Autologous organoid co-culture model reveals T cell-driven epithelial cell death in Crohn's Disease

Nassim Hammoudi, Sarah Hamoudi, Julie Bonnereau, Hugo Bottois, Kevin Pérez, Madeleine Bezault, Déborah Hassid, Victor Chardiny, Céline Grand, Brice Gergaud, Joëlle Bonnet, Leila Chedouba, My-Linh Tran Minh, Jean-Marc Gornet, Clotilde Baudry, Hélène Corte, Léon Maggiori, Antoine Toubert, Jacqueline McBride, Camille Brochier, Margaret Neighbors, Lionel Le Bourhis, Matthieu Allez, Nassim Hammoudi, Sarah Hamoudi, Julie Bonnereau, Hugo Bottois, Kevin Pérez, Madeleine Bezault, Déborah Hassid, Victor Chardiny, Céline Grand, Brice Gergaud, Joëlle Bonnet, Leila Chedouba, My-Linh Tran Minh, Jean-Marc Gornet, Clotilde Baudry, Hélène Corte, Léon Maggiori, Antoine Toubert, Jacqueline McBride, Camille Brochier, Margaret Neighbors, Lionel Le Bourhis, Matthieu Allez

Abstract

Lympho-epithelial interactions between intestinal T resident memory cells (Trm) and the epithelium have been associated with inflammatory bowel disease (IBD) activity. We developed ex vivo autologous organoid-mucosal T cell cocultures to functionally assess lymphoepithelial interactions in Crohn's Disease (CD) patients compared to controls. We demonstrate the direct epithelial cell death induced by autologous mucosal T cells in CD patients but not in controls. These findings were positively correlated with T cell infiltration of the organoids. This potential was inhibited by limiting lympho-epithelial interactions through CD103 and NKG2D blocking antibodies. These data directly demonstrate for the first time the direct deleterious effect of mucosal T cells on the epithelium of CD patients. Such ex-vivo models are promising techniques to unravel the pathophysiology of these diseases and the potential mode of action of current and future therapies.

Keywords: CD103; NKG2D; crohn’s disease; inflammatory bowel diseases; lymphoepithelial interactions; organoids.

Conflict of interest statement

MA received grant supports from Innate Pharma, Janssen, Takeda, Genentech/Roche, and honoraria for teaching activities or consultancy from Abbvie, Amgen, Biogen, Boehringer-Ingelheim, Bristol Myers Squibb, Celgene, Celltrion, Ferring, Genentech, Gilead, IQVIA, Janssen, Novartis, Pfizer, Roche, Takeda, Tillots. NH received honorarium for consultancy from Janssen and Takeda. M-LT-M received honoraria from Abbvie and Janssen. J-MG has been a speaker and/advisory board member for Abbvie, Amgen, Celltrion, Takeda, Janssen and Sanofi Genzyme. SH, JB, HB, KP, LC, MB, VC, BG, CG, DH, JB, HC, LM, CB, AT and LLB have no conflict of interest to declare. MN, JM are employed by Genentech and CB by Institut Roche. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be constructed as a potential conflict of interest.

Copyright © 2022 Hammoudi, Hamoudi, Bonnereau, Bottois, Pérez, Bezault, Hassid, Chardiny, Grand, Gergaud, Bonnet, Chedouba, Tran Minh, Gornet, Baudry, Corte, Maggiori, Toubert, McBride, Brochier, Neighbors, Le Bourhis and Allez.

