jedrzejczak D, Fish D, et al.. Incidence, clinical characteristics, and natural history of pediatric IBD in Wisconsin: a population-based epidemiological study. Inflamm Bowel Dis 2013;19:1218–23. 10.1097/MIB.0b013e318280b13e
    1. Bossuyt PM, Reitsma JB, Bruns DE, et al.. STARD 2015: an updated list of essential items for reporting diagnostic accuracy studies. BMJ 2015;351:h5527. 10.1136/bmj.h5527
    1. Benchimol EI, Bernstein CN, Bitton A, et al.. Trends in Epidemiology of Pediatric Inflammatory Bowel Disease in Canada: Distributed Network Analysis of Multiple Population-Based Provincial Health Administrative Databases. Am J Gastroenterol 2017;112:1120–34. 10.1038/ajg.2017.97
    1. Ananthakrishnan AN, Kwon J, Raffals L, et al.. Variation in treatment of patients with inflammatory bowel diseases at major referral centers in the United States. Clin Gastroenterol Hepatol 2015;13:1197–200. 10.1016/j.cgh.2014.11.020
    1. Bagheri Lankarani KGS, Hadipour M, Pourhashemi M, Mahmoodi A, Zeraatpishe M, Babaei AH. Determinants of Hospital Costs of Inflammatory Bowel Disease. Govaresh 2020;24:230–7.
    1. Park KTEO Allen JI, Meadows P Szigethy EM, Henrichsen K Kim SC, et al.. The Cost of Inflammatory Bowel Disease: An Initiative From the Crohn’s & Colitis Foundation. Inflamm Bowel Dis 2020;1:1–10.
    1. Xu F, Liu Y, Wheaton AG, Rabarison KM, Croft JB. Trends and Factors Associated with Hospitalization Costs for Inflammatory Bowel Disease in the United States. Appl Health Econ Health Policy 2019;17:77–91. 10.1007/s40258-018-0432-4
    1. Petryszyn PW, Witczak I. Costs in inflammatory bowel diseases. Prz Gastroenterol 2016;11:6–13. 10.5114/pg.2016.57883
    1. Kappelman MD, Rifas-Shiman SL, Porter CQ, et al.. Direct health care costs of Crohn’s disease and ulcerative colitis in US children and adults. Gastroenterology 2008;135:1907–13. 10.1053/j.gastro.2008.09.012
    1. Ananthakrishnan AN, Cagan A, Cai T, et al.. Comparative Effectiveness of Infliximab and Adalimumab in Crohn’s Disease and Ulcerative Colitis. Inflamm Bowel Dis 2016;22:880–5. 10.1097/MIB.0000000000000754
    1. Martini E, Krug SM, Siegmund B, Neurath MF, Becker C. Mend Your Fences: The Epithelial Barrier and its Relationship With Mucosal Immunity in Inflammatory Bowel Disease. Cell Mol Gastroenterol Hepatol 2017;4:33–46. 10.1016/j.jcmgh.2017.03.007
    1. Leung G, Muise AM. Monogenic Intestinal Epithelium Defects and the Development of Inflammatory Bowel Disease. Physiology (Bethesda) 2018;33:360–9. 10.1152/physiol.00020.2018
    1. Swidsinski A, Ladhoff A, Pernthaler A, et al.. Mucosal flora in inflammatory bowel disease. Gastroenterology 2002;122:44–54. 10.1053/gast.2002.30294
    1. Swidsinski A, Weber J, Loening-Baucke V, Hale LP, Lochs H. Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease. J Clin Microbiol 2005;43:3380–9. 10.1128/JCM.43.7.3380-3389.2005
    1. Baumgart M, Dogan B, Rishniw M, et al.. Culture independent analysis of ileal mucosa reveals a selective increase in invasive Escherichia coli of novel phylogeny relative to depletion of Clostridiales in Crohn’s disease involving the ileum. ISME J 2007;1:403–18. 10.1038/ismej.2007.52
    1. Parkes GC, Rayment NB, Hudspith BN, et al.. Distinct microbial populations exist in the mucosa-associated microbiota of sub-groups of irritable bowel syndrome. Neurogastroenterol Motil 2012;24:31–9. 10.1111/j.1365-2982.2011.01803.x
    1. Goldenring JR. Pyloric metaplasia, pseudopyloric metaplasia, ulcer-associated cell lineage and spasmolytic polypeptide-expressing metaplasia: reparative lineages in the gastrointestinal mucosa. J Pathol 2018;245:132–7. 10.1002/path.5066
    1. Martinez-Fierro ML, Garza-Veloz I, Rocha-Pizana MR, et al.. Serum cytokine, chemokine, and growth factor profiles and their modulation in inflammatory bowel disease. Medicine (Baltimore) 2019;98:e17208.
    1. Granlund A, Beisvag V, Torp SH, et al.. Activation of REG family proteins in colitis. Scand J Gastroenterol 2011;46:1316–23. 10.3109/00365521.2011.605463
    1. Thorsvik S, van Beelen Granlund A, Svendsen TD, et al.. Ulcer-associated cell lineage expresses genes involved in regeneration and is hallmarked by high neutrophil gelatinase-associated lipocalin (NGAL) levels. J Pathol 2019;248:316–25. 10.1002/path.5258
    1. Sankaran-Walters S, Hart R, Dills C. Guardians of the Gut: Enteric Defensins. Front Microbiol 2017;8:647. 10.3389/fmicb.2017.00647
    1. James SDHA, Um JW, Ballard BR, Smoot DT, M’Koma AE. The MYTHS of de novo Crohn’s Disease After Restorative Proctocolectomy with Ileal Pouch-anal Anastomosis for Ulcerative Colitis. Japenese Journal of Gastroenterology and Hepatology 2020;3:1–10.
