リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「CDX2-induced intestinal metaplasia in human gastric organoids derived from induced pluripotent stem cells」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

CDX2-induced intestinal metaplasia in human gastric organoids derived from induced pluripotent stem cells

Koide, Takahiro Koyanagi-Aoi, Michiyo Uehara, Keiichiro Kakeji, Yoshihiro Aoi, Takashi 神戸大学

2022.05.20

概要

Intestinal metaplasia is related to gastric carcinogenesis. Previous studies have suggested the important role of CDX2 in intestinal metaplasia, and several reports have shown that the overexpression of CDX2 in mouse gastric mucosa caused intestinal metaplasia. However, no study has examined the induction of intestinal metaplasia using human gastric mucosa. In the present study, to produce an intestinal metaplasia model in human gastric mucosa in vitro, we differentiated human-induced pluripotent stem cells (hiPSC) to gastric organoids, followed by the overexpression of CDX2 using a tet-on system. The overexpression of CDX2 induced, although not completely, intestinal phenotypes and the enhanced expression of many, but not all, intestinal genes and previously reported intestinal metaplasia-related genes in the gastric organoids. This model can help clarify the mechanisms underlying intestinal metaplasia and carcinogenesis in human gastric mucosa and develop therapies to restitute precursor conditions of gastric cancer to normal mucosa.

この論文で使われている画像

参考文献

Almeida, R., Silva, E., Santos-Silva, F., Silberg,

D.G., Wang, J., De Bolos, C., and David, L. (2003).

Expression of intestine-specific

transcription factors, CDX1 and CDX2, in

intestinal metaplasia and gastric carcinomas.

J. Pathol. 199, 36–40. https://doi.org/10.1002/

path.1246.

Barker, N., Huch, M., Kujala, P., van de Wetering,

M., Snippert, H.J., van Es, J.H., Sato, T., Stange,

D.E., Begthel, H., van den Born, M., et al. (2010).

Lgr5(+ve) stem cells drive self-renewal in the

stomach and build long-lived gastric units in vitro.

Cell Stem Cell 6, 25–36. https://doi.org/10.1016/j.

stem.2009.11.013.

Aoi, T., Yae, K., Nakagawa, M., Ichisaka, T., Okita,

K., Takahashi, K., Chiba, T., and Yamanaka, S.

(2008). Generation of pluripotent stem cells from

adult mouse liver and stomach cells. Science 321,

699–702. https://doi.org/10.1126/science.

1154884.

Beck, F., Chawengsaksophak, K., Waring, P.,

Playford, R.J., and Furness, J.B. (1999).

Reprogramming of intestinal differentiation and

intercalary regeneration in Cdx2 mutant mice.

Proc. Natl. Acad. Sci. U S A 96, 7318–7323. https://

doi.org/10.1073/pnas.96.13.7318.

Chen, H.Y., Hu, Y., Lu, N.H., and Zhu, Y. (2020).

Caudal type homeoboxes as a driving force in

Helicobacter pylori infection-induced gastric

intestinal metaplasia. Gut Microbes 12, 1809331–

1809412. https://doi.org/10.1080/19490976.2020.

1809331.

Chen, H.Y., Hu, Y., Xu, X.B., Zhou, Y.A., Li, N.S.,

He, C., Xie, C., Lu, N.H., and Zhu, Y. (2021).

Upregulation of oncogene Activin A

receptor type I by Helicobacter pylori

infection promotes gastric intestinal

metaplasia via regulating CDX2. Helicobacter 26,

e12849.

iScience 25, 104314, May 20, 2022

11

iScience

ll

Article

OPEN ACCESS

Chiba, T., Seno, H., Marusawa, H., Wakatsuki, Y.,

and Okazaki, K. (2006). Host factors are important

in determining clinical outcomes of Helicobacter

pylori infection. J. Gastroenterol. 41, 1–9. https://

doi.org/10.1007/s00535-005-1743-4.

Correa, P. (1992). Human gastric carcinogenesis:

a multistep and multifactorial process–first

American Cancer Society Award lecture on

cancer epidemiology and prevention. Cancer

Res. 52, 6735–6740.

Correa, P., and Shiao, Y.H. (1994). Phenotypic and

genotypic events in gastric carcinogenesis.