Figures

Figure 1
Figure 1
Ileal mucosal T cells from patients with active CD have an increased cytotoxic potential over Caco2 spheroids. (A) Caco2 spheroids alone or in coculture with mucosal T cells from CD patients in brightfield microscopy (10X) and confocal microscopy (200X: Blue: DAPI; Light Blue: E-Cadherin). Red arrows correspond to T cells infiltrating the spheroid (Red: CD3). (B) Gating strategy to assess Caco2 cell death with or without T cells in cocultures. Cell death was assessed through Annexin V and DAPI staining in E-Cadherin positive cells. (C) Comparison of Caco2 cells death rates between allogenic cocultures with mucosal T cells from controls and active CD patients. CD= Crohn’s Disease, CTR= Control. CTR: n=5; CD: n=8. *p < 0.05 Mann Whitney Test. +: p < 0.05 Wilcoxon paired Test.
Figure 2
Figure 2
Autologous co-cultures reveal direct interactions between mucosal lymphocytes and epithelial organoids especially in CD patients. (A) Experimental design of cocultures (B) Example of a two photon acquisition for a CD patient showing multiple interactions between T cells and an organoid. (C) Average speed, total displacement and arrest coefficient of lymphocytes in cocultures. Each dot corresponds to a lymphocyte. Cells from different individuals were combined in this figure. These figures represent 2 control patients and 3 IBD patients. CD= Crohn’s Disease, CTR= Control. *p < 0.05 Mann Whitney Test.
Figure 3
Figure 3
Autologous T cells from CD patients infiltrate organoids and induce epithelial cell death. (A) T cell infiltration in the organoids evaluated by the number of CD3 positive-cells in contact or infiltrating each organoid with or without LPS preincubation. Each dot corresponds to the median of the values obtained for organoids of a given patient. Each shape of point corresponds to a given patient. CTR: n=6, CD: n=7. (B) Organoid epithelial cell death evaluated by Caspase 3 fluorescence area over organoid volume in organoids alone or cocultures with or without LPS preincubation. CTR: n=6, CD: n=7 (C) Correlation between T cell infiltration and epithelial cell death in coculture with LPS preincubation. Each dot corresponds to the absolute value obtained for an organoid. (D) Cytokine detection matrix in the supernatants. Each square represents the median value of the results obtained for each well of a given condition in the distinct patients. The color of each square stands for the level expression. CTR: n=6 CD: n=6. (E) Absolute values of cytokine quantification of 6 analytes (IL-17a, IL-8, TNFalpha, IFNgamma, Perforin and IL-6) in the supernatants from CD patients. Each dot corresponds to the absolute value obtained for a given condition. Each shape of point corresponds to a given patient. CC= Coculture, CD= Crohn’s Disease, CTR= Control, LPS= Lipopolysaccharide ML= Mucosal Lymphocytes, Orga= Organoid. +: p < 0.05 Wilcoxon paired Test, *p < 0.05 Mann-Whitney Test.
Figure 4
Figure 4
Monoclonal antibodies targeting lympho-epithelial interactions reduce epithelial cell death in co-cultures. (A) Organoid in coculture with mucosal T cells from CD patients without (left) or with anti-beta7 (right) in confocal microscopy (200X: Blue: DAPI; Light Blue: E-Cadherin, Red CD3, Pink: Caspase 3). (B) T cell infiltration in the organoids evaluated by the number of CD3 positive-cells in contact or infiltrating each organoid with LPS incubation. Each dot corresponds to the median of the values obtained for organoids of a given patient. Each shape of point corresponds to a given patient. CTR: n=6, CD: n=7. (C) Organoid epithelial cell death evaluated by Caspase 3 fluorescence area over organoid volume in organoids alone or cocultures with LPS incubation. CTR: n=6, CD: n=7. (D) Cytokine detection matrix in the supernatant in LPS coculture conditions. Each square represents the median value of the results obtained for each well of a given condition in the distinct patients. The color of each square stands for the level expression (Form light blue: low expression to deep red: high expression). CTR: n=6, CD: n=6. (E) Absolute values of cytokine quantification of 6 analytes (IL-17a, IL-8, TNFalpha, IFNgamma, Perforin and IL-6) for CD patients. Each dot corresponds to the absolute value obtained for a given condition (with LPS). Each shape of point corresponds to a given patient. CC= Coculture, CD= Crohn’s Disease, CTR= Control, ML= Mucosal Lymphocytes, Orga= Organoid. +: p < 0.05 Wilcoxon paired test.
Figure 5
Figure 5
CD103 expression on mucosal T cells is associated with IBD disease activity. (A) Gating strategy for CD103 and CD45RO stainings in flow cytometry. (B) Comparison of mucosal rates of CD4 and CD8 T cells expressing CD103 between controls and patients from the ELYP cohort at week 0. (C) Evolution of mucosal rate of CD8 T cells expressing CD103 rates in IBD patients from the ELYP cohort without or with endoscopic response to biotherapy. (D) Evolution of mucosal rate of CD4 T cells expressing CD103 rates in IBD patients from the ELYP cohort without or with endoscopic response to biotherapy. CTR: ileal mucosal samples) CD= Crohn’s Disease, CTR= Control, IBD= Inflammatory Bowel Disease, NR= Non Responders, R= Responders. *p < 0.05 Mann Whitney Test; +: p < 0.05 Wilcoxon Paired Test.
Figure 6
Figure 6
NKG2D expression on mucosal T cells is associated with IBD disease activity. (A) Gating strategy for CD45RO and NKG2D stainings in flow cytometry. (B) Comparison of mucosal rates of CD4 and CD8 T cells expressing NKG2D between controls and patients from the ELYP cohort at week 0. (C) Evolution of mucosal rate of CD8 T cells expressing NKG2D rates in IBD patients from the ELYP cohort without or with endoscopic response to biotherapy. (D) Evolution of mucosal rate of CD4 T cells expressing NKG2D rates in IBD patients from the ELYP cohort without or with endoscopic response to biotherapy. CD= Crohn’s Disease, CTR= Control, IBD= Inflammatory Bowel Disease, NR= Non Responders, R= Responders. *p < 0.05 Mann Whitney Test; +: p < 0.05 Wilcoxon Paired Test.

References

    1. Giuffrida P, Corazza GR, di Sabatino A. Old and new lymphocyte players in inflammatory bowel disease. Digestive Dis Sci (2018) 63:277–88. doi: 10.1007/s10620-017-4892-4
    1. Mackay L, Kallies A. Transcriptional regulation of tissue-resident lymphocytes. Trends Immunol (2017) 38:94–103. doi: 10.1016/j.it.2016.11.004
    1. Kumar B, Ma W, Miron M, Granot T, Guyer RS, Carpenter DJ, et al. . Human tissue-resident memory T cells are defined by core transcriptional and functional signatures in lymphoid and mucosal sites. Cell Rep (2017) 20:2921–34. doi: 10.1016/j.celrep.2017.08.078
    1. Schenkel J, Masopust D. Tissue-resident memory T cells. Immunity (2014) 41:886–97. doi: 10.1016/j.immuni.2014.12.007
    1. Mayer L, Eisenhardt D. Lack of induction of suppressor T cells by intestinal epithelial cells from patients with inflammatory bowel disease. J Clin Invest (1990) 86:1255–60. doi: 10.1172/JCI114832
    1. Dotan I, Allez M, Nakazawa A, Brimnes J, Schulder-Katz M, Mayer L. Intestinal epithelial cells from inflammatory bowel disease patients preferentially stimulate CD4+ T cells to proliferate and secrete interferon-γ. Am J Physiol Gastrointest Liver Physiol (2007) 292:G1630–40. doi: 10.1152/ajpgi.00294.2006
    1. Allez M, Brimnes J, Dotan I, Mayer L. Expansion of CD8+ T cells with regulatory function after interaction with intestinal epithelial cells. Gastroenterology (2002) 123:1516–26. doi: 10.1053/gast.2002.36588
    1. Brimnes J, Allez M, Dotan I, Shao L, Nakazawa A, Mayer L. Defects in CD8+ regulatory T cells in the lamina propria of patients with inflammatory bowel disease. J Immunol (2005) 174:5814–22. doi: 10.4049/jimmunol.174.9.5814
    1. Peterson LW, Artis D. Intestinal epithelial cells: Regulators of barrier function and immune homeostasis. Nat Rev Immunol (2014) 14:141–53. doi: 10.1038/nri3608
    1. Zundler S, Becker E, Schulze LL, Neurath MF. Immune cell trafficking and retention in inflammatory bowel disease: Mechanistic insights and therapeutic advances. Gut (2019) 68:1688–700. doi: 10.1136/gutjnl-2018-317977
    1. Ichikawa R, Lamb CA, Eastham-Anderson J, Scherl A, Raffals L, Faubion WA, et al. . AlphaE integrin expression is increased in the ileum relative to the colon and unaffected by inflammation. J Crohns Colitis (2018) 12:1191. doi: 10.1093/ecco-jcc/jjy084
    1. Park CO, Kupper TS. The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nat Med (2015) 21:688–97. doi: 10.1038/nm.3883
    1. Bottois H, Ngollo M, Hammoudi N, Courau T, Bonnereau J, Chardiny V, et al. . KLRG1 and CD103 expressions define distinct intestinal tissue-resident memory CD8 T cell subsets modulated in crohn’s disease. Front Immunol (2020) 11. doi: 10.3389/fimmu.2020.00896
    1. Groh V, Bahram S, Bauer S, Herman A, Beauchamp M, Spies T. Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium. Proc Natl Acad Sci USA (1996) 93:12445–50. doi: 10.1073/pnas.93.22.12445
    1. Vadstrup K, Galsgaard ED, Jensen H, Lanier LL, Ryan JC, Chen SY, et al. . NKG2D ligand expression in crohn’s disease and NKG2D-dependent stimulation of CD8+ T cell migration. Exp Mol Pathol (2017) 103:56–70. doi: 10.1016/j.yexmp.2017.06.010
    1. Meresse B, Chen Z, Ciszewski C, Tretiakova M, Bhagat G, Krausz TN, et al. . Coordinated induction by IL15 of a TCR-independent NKG2D signaling pathway converts CTL into lymphokine-activated killer cells in celiac disease. Immunity (2004) 21:357–66. doi: 10.1016/j.immuni.2004.06.020