    1. Fabregat A, Sidiropoulos K, Viteri G, et al.. Reactome pathway analysis: a high-performance in-memory approach. BMC Bioinformatics 2017;18:142. 10.1186/s12859-017-1559-2
    1. Nielsen OH, Coskun M, Steenholdt C, Rogler G. The role and advances of immunomodulator therapy for inflammatory bowel disease. Expert Rev Gastroenterol Hepatol 2015;9:177–89. 10.1586/17474124.2014.945914
    1. Nielsen OH, Ainsworth MA. Tumor necrosis factor inhibitors for inflammatory bowel disease. N Engl J Med 2013;369:754–62. 10.1056/NEJMct1209614
    1. Nielsen OH, Ainsworth MA. Which biological agents are most appropriate for ulcerative colitis? Ann Intern Med 2014;160:733–4. 10.7326/M14-0605
    1. Pedersen J, Coskun M, Soendergaard C, Salem M, Nielsen OH. Inflammatory pathways of importance for management of inflammatory bowel disease. World J Gastroenterol 2014;20:64–77. 10.3748/wjg.v20.i1.64
    1. Andersson E, Bergemalm D, Kruse R, et al.. Subphenotypes of inflammatory bowel disease are characterized by specific serum protein profiles. PLoS One 2017;12:e0186142. 10.1371/journal.pone.0186142
    1. Beaurepaire C, Smyth D, McKay DM. Interferon-gamma regulation of intestinal epithelial permeability. J Interferon Cytokine Res 2009;29:133–44. 10.1089/jir.2008.0057
    1. Scharl M, Paul G, Barrett KE, McCole DF. AMP-activated protein kinase mediates the interferon-gamma-induced decrease in intestinal epithelial barrier function. J Biol Chem 2009;284:27952–63. 10.1074/jbc.M109.046292
    1. Coburn LA, Horst SN, Chaturvedi R, et al.. High-throughput multi-analyte Luminex profiling implicates eotaxin-1 in ulcerative colitis. PLoS One 2013;8:e82300. 10.1371/journal.pone.0082300
    1. Rodriguez-Peralvarez ML, Garcia-Sanchez V, Villar-Pastor CM, et al.. Role of serum cytokine profile in ulcerative colitis assessment. Inflamm Bowel Dis 2012;18:1864–71. 10.1002/ibd.22865
    1. Townsend P, Zhang Q, Shapiro J, et al.. Serum Proteome Profiles in Stricturing Crohn’s Disease: A Pilot Study. Inflamm Bowel Dis 2015;21:1935–41. 10.1097/MIB.0000000000000445
    1. Marini M, Bamias G, Rivera-Nieves J, et al.. TNF-alpha neutralization ameliorates the severity of murine Crohn’s-like ileitis by abrogation of intestinal epithelial cell apoptosis. Proc Natl Acad Sci U S A 2003;100:8366–71. 10.1073/pnas.1432897100
    1. Su L, Nalle SC, Shen L, et al.. TNFR2 activates MLCK-dependent tight junction dysregulation to cause apoptosis-mediated barrier loss and experimental colitis. Gastroenterology 2013;145:407–15. 10.1053/j.gastro.2013.04.011
    1. Nava P, Koch S, Laukoetter MG, et al.. Interferon-gamma regulates intestinal epithelial homeostasis through converging beta-catenin signaling pathways. Immunity 2010;32:392–402. 10.1016/j.immuni.2010.03.001
    1. Mankertz J, Schulzke JD. Altered permeability in inflammatory bowel disease: pathophysiology and clinical implications. Curr Opin Gastroenterol 2007;23:379–83. 10.1097/MOG.0b013e32816aa392
    1. Suenaert P, Bulteel V, Lemmens L, et al.. Anti-tumor necrosis factor treatment restores the gut barrier in Crohn’s disease. Am J Gastroenterol 2002;97:2000–4. 10.1111/j.1572-0241.2002.05914.x
    1. Toedter G, Li K, Sague S, et al.. Genes associated with intestinal permeability in ulcerative colitis: changes in expression following infliximab therapy. Inflamm Bowel Dis 2012;18:1399–410. 10.1002/ibd.22853
    1. Strober W, Fuss IJ. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology 2011;140:1756–67. 10.1053/j.gastro.2011.02.016
    1. Funderburg NT, Stubblefield Park SR, Sung HC, et al.. Circulating CD4(+) and CD8(+) T cells are activated in inflammatory bowel disease and are associated with plasma markers of inflammation. Immunology 2013;140:87–97. 10.1111/imm.12114
    1. Antoni L, Nuding S, Wehkamp J, Stange EF. Intestinal barrier in inflammatory bowel disease. World J Gastroenterol 2014;20:1165–79. 10.3748/wjg.v20.i5.1165
    1. Holly MK, Smith JG. Paneth Cells during Viral Infection and Pathogenesis. Viruses 2018;10. 10.3390/v10050225
    1. Holly MK, Diaz K, Smith JG. Defensins in Viral Infection and Pathogenesis. Annu Rev Virol 2017;4:369–91. 10.1146/annurev-virology-101416-041734

Source: PubMed

3
Abonnere