Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Protocol
  • Published:

In vivo labeling of epithelial cell–associated antigen passages in the murine intestine

Abstract

The intestinal immune system samples luminal contents to induce adaptive immune responses that include tolerance in the steady state and protective immunity during infection. How luminal substances are delivered to the immune system has not been fully investigated. Goblet cells have an important role in this process by delivering luminal substances to the immune system through the formation of goblet cell–associated antigen passages (GAPs). Soluble antigens in the intestinal lumen are transported across the epithelium transcellularly through GAPs and delivered to dendritic cells for presentation to T cells and induction of immune responses. GAPs can be identified and quantified by using the ability of GAP-forming goblet cells to take up fluorescently labeled dextran. Here, we describe a method to visualize GAPs and other cells that have the capacity to take up luminal substances by intraluminal injection of fluorescent dextran in mice under anesthesia, tissue sectioning for slide preparation and imaging with fluorescence microscopy. In contrast to in vivo two-photon imaging previously used to identify GAPs, this technique is not limited by anatomical constraints and can be used to visualize GAP formation throughout the length of the intestine. In addition, this method can be combined with common immunohistochemistry protocols to visualize other cell types. This approach can be used to compare GAP formation following different treatments or changes to the luminal environment and to uncover how sampling of luminal substances is altered in pathophysiological conditions. This protocol requires 8 working hours over 2–3 d to be completed.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1
Fig. 2: GAP formation in the small intestine.
Fig. 3: GAP formation per goblet cell.
Fig. 4: GAP formation in the colon.

Similar content being viewed by others

References

  1. McDole, J. R. et al. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature 483, 345–349 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Knoop, K. A., McDonald, K. G., McCrate, S., McDole, J. R. & Newberry, R. D. Microbial sensing by goblet cells controls immune surveillance of luminal antigens in the colon. Mucosal Immunol. 8, 198–210 (2015).

    CAS  PubMed  Google Scholar 

  3. Knoop, K. A. et al. Antibiotics promote the sampling of luminal antigens and bacteria via colonic goblet cell associated antigen passages. Gut Microbes 8, 400–411 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Kulkarni, D. H. et al. Goblet cell associated antigen passages are inhibited during Salmonella typhimurium infection to prevent pathogen dissemination and limit responses to dietary antigens. Mucosal Immunol. 11, 1103–1113 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Knoop, K. A. et al. Microbial antigen encounter during a preweaning interval is critical for tolerance to gut bacteria. Sci. Immunol. 2, eaa01314 (2017).

    Google Scholar 

  6. Yu, Q. H. & Yang, Q. Diversity of tight junctions (TJs) between gastrointestinal epithelial cells and their function in maintaining the mucosal barrier. Cell Biol. Int. 33, 78–82 (2009).

    PubMed  Google Scholar 

  7. Turner, J. R. Intestinal mucosal barrier function in health and disease. Nat. Rev. Immunol. 9, 799–809 (2009).

    CAS  PubMed  Google Scholar 

  8. Heller, F. et al. Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology 129, 550–564 (2005).

    CAS  PubMed  Google Scholar 

  9. Chang, J. et al. Impaired intestinal permeability contributes to ongoing bowel symptoms in patients with inflammatory bowel disease and mucosal healing. Gastroenterology 153, 723–731.e1 (2017).

    PubMed  Google Scholar 

  10. Michielan, A. & D’Incà, R. Intestinal permeability in inflammatory bowel disease: pathogenesis, clinical evaluation, and therapy of leaky gut. Mediators Inflamm. 2015, 628157 (2015).

    PubMed  PubMed Central  Google Scholar 

  11. Cukrowska, B. et al. Intestinal epithelium, intraepithelial lymphocytes and the gut microbiota—key players in the pathogenesis of celiac disease. World J. Gastroenterol. 23, 7505–7518 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Obrenovich, M. E. M. Leaky gut, leaky brain? Microorganisms 6, E107 (2018).