Cancer Res. 54, 1941s–1943s.

Couvelard, A., Cauvin, J.M., Goldfain, D.,

Rotenberg, A., Robaszkiewicz, M., and Flejou, J.F.

(2001). Cytokeratin immunoreactivity of intestinal

metaplasia at normal oesophagogastric junction

indicates its aetiology. Gut 49, 761–766. https://

doi.org/10.1136/gut.49.6.761.

de Vries, A.C., van Grieken, N.C., Looman, C.W.,

Casparie, M.K., de Vries, E., Meijer, G.A., and

Kuipers, E.J. (2008). Gastric cancer risk in patients

with premalignant gastric lesions: a nationwide

cohort study in the Netherlands.

Gastroenterology 134, 945–952. https://doi.org/

10.1053/j.gastro.2008.01.071.

Fagerberg, L., Hallstrom, B.M., Oksvold, P.,

Kampf, C., Djureinovic, D., Odeberg, J., Habuka,

M., Tahmasebpoor, S., Danielsson, A., Edlund, K.,

et al. (2014). Analysis of the human tissue-specific

expression by genome-wide integration of

transcriptomics and antibody-based proteomics.

Mol. Cell Proteomics 13, 397–406. https://doi.

org/10.1074/mcp.m113.035600.

Fordham, R.P., Yui, S., Hannan, N.R.,

Soendergaard, C., Madgwick, A., Schweiger, P.J.,

Nielsen, O.H., Vallier, L., Pedersen, R.A.,

Nakamura, T., et al. (2013). Transplantation of

expanded fetal intestinal progenitors contributes

to colon regeneration after injury. Cell Stem Cell

13, 734–744. https://doi.org/10.1016/j.stem.2013.

09.015.

Fujii, Y., Yoshihashi, K., Suzuki, H., Tsutsumi, S.,

Mutoh, H., Maeda, S., Yamagata, Y., Seto, Y.,

Aburatani, H., and Hatakeyama, M. (2012). CDX1

confers intestinal phenotype on gastric epithelial

cells via induction of stemness-associated

reprogramming factors SALL4 and KLF5. Proc.

Natl. Acad. Sci. U S A 109, 20584–20589. https://

doi.org/10.1073/pnas.1208651109.

Grainger, S., Hryniuk, A., and Lohnes, D. (2013).

Cdx1 and Cdx2 exhibit transcriptional specificity

in the intestine. PLoS One 8, e54757. https://doi.

org/10.1371/journal.pone.0054757.

Guo, R.J., Suh, E.R., and Lynch, J.P. (2004). The

role of Cdx proteins in intestinal development

and cancer. Cancer Biol. Ther. 3, 593–601. https://

doi.org/10.4161/cbt.3.7.913.

Hattori, T. (1986). Development of

adenocarcinomas in the stomach. Cancer 57,

1528–1534. https://doi.org/10.1002/10970142(19860415)57:8<1528::aidcncr2820570815>3.0.co;2-3.

Helal, N., Omran, Z., Aboushousha, T., Youssef,

M., Badawy, A., Abdel Aziz, A., Aboul-Ezz, M.,

and Moussa, M.M. (2020). Predictive value of

CDX2 and SOX2 in chronic gastritis and

intestinal-type gastric cancer. Open Access

12

iScience 25, 104314, May 20, 2022

Maced. J. Med. Sci. 8, 947–955. https://doi.org/

10.3889/oamjms.2020.5570.

Honda, S., Fujioka, T., Tokieda, M., Satoh, R.,

Nishizono, A., and Nasu, M. (1998). Development

of Helicobacter pylori-induced gastric carcinoma

in Mongolian gerbils. Cancer Res. 58, 4255–4259.

Jung, P., Sato, T., Merlos-Suarez, A., Barriga,

F.M., Iglesias, M., Rossell, D., Auer, H., Gallardo,

M., Blasco, M.A., Sancho, E., et al. (2011). Isolation

and in vitro expansion of human colonic stem

cells. Nat. Med. 17, 1225–1227. https://doi.org/

10.1038/nm.2470.

Karlsson, M., Zhang, C., Mear, L., Zhong, W.,

Digre, A., Katona, B., Sjostedt, E., Butler, L.,

Odeberg, J., Dusart, P., et al. (2021). A single-cell

type transcriptomics map of human tissues. Sci.

Adv. 7, eabh2169. https://doi.org/10.1126/

sciadv.abh2169.

Kawachi, H., Takizawa, T., Eishi, Y., Shimizu, S.,

Kumagai, J., Funata, N., and Koike, M. (2003).

Absence of either gastric or intestinal phenotype

in microscopic differentiated gastric carcinomas.

J. Pathol. 199, 436–446. https://doi.org/10.1002/

path.1323.

Kim, S.I., Oceguera-Yanez, F., Sakurai, C.,

Nakagawa, M., Yamanaka, S., and Woltjen, K.

(2016). Inducible transgene expression in human

iPS cells using versatile all-in-one piggyBac

transposons. Methods Mol. Biol. 1357, 111–131.

https://doi.org/10.1007/7651_2015_251.

Kirchner, T., Muller, S., Hattori, T., Mukaisyo, K.,

Papadopoulos, T., Brabletz, T., and Jung, A.

(2001). Metaplasia, intraepithelial neoplasia and

early cancer of the stomach are related to

dedifferentiated epithelial cells defined by

cytokeratin-7 expression in gastritis. Virchows

Arch. 439, 512–522. https://doi.org/10.1007/

s004280100477.

Kohlnhofer, B.M., Thompson, C.A., Walker, E.M.,

and Battle, M.A. (2016). GATA4 regulates

epithelial cell proliferation to control intestinal

growth and development in mice. Cell Mol

Gastroenterol Hepatol 2, 189–209. https://doi.

org/10.1016/j.jcmgh.2015.11.010.

Lee, H.J., Nam, K.T., Park, H.S., Kim, M.A., Lafleur,

B.J., Aburatani, H., Yang, H.K., Kim, W.H., and

Goldenring, J.R. (2010). Gene expression

profiling of metaplastic lineages identifies

CDH17 as a prognostic marker in early stage

gastric cancer. Gastroenterology 139, 213–

225.e3. https://doi.org/10.1053/j.gastro.2010.04.

008.

Li, T., Guo, H., Li, H., Jiang, Y., Zhuang, K., Lei, C.,

Wu, J., Zhou, H., Zhu, R., Zhao, X., et al. (2019).

MicroRNA-92a-1-5p increases CDX2 by targeting

FOXD1 in bile acids-induced gastric intestinal

metaplasia. Gut 68, 1751–1763. https://doi.org/

10.1136/gutjnl-2017-315318.

Lopes, N., Bergsland, C.H., Bjornslett, M.,

Pellinen, T., Svindland, A., Nesbakken, A.,

Almeida, R., Lothe, R.A., David, L., and Bruun, J.

(2020). Digital image analysis of multiplex

fluorescence IHC in colorectal cancer recognizes

the prognostic value of CDX2 and its negative

correlation with SOX2. Lab. Invest. 100, 120–134.

https://doi.org/10.1038/s41374-019-0336-4.

McCracken, K.W., Cata, E.M., Crawford, C.M.,

Sinagoga, K.L., Schumacher, M., Rockich, B.E.,

Tsai, Y.H., Mayhew, C.N., Spence, J.R., Zavros, Y.,

and Wells, J.M. (2014). Modelling human

development and disease in pluripotent stemcell-derived gastric organoids. Nature 516,

400–404. https://doi.org/10.1038/nature13863.

McCracken, K.W., Aihara, E., Martin, B., Crawford,

C.M., Broda, T., Treguier, J., Zhang, X., Shannon,

J.M., Montrose, M.H., and Wells, J.M. (2017).

Wnt/b-catenin promotes gastric fundus

specification in mice and humans. Nature 541,

182–187. https://doi.org/10.1038/nature21021.

Mutoh, H., Hakamata, Y., Sato, K., Eda, A.,

Yanaka, I., Honda, S., Osawa, H., Kaneko, Y., and

Sugano, K. (2002). Conversion of gastric mucosa

to intestinal metaplasia in Cdx2-expressing

transgenic mice. Biochem. Biophys. Res.

