Cell-Matrix Interactions Contribute to Barrier Function in Human Colon Organoids

James Varani, Shannon D McClintock, Muhammad N Aslam, James Varani, Shannon D McClintock, Muhammad N Aslam

Abstract

The importance of cell-matrix adhesion to barrier control in the colon is unclear. The goals of the present study were to: (i) determine if disruption of colon epithelial cell interactions with the extracellular matrix alters permeability control measurement and (ii) determine if increasing the elaboration of protein components of cell-matrix adhesion complexes can mitigate the effects of cell-matrix disruption. Human colon organoids were interrogated for transepithelial electrical resistance (TEER) under control conditions and in the presence of Aquamin®, a multi-mineral product. A function-blocking antibody directed at the C-terminal region of the laminin α chain was used in parallel. The effects of Aquamin® on cell-matrix adhesion protein expression were determined in a proteomic screen and by Western blotting. Aquamin® increased the expression of multiple basement membrane, hemidesmosomal and focal adhesion proteins as well as keratin 8 and 18. TEER values were higher in the presence of Aquamin® than they were under control conditions. The blocking antibody reduced TEER values under both conditions but was most effective in the absence of Aquamin®, where expression of cell-matrix adhesion proteins was lower to begin with. These findings provide evidence that cell-matrix interactions contribute to barrier control in the colon.

Keywords: Aquamin®; basement membrane; cell-cell junction; cell-matrix adhesion; colonoid; gut barrier; laminin; proteomics.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Varani, McClintock and Aslam.

Figures

Figure 1
Figure 1
Effects of Aquamin® on expression of proteins involved in cell-matrix interactions: Proteomic screen. Organoid cultures were established with colon biopsies from four healthy subjects and three with ulcerative colitis in remission. Organoids were cultured in the presence or absence of Aquamin® as described in the Materials and Methods section. At the end of the incubation period, protein was isolated from the cultures and assessed in the proteomic screen. Values from respective controls were set at 1.0 and the values from Aquamin®-treated samples reflect a ratio (increase or decrease fold-change) relative to respective control. Values shown reflect mean (±SD) fold-change from respective control in colon organoids from normal subjects (N = 4) and subjects with UC in remission (N = 3). Methods for generating the data sets were described in detail in past reports (22, 24). Data for individual proteins were compared for statistical differences using the student t-test. Asterisk (*) indicates a difference from respective control at p < 0.05.
Figure 2
Figure 2
Effects of Aquamin® on expression of Laminin β1: Western blotting. Protein isolated from control and Aquamin®-treated (healthy normal subjects) colon organoid-derived monolayer cells was assessed for laminin β1 expression by Western blotting as described in the Materials and Methods section. 10 μg of protein from each condition was used. β-actin was assessed in parallel (as a loading control). Band quantitation was done using ImageJ software. Relative band density is presented for laminin β1 and β-actin.
Figure 3
Figure 3
Transepithelial electrical resistance in differentiation medium (Preliminary assessment). (A) Time-dependent changes in TEER values. Values shown are means and standard deviations based on four separate experiments with four samples (individual transwell insert filters seeded with healthy colon organoid-derived monolayer cells) per data point at each time-point in each experiment. Insert: Confocal fluorescent microscopic (max-projected) images of organoids and organoid-derived cells on transwell inserts stained after the day-2 and day-5 readings with antibody to occludin and with a combination of antibody to occludin and DAPI. Scale bars = 50 μm. (B) Effects of anti-laminin antibody on TEER values. Values shown are means and standard deviations based on two separate experiments with 4 samples (individual transwell insert filters seeded with healthy colon organoid-derived monolayer cells) per data point at each time-point in each experiment. Insert: hematoxylin and eosin-stained images of the cell monolayers still attached to the transwell inserts from IgG-treated and anti-laminin-treated wells. Arrows in the anti-laminin-treated image show areas where cell detachment from the underlying transwell insert was visible. Scale bar = 100 μm (small) and 50 μm (Large).
Figure 4
Figure 4
Transepithelial electrical resistance in KGM Gold-growth medium with or without Aquamin® and with or without anti-laminin antibody. TEER values shown are means and standard deviations based on three separate experiments with four samples (individual transwell insert filters seeded with healthy colon organoid-derived monolayer cells) per data point in each experiment. Data were compared for statistical differences using ANOVA followed by unpaired-group comparisons. Asterisk (*) above the open Aquamin® bar indicates a difference from control at p < 0.05. Asterisks (*) above the closed bars indicates difference from respective IgG control at p < 0.05.
Figure 5
Figure 5
Colon organoid cohesion in KGM Gold-growth medium mix: Effect of anti-laminin antibody. Colon organoids were maintained for 7 days in KGM Gold-growth medium with either IgG or anti-laminin. At the end of the incubation period, organoid cohesion was assessed as described in the Materials and Methods section. Values shown represent the change in organoid size (i.e., mean surface area ± SD of individual colon organoids based on two separate experiments with a minimum of 53-104 colon or-ganoids assessed individually per treatment group in both pre- and post-harvest cultures. Data were compared for statistical differences using ANOVA followed by unpaired-group comparisons. While the decrease in organoid size between post-harvest and pre-harvest organoids were statistically significant with either IgG or anti-laminin, the differences between anti-laminin and IgG were not different. Inset: Representative examples of organoid appearance immediately prior to harvest (upper) and 1 day after the harvested organoids had been reestablished in culture. Scale bar = 200 μm.
Figure 6
Figure 6
Aquamin®-responsive cell-matrix adhesion structures in the colon. A cartoon depicting structures important to cell-matrix adhesion in the colon and components of those structures that are responsive to Aquamin® (shown in red). Based on the profile of proteins that are induced by Aquamin®, cell-matrix adhesion through both focal adhesions and hemidesmosomes could be affected. Laminin-binding integrins did not alter with Aquamin®.

References

    1. Salim SY, Söderholm JD. Importance of disrupted intestinal barrier in inflammatory bowel diseases. Inflamm Bowel Dis. (2011) 17:362-81. 10.1002/ibd.21403
    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. Lee JY, Wasinger VC, Yau YY, Chuang E, Yajnik V, Leong RW. Molecular pathophysiology of epithelial barrier dysfunction in inflammatory bowel diseases. Proteomes. (2018) 6:17. 10.3390/proteomes6020017
    1. Vivinus-Nébot M, Frin-Mathy G, Bzioueche H, Dainese R, Bernard G, Anty R, et al. . Functional bowel symptoms in quiescent inflammatory bowel diseases: role of epithelial barrier disruption and low-grade inflammation. Gut. (2014) 63:744-52. 10.1136/gutjnl-2012-304066
    1. Pearson AD, Eastham EJ, Laker MF, Craft AW, Nelson R. Intestinal permeability in children with Crohn's disease and coeliac disease. Br Med J. (1982) 285:20-1. 10.1136/bmj.285.6334.20
    1. Luissint AC, Parkos CA, Nusrat A. Inflammation and the intestinal barrier: leukocyte-epithelial cell interactions, cell junction remodeling, and mucosal repair. Gastroenterology. (2016) 151:616-32. 10.1053/j.gastro.2016.07.008
    1. Dunlop SP, Hebden J, Campbell E, Naesdal J, Olbe L, Perkins AC, et al. . Abnormal intestinal permeability in subgroups of diarrhea-predominant irritable bowel syndromes. Am J Gastroenterol. (2006) 101:1288-94. 10.1111/j.1572-0241.2006.00672.x
    1. Flügel A, Schulze-Koops H, Heesemann J, Kühn K, Sorokin L, Burkhardt H, et al. . Interaction of enteropathogenic Yersinia enterocolitica with complex basement membranes and the extracellular matrix proteins collagen type IV, laminin-1 and−2, and nidogen/entactin. J Biol Chem. (1994) 269:29732-8. 10.1016/S0021-9258(18)43942-7
    1. Moreira AP, Texeira TF, Ferreira AB, Peluzio Mdo C, Alfenas Rde C. Influence of a high-fat diet on gut microbiota, intestinal permeability and metabolic endotoxaemia. Br J Nutr. (2012) 108:801-9. 10.1017/S0007114512001213
    1. Thaiss CA, Levy M, Grosheva I, Zheng D, Soffer E, Blacher E, et al. . Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science. (2018) 359:1376-83. 10.1126/science.aar3318
    1. Meddings JB, Swain MG. Environmental stress-induced gastrointestinal permeability is mediated by endogenous glucocorticoids in the rat. Gastroenterology. (2000) 119:1019-28. 10.1053/gast.2000.18152
    1. Clayburgh DR, Shen L, Turner JR. A porous defense: the leaky epithelial barrier in intestinal disease. Lab Invest. (2004) 84:282-91. 10.1038/labinvest.3700050
    1. Turner JR. Molecular basis of epithelial barrier regulation: from basic mechanisms to clinical application. Am J Pathol. (2006) 169:1901-9. 10.2353/ajpath.2006.060681
    1. Shen L, Su L, Turner JR. Mechanisms and functional implications of intestinal barrier defects. Dig Dis. (2009) 27:443-9. 10.1159/000233282
    1. Turner JR. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol. (2009) 9:799-809. 10.1038/nri2653
    1. Camilleri M, Madsen K, Spiller R, Greenwood-Van Meerveld B, Verne GN. Intestinal barrier function in health and gastrointestinal disease. Neurogastroenterol Motil. (2012) 24:503-12. 10.1111/j.1365-2982.2012.01921.x
    1. Cereijido M, Valdés J, Shoshani L, Contreras RG. Role of tight junctions in establishing and maintaining cell polarity. Annu Rev Physiol. (1998) 60:161-77. 10.1146/annurev.physiol.60.1.161
    1. Aijaz S, Balda MS, Matter K. Tight junctions: molecular architecture and function. Int Rev Cytol. (2006) 248:261-98. 10.1016/S0074-7696(06)48005-0
    1. Green KJ, Simpson CL. Desmosomes: new perspectives on a classic. J Invest Dermatol. (2007) 127:2499-515. 10.1038/sj.jid.5701015
    1. Kowalczyk AP, Green KJ. Structure, function, and regulation of desmosomes. Prog Mol Biol Transl Sci. (2013) 116:95-118. 10.1016/B978-0-12-394311-8.00005-4
    1. Adey WH, McKibbin DL. Studies on the maerl species Phymatolithon calcareum (Pallas) nov. comb. and Lithothamnium corallioides Crouan in the Ria de Vigo. Botanical Marina. (1970) 13:100–6. 10.1515/botm.1970.13.2.100
    1. Attili D, McClintock SD, Rizvi AH, Pandya S, Rehman H, Nadeem DM, et al. . Calcium-induced differentiation in normal human colonoid cultures: Cell-cell / cell-matrix adhesion, barrier formation and tissue integrity. PLoS ONE. (2019) 14:e0215122. 10.1371/journal.pone.0215122
    1. McClintock SD, Attili D, Dame MK, Richter A, Silvestri SS, Berner MM, et al. . Differentiation of human colon tissue in culture: effects of calcium on trans-epithelial electrical resistance and tissue cohesive properties. PLoS ONE. (2020) 15:e0222058. 10.1371/journal.pone.0222058
    1. Aslam MN, McClintock SD, Attili D, Pandya S, Rehman H, Nadeem DM, et al. . Ulcerative colitis-derived colonoid culture: a multi-mineral-approach to improve barrier protein expression. Front Cell Dev Biol. (2020) 8:577221. 10.3389/fcell.2020.577221
    1. Schmehl K, Florian S, Jacobasch G, Salomon A, Körber J. Deficiency of epithelial basement membrane laminin in ulcerative colitis affected human colonic mucosa. Int J Colorectal Dis. (2000) 15:39-48. 10.1007/s003840050006
    1. Verbeke S, Gotteland M, Fernández M, Bremer J, Ríos G, Brunser O. Basement membrane and connective tissue proteins in intestinal mucosa of patients with coeliac disease. J Clin Pathol. (2002) 55:440-5. 10.1136/jcp.55.6.440
    1. Bouatrouss Y, Herring-Gillam FE, Gosselin J, Poisson J, Beaulieu JF. Altered expression of laminins in Crohn's disease small intestinal mucosa. Am J Pathol. (2000) 156:45-50. 10.1016/S0002-9440(10)64704-9
    1. Spenlé C, Lefebvre O, Lacroute J, Méchine-Neuville A, Barreau F, Blottière HM, et al. . The laminin response in inflammatory bowel disease: protection or malignancy? PLoS ONE. (2014) 9:e111336. 10.1371/journal.pone.0111336
    1. Timpl R, Tisi D, Talts JF, Andac Z, Sasaki T, Hohenester E. Structure and function of laminin LG modules. Matrix Biol. (2000) 19:309-17. 10.1016/S0945-053X(00)00072-X
    1. Colognato H, Yurchenco PD. Form and function: the laminin family of heterotrimers. Dev Dyn. (2000) 218:213-34. 10.1002/(SICI)1097-0177(200006)218:2<213::AID-DVDY1>;2-R
    1. Turck N, Gross I, Gendry P, Stutzmann J, Freund JN, Kedinger M, et al. . Laminin isoforms: biological roles and effects on the intracellular distribution of nuclear proteins in intestinal epithelial cells. Exp Cell Res. (2005) 303:494-503. 10.1016/j.yexcr.2004.10.025
    1. Gonzales M, Haan K, Baker SE, Fitchmun M, Todorov I, Weitzman S, et al. . A cell signal pathway involving laminin-5, alpha3beta1 integrin, and mitogen-activated protein kinase can regulate epithelial cell proliferation. Mol Biol Cell. (1999) 10:259-70. 10.1091/mbc.10.2.259
    1. Miyoshi H, Stappenbeck TS. In vitro expansion and genetic modification of gastrointestinal stem cells in spheroid culture. Nat Protoc. (2013) 8:2471-82. 10.1038/nprot.2013.153
    1. Zou WY, Blutt SE, Crawford SE, Ettayebi K, Zeng XL, Saxena K, et al. . Human intestinal enteroids: new models to study gastrointestinal virus infections. Methods Mol Biol. (2019) 1576:229-47. 10.1007/7651_2017_1
    1. Fabregat A, Sidiropoulos K, Viteri G, Forner O, Marin-Garcia P, Arnau V, et al. . Reactome pathway analysis: a high-performance in-memory approach. BMC Bioinformatics. (2017) 18:142. 10.1186/s12859-017-1559-2
    1. Sekiguchi R, Yamada KM. Basement membranes in development and disease. Curr Top Dev Biol. (2018) 130:143-91. 10.1016/bs.ctdb.2018.02.005
    1. Pozzi A, Yurchenco PD, Iozzo RV. The nature and biology of basement membranes. Matrix Biol. (2017) 57-58:1-11. 10.1016/j.matbio.2016.12.009
    1. Baker SE, Hopkinson SB, Fitchmun M, Andreason GL, Frasier F, Plopper G, et al. . Laminin-5 and hemidesmosomes: role of the alpha 3 chain subunit in hemidesmosome stability and assembly. J Cell Sci. (1996) 109:2509-20. 10.1242/jcs.109.10.2509
    1. Green KJ, Jones JC. Desmosomes and hemidesmosomes: structure and function of molecular components. FASEB J. (1996) 10:871-81. 10.1096/fasebj.10.8.8666164
    1. Chidgey M. Plakin Proteins, Hemidesmosomes and Human Disease. In: eLS editor. John Wiley & Sons; (2012). 10.1002/9780470015902.a0024527
    1. Has C. Hemidesmosomes: how much plakins do they need? Exp Dermatol. (2016) 25:263-4. 10.1111/exd.12939
    1. Polari L, Alam CM, Nyström JH, Heikkilä T, Tayyab M, Baghestani S, et al. . Keratin intermediate filaments in the colon: guardians of epithelial homeostasis. Int J Biochem Cell Biol. (2020) 129:105878. 10.1016/j.biocel.2020.105878
    1. Zupancic T, Stojan J, Lane EB, Komel R, Bedina-Zavec A, Liovic M. Intestinal cell barrier function in vitro is severely compromised by keratin 8 and 18 mutations identified in patients with inflammatory bowel disease. PLoS ONE. (2014) 9:e99398. 10.1371/journal.pone.0099398
    1. Corfe BM, Majumdar D, Assadsangabi A, Marsh AM, Cross SS, Connolly JB, et al. . Inflammation decreases keratin level in ulcerative colitis; inadequate restoration associates with increased risk of colitis-associated cancer. BMJ Open Gastroenterol. (2015) 2:e000024. 10.1136/bmjgast-2014-000024
    1. Nishiuchi R, Takagi J, Hayashi M, Ido H, Yagi Y, Sanzen N, et al. . Ligand-binding specificities of laminin-binding integrins: a comprehensive survey of laminin-integrin interactions using recombinant alpha3beta1, alpha6beta1, alpha7beta1 and alpha6beta4 integrins. Matrix Biol. (2006) 25:189-97. 10.1016/j.matbio.2005.12.001
    1. De Arcangelis A, Hamade H, Alpy F, Normand S, Bruyère E, Lefebvre O, et al. . Hemidesmosome integrity protects the colon against colitis and colorectal cancer. Gut. (2017) 66:1748-60. 10.1136/gutjnl-2015-310847
    1. Stutzmann J, Bellissent-Waydelich A, Fontao L, Launay JF, Simon-Assmann P. Adhesion complexes implicated in intestinal epithelial cell-matrix interactions. Microsc Res Tech. (2000) 51:179-90. 10.1002/1097-0029(20001015)51:2<179::AID-JEMT9>;2-4
    1. Rousselle P, Lunstrum GP, Keene DR, Burgeson RE. Kalinin: an epithelium-specific basement membrane adhesion molecule that is a component of anchoring filaments. J Cell Biol. (1991) 114:567-76. 10.1083/jcb.114.3.567
    1. Hennings H, Holbrook KA. Calcium regulation of cell-cell contact and differentiation of epidermal cells in culture. An ultrastructural study. Exp Cell Res. (1983) 143:127-42. 10.1016/0014-4827(83)90115-5
    1. Huang Y, Zhou Y, Castiblanco A, Yang W, Brown EM, Yang JJ. Multiple Ca(2+)-binding sites in the extracellular domain of the Ca(2+)-sensing receptor corresponding to cooperative Ca(2+) response. Biochemistry. (2009) 48:388-98. 10.1021/bi8014604
    1. Singh N, Aslam MN, Varani J, Chakrabarty S. Induction of calcium sensing receptor in human colon cancer cells by calcium, vitamin D and aquamin: promotion of a more differentiated, less malignant and indolent phenotype. Mol Carcinog. (2015) 54:543-53. 10.1002/mc.22123
    1. Carrillo-López N, Fernández-Martín JL, Alvarez-Hernández D, González-Suárez I, Castro-Santos P, Román-García P, et al. . Lanthanum activates calcium-sensing receptor and enhances sensitivity to calcium. Nephrol Dial Transplant. (2010) 25:2930-7. 10.1093/ndt/gfq124
    1. Tiwari S, Askari JA, Humphries MJ, Bulleid NJ. Divalent cations regulate the folding and activation status of integrins during their intracellular trafficking. J Cell Sci. (2011) 124:1672-80. 10.1242/jcs.084483
    1. Varani J, McClintock SD, Aslam MN. Organoid culture to study epithelial cell differentiation and barrier formation in the colon: bridging the gap between monolayer cell culture and human subject research. In Vitro Cell Dev Biol Anim. (2021) 57:174-90. 10.1007/s11626-020-00534-6
    1. Aslam MN, Bassis CM, Bergin IL, Knuver K, Zick SM, Sen A, et al. . A calcium-rich multimineral intervention to modulate colonic microbial communities and metabolomic profiles in humans: results from a 90-day trial. Cancer Prev Res. (2020) 13:101-16. 10.1158/1940-6207.CAPR-19-0325
    1. Aslam MN, McClintock SD, Jawad-Makki MAH, Knuver K, Ahmad HM, Basrur V, et al. . A multi-mineral intervention to modulate colonic mucosal protein profile: results from a 90-day trial in human subjects. Nutrients. (2021) 13:939. 10.3390/nu13030939
    1. Sequeira IR, Lentle RG, Kruger MC, Hurst RD. Standardising the lactulose mannitol test of gut permeability to minimise error and promote comparability. PLoS ONE. (2014) 9:e99256. 10.1371/journal.pone.0099256
    1. Balk EM, Adam GP, Langberg VN, Earley A, Clark P, Ebeling PR, et al. . International osteoporosis foundation calcium steering committee. global dietary calcium intake among adults: a systematic review. Osteoporos Int. (2017) 28:3315-24. 10.1007/s00198-017-4230-x
    1. Aslam MN, Varani J. The western-style diet, calcium deficiency and chronic disease. J Nutr Food Sci. (2016) 6:3. 10.4172/2155-9600.1000496
    1. U.S. Department of Health and Human Services; U.S. Department of Agriculture . 2015-−2020 Dietary Guidelines for Americans. 8th ed. Washington, DC: U.S. Department of Health and Human Services; (2015).
    1. Fine JD, Eady RA, Bauer EA, Bauer JW, Bruckner-Tuderman L, Heagerty A, et al. . The classification of inherited epidermolysis bullosa (EB): report of the third international consensus meeting on diagnosis and classification of EB. J Am Acad Dermatol. (2008) 58:931-50. 10.1016/j.jaad.2008.02.004
    1. Sachsenberg-Studer EM, Runne U, Wehrmann T, Wolter M, Kriener S, Engels K, et al. . Bullous colon lesions in a patient with bullous pemphigoid. Gastrointest Endosc. (2001) 54:104-8. 10.1067/mge.2001.115472
    1. Seo JW, Park J, Lee J, Kim MY, Choi HJ, Jeong HJ, et al. . A case of pemphigus vulgaris associated with ulcerative colitis. Intest Res. (2018) 16:147-50. 10.5217/ir.2018.16.1.147
    1. Chen YJ, Juan CK, Chang YT, Wu CY, Ho HJ, Tseng HC. Association between inflammatory bowel disease and bullous pemphigoid: a population-based case-control study. Sci Rep. (2020) 10:12727. 10.1038/s41598-020-69475-0
    1. Natsuga K, Shinkuma S, Nishie W, Shimizu H. Animal models of epidermolysis bullosa. Dermatol Clin. (2010) 28:137-42. 10.1016/j.det.2009.10.016
    1. Bruckner-Tuderman L, McGrath JA, Robinson EC, Uitto J. Animal models of epidermolysis bullosa: update 2010. J Invest Dermatol. (2010) 130:1485-8. 10.1038/jid.2010.75
    1. Heimbach L, Li N, Diaz A, Liu Z. Experimental animal models of bullous pemphigoid. G Ital Dermatol Venereol. (2009) 144:423-31.

Source: PubMed

3
Subscribe