    PubMed  Google Scholar 

  13. Hamilton, M. K. & Raybould, H. E. Bugs, guts and brains, and the regulation of food intake and body weight. Int. J. Obes. Suppl. 6, S8–S14 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Araújo, J. R., Tomas, J., Brenner, C. & Sansonetti, P. J. Impact of high-fat diet on the intestinal microbiota and small intestinal physiology before and after the onset of obesity. Biochimie 141, 97–106 (2017).

    PubMed  Google Scholar 

  15. Shen, L., Weber, C. R., Raleigh, D. R., Yu, D. & Turner, J. R. Tight junction pore and leak pathways: a dynamic duo. Annu. Rev. Physiol. 73, 283–309 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Utzeri, E. & Usai, P. Role of non-steroidal anti-inflammatory drugs on intestinal permeability and nonalcoholic fatty liver disease. World J. Gastroenterol. 23, 3954–3963 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Knoop, K. A., Miller, M. J. & Newberry, R. D. Transepithelial antigen delivery in the small intestine: different paths, different outcomes. Curr. Opin. Gastroenterol. 29, 112–118 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Volynets, V. et al. Assessment of the intestinal barrier with five different permeability tests in healthy C57BL/6J and BALB/cJ mice. Dig. Dis. Sci. 61, 737–746 (2016).

    CAS  PubMed  Google Scholar 

  19. Wang, L. et al. Methods to determine intestinal permeability and bacterial translocation during liver disease. J. Immunol. Methods 421, 44–53 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Wu, L.-L. et al. Commensal bacterial endocytosis in epithelial cells is dependent on myosin light chain kinase-activated brush border fanning by interferon-γ. Am. J. Pathol. 184, 2260–2274 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Rock, K. L., Reits, E. & Neefjes, J. Present yourself! By MHC class I and MHC class II molecules. Trends Immunol. 37, 724–737 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Blum, J. S., Wearsch, P. A. & Cresswell, P. Pathways of antigen processing. Annu. Rev. Immunol. 31, 443–473 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Kang, S. K. et al. Identification of a peptide sequence that improves transport of macromolecules across the intestinal mucosal barrier targeting goblet cells. J. Biotechnol. 135, 210–216 (2008).

    CAS  PubMed  Google Scholar 

  24. Jin, Y. et al. Goblet cell-targeting nanoparticles for oral insulin delivery and the influence of mucus on insulin transport. Biomaterials 33, 1573–1582 (2012).

    CAS  PubMed  Google Scholar 

  25. Fan, T. et al. Design and evaluation of solid lipid nanoparticles modified with peptide ligand for oral delivery of protein drugs. Eur. J. Pharm. Biopharm. 88, 518–528 (2014).

    CAS  PubMed  Google Scholar 

  26. Lee, J. Y. et al. Production of recombinant human growth hormone conjugated with a transcytotic peptide in Pichia pastoris for effective oral protein delivery. Mol. Biotechnol. 57, 430–438 (2015).

    CAS  PubMed  Google Scholar 

  27. Xia, D. et al. Wheat germ agglutinin nanocage stabilized drug nanocrystals cross intestinal epithelium barrier via goblet cells. J. Control Release 213, e25– e26 (2015).

    PubMed  Google Scholar 

  28. Xia, D. et al. Enhanced transport of nanocage stabilized pure nanodrug across intestinal epithelial barrier mimicking Listeria monocytogenes. Biomaterials 37, 320–332 (2015).

    CAS  PubMed  Google Scholar 

  29. Kenngott, E. E. et al. Identification of targeting peptides for mucosal delivery in sheep and mice. Mol. Pharm. 13, 202–210 (2016).

    CAS  PubMed  Google Scholar 

  30. Kiliaan, A. J. et al. Stress stimulates transepithelial macromolecular uptake in rat jejunum. Am. J. Physiol. 275, G1037– G1044 (1998).

    CAS  PubMed  Google Scholar 

  31. Madara, J. L. & Trier, J. S. Structure and permeability of goblet cell tight junctions in rat small intestine. J. Membr. Biol. 66, 145–157 (1982).