    1. Setty M, Discepolo V, Abadie V, Kamhawi S, Mayassi T, Kent A, et al. . Distinct and synergistic contributions of epithelial stress and adaptive immunity to functions of intraepithelial killer cells and active celiac disease. Gastroenterology (2015) 149:681e10–691.e10. doi: 10.1053/j.gastro.2015.05.013
    1. Tang F, Sally B, Lesko K, Discepolo V, Abadie V, Ciszewski C, et al. . Cysteinyl leukotrienes mediate lymphokine killer activity induced by NKG2D and IL-15 in cytotoxic T cells during celiac disease. J Exp Med (2015) 212:1487–95. doi: 10.1084/jem.20150303
    1. Allez M, Tieng V, Nakazawa A, Treton X, Pacault V, Dulphy N, et al. . CD4+NKG2D+ T cells in crohn’s disease mediate inflammatory and cytotoxic responses through MICA interactions. Gastroenterology (2007) 132:2346–58. doi: 10.1053/j.gastro.2007.03.025
    1. Pariente B, Mocan I, Camus M, Dutertre CA, Ettersperger J, Cattan P, et al. . Activation of the receptor NKG2D leads to production of Th17 cytokines in CD4+ T cells of patients with crohn’s disease. Gastroenterology (2011) 141:217–26. doi: 10.1053/j.gastro.2011.03.061
    1. Camus M, Esses S, Pariente B, Le Bourhis L, Douay C, Chardiny V, et al. . Oligoclonal expansions of mucosal T cells in crohn’s disease predominate in NKG2D-expressing CD4 T cells. Mucosal Immunol (2014) 7:325–34. doi: 10.1038/mi.2013.51
    1. La Scaleia R, Stoppacciaro A, Oliva S, Morrone S, Di Nardo G, Santoni A, et al. . NKG2D/Ligand dysregulation and functional alteration of innate immunity cell populations in pediatric IBD. Inflammation Bowel Dis (2012) 18:1910–22. doi: 10.1002/ibd.22899
    1. Bar-Ephraim YE, Kretzschmar K, Clevers H. Organoids in immunological research. Nat Rev Immunol (2020) 20:279–93. doi: 10.1038/s41577-019-0248-y
    1. Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, et al. . Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and barrett’s epithelium. Gastroenterology (2011) 141:1762–72. doi: 10.1053/j.gastro.2011.07.050
    1. Arnauts K, Verstockt B, Ramalho AS, Vermeire S, Verfaillie C, Ferrante M. Ex vivo mimicking of inflammation in organoids derived from patients with ulcerative colitis. Gastroenterology (2020) 159:1564–7. doi: 10.1053/j.gastro.2020.05.064
    1. d'Aldebert E, Quaranta M, Sébert M, Bonnet D, Kirzin S, Portier G, et al. . Characterization of human colon organoids from inflammatory bowel disease patients. Front Cell Dev Biol (2020) 8. doi: 10.3389/fcell.2020.00363
    1. Biton M, Haber AL, Rogel N, Burgin G, Beyaz S, Schnell A, et al. . T Helper cell cytokines modulate intestinal stem cell renewal and differentiation. Cell (2018) 175:1307–1320.e22. doi: 10.1016/j.cell.2018.10.008
    1. Takashima S, Martin ML, Jansen SA, Fu Y, Bos J, Chandra D, et al. . T Cell-derived interferon-γ programs stem cell death in immune-mediated intestinal damage. Sci Immunol (2019) 4:8556. doi: 10.1126/sciimmunol.aay8556
    1. Schreider C, Peignon G, Thenet S, Chambaz J, Pinçon-Raymond M. Integrin-mediated functional polarization of caco-2 cells through e-cadherin–actin complexes. J Cell Sci (2002) 115:543–52. doi: 10.1242/jcs.115.3.543
    1. Wrobel P, Shojaei H, Schittek B, Gieseler F, Wollenberg B, Kalthoff H, et al. . Lysis of a broad range of epithelial tumour cells by human gamma delta T cells: Involvement of NKG2D ligands and T-cell receptor- versus NKG2D-dependent recognition. Scand J Immunol (2007) 66:320–8. doi: 10.1111/j.1365-3083.2007.01963.x
    1. Roosenboom B, Wahab PJ, Smids C, Groenen MJM, van Koolwijk E, van Lochem EG, et al. . Intestinal CD103+CD4+ and CD103+CD8+ T-cell subsets in the gut of inflammatory bowel disease patients at diagnosis and during follow-up. Inflammation Bowel Dis (2019) 25:1497–509. doi: 10.1093/ibd/izz049