Commun. 294, 470–479. https://doi.org/10.1016/

s0006-291x(02)00480-1.

Mutoh, H., Sakurai, S., Satoh, K., Osawa, H.,

Hakamata, Y., Takeuchi, T., and Sugano, K. (2004).

Cdx1 induced intestinal metaplasia in the

transgenic mouse stomach: comparative study

with Cdx2 transgenic mice. Gut 53, 1416–1423.

https://doi.org/10.1136/gut.2003.032482.

Mutoh, H., Hayakawa, H., Sakamoto, H.,

Sashikawa, M., and Sugano, K. (2009). Transgenic

Cdx2 induces endogenous Cdx1 in intestinal

metaplasia of Cdx2-transgenic mouse stomach.

FEBS J. 276, 5821–5831. https://doi.org/10.1111/

j.1742-4658.2009.07263.x.

Mutoh, H., Sashikawa, M., and Sugano, K. (2011).

Sox2 expression is maintained while gastric

phenotype is completely lost in Cdx2-induced

intestinal metaplastic mucosa. Differentiation 81,

92–98. https://doi.org/10.1016/j.diff.2010.10.002.

Nakagawa, M., Taniguchi, Y., Senda, S., Takizawa,

N., Ichisaka, T., Asano, K., Morizane, A., Doi, D.,

Takahashi, J., Nishizawa, M., et al. (2014). A novel

efficient feeder-free culture system for the

derivation of human induced pluripotent stem

cells. Sci. Rep. 4, 3594. https://doi.org/10.1038/

srep03594.

Nature Medicine Editorial (2013). Of men, not

mice. Nat. Med. 19, 379. https://doi.org/10.1038/

nm.3163.

Niu, H., Jia, Y., Li, T., and Su, B. (2017). SOX2

inhibition promotes promoter demethylation of

CDX2 to facilitate gastric intestinal metaplasia.

Dig. Dis. Sci. 62, 124–132. https://doi.org/10.

1007/s10620-016-4361-5.

Okita, K., Matsumura, Y., Sato, Y., Okada, A.,

Morizane, A., Okamoto, S., Hong, H., Nakagawa,

M., Tanabe, K., Tezuka, K.-I., et al. (2011). A more

efficient method to generate integration-free

human iPS cells. Nat. Methods 8, 409–412.

https://doi.org/10.1038/nmeth.1591.

Ormsby, A.H., Goldblum, J.R., Rice, T.W., Richter,

J.E., Falk, G.W., Vaezi, M.F., and Gramlich, T.L.

(1999). Cytokeratin subsets can reliably

distinguish Barrett’s esophagus from intestinal

metaplasia of the stomach. Hum. Pathol. 30,

288–294. https://doi.org/10.1016/s0046-8177(99)

90007-2.

Park, D.Y., Srivastava, A., Kim, G.H., MinoKenudson, M., Deshpande, V., Zukerberg, L.R.,

Song, G.A., and Lauwers, G.Y. (2010). CDX2

expression in the intestinal-type gastric epithelial

neoplasia: frequency and significance. Mod.

iScience

ll

Article

Pathol. 23, 54–61. https://doi.org/10.1038/

modpathol.2009.135.

Plummer, M., Franceschi, S., Vignat, J., Forman,

D., and de Martel, C. (2015). Global burden of

gastric cancer attributable to Helicobacter pylori.

Int. J. Cancer 136, 487–490. https://doi.org/10.

1002/ijc.28999.

Que, J., Okubo, T., Goldenring, J.R., Nam, K.T.,

Kurotani, R., Morrisey, E.E., Taranova, O., Pevny,

L.H., and Hogan, B.L.M. (2007). Multiple dosedependent roles for Sox2 in the patterning and

differentiation of anterior foregut endoderm.

Development 134, 2521–2531. https://doi.org/10.

1242/dev.003855.

Reis, C.A., David, L., Correa, P., Carneiro, F., de

Bolos, C., Garcia, E., Mandel, U., Clausen, H., and

Sobrinho-Simoes, M. (1999). Intestinal metaplasia

of human stomach displays distinct patterns of

mucin (MUC1, MUC2, MUC5AC, and MUC6)

expression. Cancer Res. 59, 1003–1007.

San Roman, A.K., Aronson, B.E., Krasinski, S.D.,

Shivdasani, R.A., and Verzi, M.P. (2015).

Transcription factors GATA4 and HNF4A control

distinct aspects of intestinal homeostasis in

conjunction with transcription factor CDX2. J Biol

Chem 290, 1850–1860. https://doi.org/10.1074/

jbc.M114.620211.

Sato, T., Vries, R.G., Snippert, H.J., van de

Wetering, M., Barker, N., Stange, D.E., van Es,

J.H., Abo, A., Kujala, P., Peters, P.J., and Clevers,

H. (2009). Single Lgr5 stem cells build crypt-villus

structures in vitro without a mesenchymal niche.

Nature 459, 262–265. https://doi.org/10.1038/

nature07935.

Seok, J., Warren, H.S., Cuenca, A.G., Mindrinos,

M.N., Baker, H.V., Xu, W., Richards, D.R.,

McDonald-Smith, G.P., Gao, H., Hennessy, L.,

et al. (2013). Genomic responses in mouse

models poorly mimic human inflammatory

diseases. Proc. Natl. Acad. Sci. U S A 110, 3507–

3512. https://doi.org/10.1073/pnas.1222878110.

OPEN ACCESS

Silberg, D.G., Sullivan, J., Kang, E., Swain, G.P.,

Moffett, J., Sund, N.J., Sackett, S.D., and

Kaestner, K.H. (2002). Cdx2 ectopic expression

induces gastric intestinal metaplasia in transgenic

mice. Gastroenterology 122, 689–696. https://

doi.org/10.1053/gast.2002.31902.

Simmini, S., Bialecka, M., Huch, M., Kester, L., van

de Wetering, M., Sato, T., Beck, F., van

Oudenaarden, A., Clevers, H., and Deschamps, J.

(2014). Transformation of intestinal stem cells into

gastric stem cells on loss of transcription factor

Cdx2. Nat. Commun. 5, 5728. https://doi.org/10.

1038/ncomms6728.

Slack, J.M. (2009). Metaplasia and somatic cell

reprogramming. J. Pathol. 217, 161–168. https://

doi.org/10.1002/path.2442.

Suzuki, K., Koyanagi-Aoi, M., Uehara, K., Hinata,

N., Fujisawa, M., and Aoi, T. (2019). Directed

differentiation of human induced pluripotent

stem cells into mature stratified bladder

urothelium. Sci. Rep. 9, 10506. https://doi.org/10.

1038/s41598-019-46848-8.

Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M.,

Ichisaka, T., Tomoda, K., and Yamanaka, S. (2007).

Induction of pluripotent stem cells from adult

human fibroblasts by defined factors. Cell 131,

861–872. https://doi.org/10.1016/j.cell.2007.11.

019.

Takao, K., and Miyakawa, T. (2015). Genomic

responses in mouse models greatly mimic human

inflammatory diseases. Proc. Natl. Acad. Sci.

U S A 112, 1167–1172. https://doi.org/10.1073/

pnas.1401965111.

Tsukamoto, T., Mizoshita, T., Mihara, M., Tanaka,

H., Takenaka, Y., Yamamura, Y., Nakamura, S.,

Ushijima, T., and Tatematsu, M. (2005). Sox2

expression in human stomach adenocarcinomas

with gastric and gastric-and-intestinal-mixed

phenotypes. Histopathology 46, 649–658. https://

doi.org/10.1111/j.1365-2559.2005.02170.x.

Tsukamoto, T., Mizoshita, T., and Tatematsu, M.

(2006). Gastric-and-intestinal mixed-type

intestinal metaplasia: aberrant expression of

transcription factors and stem cell

intestinalization. Gastric Cancer 9, 156–166.

https://doi.org/10.1007/s10120-006-0375-6.

Uhlen, M., Fagerberg, L., Hallstrom, B.M.,

Lindskog, C., Oksvold, P., Mardinoglu, A.,

Sivertsson, A., Kampf, C., Sjostedt, E., Asplund,

A., et al. (2015). Proteomics. Tissue-based map of

the human proteome. Science 347, 1260419.

https://doi.org/10.1126/science.1260419.

Xu, X., Cheng, J., Luo, S., Huang, D., Xu, J., Qian,

Y., Zhou, H., and Wan, X. (2020). Deoxycholic

acid-stimulated macrophage-derived exosomes

promote intestinal metaplasia and suppress

proliferation in human gastric epithelial cells. Eur.

J. Pharmacol. 887, 173566. https://doi.org/10.

1016/j.ejphar.2020.173566.

Yu, J.H., Zheng, J.B., Qi, J., Yang, K., Wu, Y.H.,

Wang, K., Wang, C.B., and Sun, X.J. (2019). Bile

acids promote gastric intestinal metaplasia by

upregulating CDX2 and MUC2 expression via the

FXR/NF-kB signalling pathway. Int. J. Oncol. 54,

879–892. https://doi.org/10.3892/ijo.2019.4692.

Yuan, T., Ni, Z., Han, C., Min, Y., Sun, N., Liu, C.,

Shi, M., Lu, W., Wang, N., Du, F., et al. (2019).

SOX2 interferes with the function of CDX2 in bile

acid-induced gastric intestinal metaplasia.

Cancer Cell Int. 19, 24. https://doi.org/10.1186/

s12935-019-0739-8.

Zhao, H., Yan, P., Zhang, N., Feng, L., Chu, X., Cui,

G., Qin, Y., Yang, C., Wang, S., and Yang, K.

(2021). The recurrence rate of Helicobacter pylori

in recent 10 years: a systematic review and metaanalysis. Helicobacter 26, e12852.

Zheng, Q., Chen, X.Y., Shi, Y., and Xiao, S.D.

(2004). Development of gastric adenocarcinoma

in Mongolian gerbils after long-term infection

with Helicobacter pylori. J. Gastroenterol.

Hepatol. 19, 1192–1198. https://doi.org/10.1111/

j.1440-1746.2004.03469.x.

iScience 25, 104314, May 20, 2022

13

iScience

ll

Article

OPEN ACCESS

STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE

SOURCE

IDENTIFIER

Antibodies

mouse monoclonal anti-OCT3/4

BD Biosciences

Cat# 611202; RRID: AB_398736

rabbit polyclonal anti-SOX2

Abcam

Cat# ab97959; RRID: AB_2341193

rabbit monoclonal anti-SOX2

Abcam

Cat# ab93689; RRID: AB_10562630

mouse monoclonal anti-CDX2

Thermo Fisher Scientific

Cat# 39-7800; RRID: AB_2533435

mouse monoclonal anti-MUC5AC

Leica Biosystems

Cat# NCL-MUC-5AC; RRID: AB_442113

mouse monoclonal anti-MUC2

Leica Biosystems

Cat# NCL-MUC-2; RRID: AB_442112

rat monoclonal anti-E-cadherin

Takara

Cat# M108; RRID: AB_2895157

goat polyclonal anti-NANOG

R&D Systems

Cat# AF1997; RRID: AB_355097

goat polyclonal anti-SOX17

R&D Systems

Cat# AF1924; RRID: AB_355060

mouse monoclonal anti-a-SMA

Dako

Cat# M0851; RRID: AB_2223500

mouse monoclonal anti-b-III tubulin

Millipore

Cat# MAB1637; RRID: AB_2210524

mouse monoclonal anti-GATA4

Santa Cruz Biotechnology

Cat# sc-25310; RRID: AB_627667

mouse monoclonal anti-E-cadherin (E-cad)

Thermo Fisher Scientific

Cat# 33-4000; RRID: AB_2533118

goat polyclonal anti-E-cad

R&D systems

Cat# AF648; RRID: AB_355504

mouse monoclonal anti- cytokeratin 20 (CK20)

Dako

Cat# IR777; RRID: AB_2133718

mouse monoclonal anti- cytokeratin 7 (CK7)