    CAS  PubMed  Google Scholar 

  32. Barbour, W. M. & Hopwood, D. Uptake of cationized ferritin by colonic epithelium. J. Pathol. 139, 167–178 (1983).

    CAS  PubMed  Google Scholar 

  33. Colony, P. C. & Specian, R. D. Endocytosis and vesicular traffic in fetal and adult colonic goblet cells. Anat. Rec. 218, 365–372 (1987).

    CAS  PubMed  Google Scholar 

  34. Knoop, K. A., McDonald, K. G., Kulkarni, D. H. & Newberry, R. D. Antibiotics promote inflammation through the translocation of native commensal colonic bacteria. Gut 65, 1100–1109 (2016).

    CAS  PubMed  Google Scholar 

  35. Nagatake, T., Fujita, H., Minato, N. & Hamazaki, Y. Enteroendocrine cells are specifically marked by cell surface expression of claudin-4 in mouse small intestine. PLoS ONE 9, e90638 (2014).

    PubMed  PubMed Central  Google Scholar 

  36. Barbosa, F. L. et al. Goblet cells contribute to ocular surface immune tolerance-implications for dry eye disease. Int. J. Mol. Sci. 18, E978 (2017).

    PubMed  Google Scholar 

  37. Mammoto, A. et al. Control of lung vascular permeability and endotoxin-induced pulmonary oedema by changes in extracellular matrix mechanics. Nat. Commun. 4, 1759 (2013).

    PubMed  Google Scholar 

  38. Ko, B. Y., Xiao, Y., Barbosa, F. L., de Paiva, C. S. & Pflugfelder, S. C. Goblet cell loss abrogates ocular surface immune tolerance. JCI Insight 3, 98222 (2018).

    PubMed  Google Scholar 

  39. Weiner, M. L. Intestinal transport of some macromolecules in food. Food Chem. Toxicol. 26, 867–880 (1988).

    CAS  PubMed  Google Scholar 

  40. Specian, R. D. & Neutra, M. R. Mechanism of rapid mucus secretion in goblet cells stimulated by acetylcholine. J. Cell Biol. 85, 626–640 (1980).

    CAS  PubMed  Google Scholar 

  41. Neutra, M. R., O’Malley, L. J. & Specian, R. D. Regulation of intestinal goblet cell secretion. II. A survey of potential secretagogues. Am. J. Physiol. 242, G380– G387 (1982).

    CAS  PubMed  Google Scholar 

  42. Pickett, J. A. & Edwardson, J. M. Compound exocytosis: mechanisms and functional significance. Traffic 7, 109–116 (2006).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by grants AI136515, AI140755, AI12626, AI131342 and DK097317 (to R.D.N.); grants DK052574, DK09789 and AI095542 (to K.A.K.); Swedish Research Council International Postdoc Award 2014-00366 (to J.K.G.); and Crohn’s and Colitis Foundation 610605 (to D.H.K.).

Author information

Authors and Affiliations

Authors

Contributions

All authors conceived the protocol and reviewed and discussed the manuscript. K.A.K. and D.H.K. prepared the figures. K.A.K., J.E.D. and A.N.F. prepared the supplementary videos.

Corresponding author

Correspondence to Rodney D. Newberry.

Ethics declarations

Competing interests

R.D.N., K.A.K. and K.G.M. are inventors on the US Non-provisional Application Serial No. 15/880,658: Compositions and methods for modulation of dietary and microbial exposure.

Supplementary information

Reporting Summary

Supplementary Video 1

Mouse surgery and intraluminal dextran administration

Supplementary Video 2

Embedding and freezing of tissue

Supplementary Video 3

Sectioning of tissue for slide preparation

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Knoop, K.A., Kulkarni, D.H., McDonald, K.G. et al. In vivo labeling of epithelial cell–associated antigen passages in the murine intestine. Lab Anim 49, 79–88 (2020). https://doi.org/10.1038/s41684-019-0438-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41684-019-0438-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing