Skip to main content

Advertisement

Log in

Organoids transplantation as a new modality to design epithelial signature to create a membrane-protective sulfomucin-enriched segment

  • Original Article—Alimentary Tract
  • Published:
Journal of Gastroenterology Aims and scope Submit manuscript

Abstract

Background

The organoids therapy for ulcerative colitis (UC) is under development. It is important to dissect how the engrafted epithelium can provide benefits for overcoming the vulnerability to inflammation. We mainly focused on the deliverability of sulfomucin, which is reported to play an important role in epithelial function.

Methods

We analyzed each segment of colon epithelium to determine differences in sulfomucin production in both mice and human. Subsequently, we transplanted organoids established from sulfomucin-enriched region into the injured recipient epithelium following dextran sulfate sodium-induced colitis and analyzed the engrafted epithelium in mouse model.

Results

In human normal colon, sulfomucin production was increased in proximal colon, whereas it was decreased in the inflammatory region of UC. In murine colon epithelium, increased sulfomucin production was found in cecum compared to distal small intestine and proximal colon. RNA sequencing analysis revealed that several key genes associated with sulfomucin production such as Papss2 and Slc26a1 were enriched in isolated murine cecum crypts. Then we established murine cecum organoids and transplanted them into the injured epithelium of distal colon. Although the expression of sulfomucin was temporally decreased in cecum organoids, its secretion was restored again in the engrafted patches after transplantation. Finally, we verified a part of mechanisms controlling sulfomucin production in human samples.

Conclusion

This study illustrated the deliverability of sulfomucin in the disease-relevant grafting model to design sulfomucin-producing epithelial units in severely injured distal colon. The current study is the basis for the better promotion of organoids transplantation therapy for refractory UC.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Molodecky NA, Soon IS, Rabi DM, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142:46-54e42.

    Article  PubMed  Google Scholar 

  2. Ungaro R, Mehandru S, Allen PB, et al. Ulcerative colitis. Lancet. 2017;389:1756–70.

    Article  PubMed  Google Scholar 

  3. Rungoe C, Langholz E, Andersson M, et al. Changes in medical treatment and surgery rates in inflammatory bowel disease: a nationwide cohort study 1979–2011. Gut. 2014;63:1607–16.

    Article  CAS  PubMed  Google Scholar 

  4. Nakase H, Uchino M, Shinzaki S, et al. Evidence-based clinical practice guidelines for inflammatory bowel disease 2020. J Gastroenterol. 2021;56:489–526.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Naganuma M. Solving the questions regarding 5-aminosalitylate formulation in the treatment of ulcerative colitis. J Gastroenterol. 2020;55:1013–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Targownik LE, Singh H, Nugent Z, et al. The epidemiology of colectomy in ulcerative colitis: results from a population-based cohort. Am J Gastroenterol. 2012;107:1228–35.

    Article  PubMed  Google Scholar 

  7. Kakiuchi N, Yoshida K, Uchino M, et al. Frequent mutations that converge on the NFKBIZ pathway in ulcerative colitis. Nature. 2020;577:260–5.

    Article  CAS  PubMed  Google Scholar 

  8. Snippert HJ, van der Flier LG, Sato T, et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell. 2010;143:134–44.

    Article  CAS  PubMed  Google Scholar 

  9. Vermeulen L, Snippert HJ. Stem cell dynamics in homeostasis and cancer of the intestine. Nat Rev Cancer. 2014;14:468–80.

    Article  CAS  PubMed  Google Scholar 

  10. Santos AJM, Lo YH, Mah AT, et al. The intestinal stem cell niche: homeostasis and adaptations. Trends Cell Biol. 2018;28:1062–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Okamoto R, Watanabe M. Role of epithelial cells in the pathogenesis and treatment of inflammatory bowel disease. J Gastroenterol. 2016;51:11–21.

    Article  CAS  PubMed  Google Scholar 

  12. Johansson ME, Larsson JM, Hansson GC. The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of host-microbial interactions. Proc Natl Acad Sci U S A. 2011;108(Suppl 1):4659–65.

    Article  CAS  PubMed  Google Scholar 

  13. Filipe MI. Mucins in the human gastrointestinal epithelium: a review. Invest Cell Pathol. 1979;2:195–216.

    CAS  PubMed  Google Scholar 

  14. Xu P, Xi Y, Zhu J, et al. Intestinal sulfation is essential to protect against colitis and colonic carcinogenesis. Gastroenterology. 2021;161(271–286): e11.

    Google Scholar 

  15. Dawson PA, Huxley S, Gardiner B, et al. Reduced mucin sulfonation and impaired intestinal barrier function in the hyposulfataemic NaS1 null mouse. Gut. 2009;58:910–9.

    Article  CAS  PubMed  Google Scholar 

  16. Tobisawa Y, Imai Y, Fukuda M, et al. Sulfation of colonic mucins by N-acetylglucosamine 6-O-sulfotransferase-2 and Its protective function in experimental colitis in mice. J Biol Chem. 2010;285:6750–60.

    Article  CAS  PubMed  Google Scholar 

  17. Watanabe S, Kobayashi S, Ogasawara N, et al. Transplantation of intestinal organoids into a mouse model of colitis. Nat Protoc. 2022;17:649–71.

    Article  CAS  PubMed  Google Scholar 

  18. Akiyama S, Mochizuki W, Nibe Y, et al. CCN3 expression marks a sulfomucin-nonproducing unique subset of colonic goblet cells in mice. Acta Histochem Cytochem. 2017;50:159–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Matsuo K, Ota H, Akamatsu T, et al. Histochemistry of the surface mucous gel layer of the human colon. Gut. 1997;40:782–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Barker N, van Es JH, Kuipers J, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449:1003–7.

    Article  CAS  PubMed  Google Scholar 

  21. Porter EM, Bevins CL, Ghosh D, et al. The multifaceted Paneth cell. Cell Mol Life Sci. 2002;59:156–70.

    Article  CAS  PubMed  Google Scholar 

  22. Hollingsworth MA, Swanson BJ. Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer. 2004;4:45–60.

    Article  CAS  PubMed  Google Scholar 

  23. Dawson PA, Russell CS, Lee S, et al. Urolithiasis and hepatotoxicity are linked to the anion transporter Sat1 in mice. J Clin Invest. 2010;120:706–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kandori H, Hirayama K, Takeda M, et al. Histochemical, lectin-histochemical and morphometrical characteristics of intestinal goblet cells of germfree and conventional mice. Exp Anim. 1996;45:155–60.

    Article  CAS  PubMed  Google Scholar 

  25. Sato T, Vries RG, Snippert HJ, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–5.

    Article  CAS  PubMed  Google Scholar 

  26. Yui S, Azzolin L, Maimets M, et al. YAP/TAZ-dependent reprogramming of colonic epithelium links ECM remodeling to tissue regeneration. Cell Stem Cell. 2018;22:35-49.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sato T, van Es JH, Snippert HJ, et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011. https://doi.org/10.1038/nature09637.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Sansom OJ, Reed KR, Hayes AJ, et al. Loss of Apc in vivo immediately perturbs Wnt signaling, differentiation, and migration. Genes Dev. 2004;18:1385–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Venkatachalam KV. Human 3′-phosphoadenosine 5′-phosphosulfate (PAPS) synthase: biochemistry, molecular biology and genetic deficiency. IUBMB Life. 2003;55:1–11.

    Article  CAS  PubMed  Google Scholar 

  30. Yui S, Nakamura T, Sato T, et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5(+) stem cell. Nat Med. 2012;18:618–23.

    Article  CAS  PubMed  Google Scholar 

  31. Fukuda M, Mizutani T, Mochizuki W, et al. Small intestinal stem cell identity is maintained with functional Paneth cells in heterotopically grafted epithelium onto the colon. Genes Dev. 2014;28:1752–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Fordham RP, Yui S, Hannan NR, et al. Transplantation of expanded fetal intestinal progenitors contributes to colon regeneration after injury. Cell Stem Cell. 2013;13:734–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all members of the Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University (TMDU) for their feedback on this study. We also thank Shintaro Akiyama, Department of Gastroenterology, Faculty of Medicine, University of Tsukuba for his commentary support on the manuscript. We also thank Ichiro Sekiya, Norio Shimizu, and all other members of Center for Stem cell and Regenerative Medicine, Tokyo Medical and Dental University (TMDU) for their feedback on this study.

Funding

This work was supported by MEXT/JSPS KAKENHI [18K15743 to SY; 20H03657 to SY; 19H01050 and 22H00472 to MW; 19H03634 to RO], Japan Agency for Medical Research and Development (AMED) [20bm0704029h0003 to SY; 20bm0304001h0008 and 20bk0104008h0003 to MW, 20bm0404055h0002 to RO], Japan Science and Technology Agency (JST) Forrest Program [JPMJFR2012 to SY], Young Innovative Medical Science Unit (TMDU) to SY, and Naoki Tsuchida Research Grant to SY.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: SW, MW, SY. Methodology: SW, MW, SY. Investigation: SW, NO, SK, SK, MI, YH, SN, HS, GI, TM. Visualization: SW, NO, SY. Funding acquisition: MW, RO, SY. Sample offer: YN, KT, RO. Supervision: MW. Writing—original draft: SW, NO, SY.

Corresponding author

Correspondence to Shiro Yui.

Ethics declarations

Conflict of interest

All authors declare no COI.

Data availability

RNA sequencing will be available from a corresponding author on reasonable request. Human organoids used in this study can be used only at TMDU according to the regulation of the study.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material (HDR 16 kb)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Watanabe, S., Ogasawara, N., Kobayashi, S. et al. Organoids transplantation as a new modality to design epithelial signature to create a membrane-protective sulfomucin-enriched segment. J Gastroenterol 58, 379–393 (2023). https://doi.org/10.1007/s00535-023-01959-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00535-023-01959-y

Keywords

Navigation