    1. Smids C, Horjus Talabur Horje CS, Drylewicz J, Roosenboom B, Groenen MJM, van Koolwijk E, et al. . Intestinal T cell profiling in inflammatory bowel disease: Linking T cell subsets to disease activity and disease course. J Crohns Colitis (2018) 12:465–75. doi: 10.1093/ecco-jcc/jjx160
    1. Chakrabarti J, Koh V, So J, Yong W, Zavros Y. A preclinical human-derived autologous gastric cancer Organoid/Immune cell Co-culture model to predict the efficacy of targeted therapies. J Vis Exp (2021) (173). doi: 10.3791/61443
    1. Courau T, Bonnereau J, Chicoteau J, Bottois H, Remark R, Assante Miranda L, et al. . Cocultures of human colorectal tumor spheroids with immune cells reveal the therapeutic potential of MICA/B and NKG2A targeting for cancer treatment. J Immunother Cancer (2019) 7:74. doi: 10.1186/s40425-019-0553-9
    1. Stephens M, von der Weid PY. Lipopolysaccharides modulate intestinal epithelial permeability and inflammation in a species-specific manner. Gut Microbes (2020) 11:421–32. doi: 10.1080/19490976.2019.1629235
    1. Dai B, Hackney JA, Ichikawa R, Nguyen A, Elstrott J, Orozco LD, et al. . Dual targeting of lymphocyte homing andretention through alphaEbeta7 and alpha4beta7 inhibition in inflammatory bowel disease. Cell Rep Med (2021) 2:100381. doi: 10.1016/j.xcrm.2021.100381
    1. Allez M, Skolnick BE, Wisniewska-Jarosinska M, Petryka R, Overgaard RV. Anti-NKG2D monoclonal antibody (NNC0142-0002) in active crohn’s disease: A randomised controlled trial. Gut (2017) 66:1918–25. doi: 10.1136/gutjnl-2016-311824
    1. Sandborn WJ, Vermeire S, Tyrrell H, Hassanali A, Lacey S, Tole S, et al. . Etrolizumab for the treatment of ulcerative colitis and crohn’s disease: An overview of the phase 3 clinical program. Adv Ther (2020) 37:3417–31. doi: 10.1007/s12325-020-01366-2
    1. Jaeger N, Gamini R, Cella M, Schettini JL, Bugatti M, Zhao S, et al. . Single-cell analyses of crohn’s disease tissues reveal intestinal intraepithelial T cells heterogeneity and altered subset distributions. Nat Commun (2021) 12:1921. doi: 10.1038/s41467-021-22164-6
    1. Bartolomé-Casado R, Landsverk OJB, Chauhan SK, Richter L, Phung D, Greiff V, et al. . Resident memory CD8 T cells persist for years in human small intestine. J Exp Med (2019) 216:2412–26. doi: 10.1084/jem.20190414
    1. Enderle K, Dinkel M, Spath EM, Schmid B, Zundler S, Tripal P, et al. . Dynamic imaging of iel-iec co-cultures allows for quantification of cd103-dependent t cell migration. Int J Mol Sci (2021) 22:5148. doi: 10.3390/ijms22105148
    1. Habtezion A, Nguyen LP, Hadeiba H, Butcher EC. Leukocyte trafficking to the small intestine and colon. Gastroenterology (2016) 150:340. doi: 10.1053/j.gastro.2015.10.046
    1. Ma H, Tao W, Zhu S. T Lymphocytes in the intestinal mucosa: defense and tolerance. Cell Mol Immunol (2019) 16:216. doi: 10.1038/s41423-019-0208-2
    1. Sakai T, Kusugami K, Nishimura H, Ando T, Yamaguchi T, Ohsuga M, et al. . Interleukin 15 activity in the rectal mucosa of inflammatory bowel disease. Gastroenterology (1998) 114:1237–43. doi: 10.1016/S0016-5085(98)70430-5
    1. Liu Z, Geboes K, Colpaert S, D'Haens GR, Rutgeerts P, Ceuppens JL. IL-15 is highly expressed in inflammatory bowel disease and regulates local T cell-dependent cytokine production. J Immunol (2000) 164:3608–15. doi: 10.4049/jimmunol.164.7.3608
    1. Abadie V, Kim SM, Lejeune T, Palanski BA, Ernest JD, Tastet O, et al. . IL-15, gluten and HLA-DQ8 drive tissue destruction in coeliac disease. Nature (2020) 578:600–4. doi: 10.1038/s41586-020-2003-8

Source: PubMed

3
S'abonner