Dako

Cat# M7018; RRID: AB_2134589

mouse monoclonal anti-CDX2

Biocare Medical

Cat# CM226-C; RRID: AB_2335616

rabbit monoclonal anti-SOX2

Thermo Fisher Scientific

Cat# MA5-16399; RRID: AB_2537918

goat polyclonal anti-PDX1

Abcam

Cat# ab47383 RRID: AB_2162359

rabbit polyclonal anti-Somatostatin

Dako

Cat# A0566; RRID: AB_2688022

mouse monoclonal anti-H, K-ATase

MBL International

Cat# D031-3; RRID: AB_590576

rabbit monoclonal anti-Synaptophysin

Roche

Cat# 760-4595; RRID: AB_2857955

mouse monoclonal anti-Chromogranin A

DAKO

Cat# M0869; RRID: AB_2081135

mouse monoclonal anti-b-actin

Sigma Aldrich

Cat# A5441; RRID: AB_476744

iMatrix-511

Nippi

Cat# 892-021

Stem Fit

Ajinomoto

Chemicals, peptides, and recombinant proteins

penicillin and streptomycin

Life Technologies

Cat# 15140-122

TrypLE Select

Life Technologies

Cat# A12177-01

Rock inhibitor(Y-27632)

WAKO

Cat# 253-00513

Fugene HD

Promega

Cat# E2311

G418

Nacalai Tesque

Cat# 16512-36

Activin A

Peprotech

Cat# 120-14-250UG

BMP4

Peprotech

Cat# 314-BP

B27

Life Technologies

Cat# 17504-001

L-glutamine

Life Technologies

Cat# 25030-081

FBS

BIOWEST

Cat# F7524

CHIR99021

TOCRIS

Cat# 4423

FGF4

Cell Guidance Systems

Cat# GFH31

NOGGIN

R&D Systems

Cat# 6057-NG

(Continued on next page)

14

iScience 25, 104314, May 20, 2022

iScience

ll

Article

OPEN ACCESS

Continued

REAGENT or RESOURCE

SOURCE

HEPES

Invitrogen

Cat# 15630-080

EGF

R&D Systems

Cat# 236-EG

Retinoic acid

WAKO

N2

Life Technologies

IDENTIFIER

Cat# 17502-001

Critical commercial assays

Prime Script II 1st strand cDNA Synthesis Kit

Takara

Cat# 6210B

Takara Ex Taq PCR kit

Takara

Cat# RR001A

TURBO DNA-free kit

Thermo Fisher Scientific

Cat# AM1907

TB Green Premix Ex Taq II

Takara

Cat# RR420A

LR clonase II

Life Technologies

Cat# 11791-020

XT ultraView Universal DAB Detection Kit

Ventana Medical Systems, Inc.

Cat# 760-500

Deposited data

RNA-seq

This paper

GEO: GSE173624

Listing genes highly expressed in the intestine

Fagerberg et al. (2014)

Array Express Archive

(www.ebi.ac.uk/arrayexpress/)

under the accession number E-MTAB-1733

the Human Protein Atlas

Karlsson et al. (2021)

https://www.proteinatlas.org

iPSC line (201B7)

Riken BRC (Tsukuba, Japan)

HPS0001, RRID:CVCL_A324

iPSC line (FF-PB-3AB4)

Suzuki et al. (2019)

iPSC line (CDX2-iPSC)

This paper

Experimental models: Cell lines

Colo320 DM

The Japanese Collection

JCRB0225, RRID:CVCL_0219

of Research Bioresources (JCRB)

Cell bank (Osaka, Japan)

Oligonucleotides

human CDX2 cloning primer, forward primer

This paper

50 -CACCATGTACGTGAGCTACCTCCTGGACAAGGAC-30

human CDX2 cloning primer, reverse primer 50 -TCAC

This paper

TGGGTGACGGTGGGGTTTAGCACCCCCCCAGTTG-30

Primers for hGAPDH, see Table S1

Okita et al. (2011)

Primers for hOCT3/4, see Table S1

Takahashi et al., 2007

Primers for hSOX2, see Table S1

Takahashi et al., 2007

Primers for hNANOG, see Table S1

Takahashi et al., 2007

Primers for hCDX2, see Table S1

This paper

Primers for hPDX1, see Table S1

This paper

Primers for hMUC5AC, see Table S1

This paper

Primers for hCK7, see Table S1

This paper

Primers for hCK20, see Table S1

This paper

Primers for hSOX17, see Table S1

This paper

Primers for hFOXA2, see Table S1

This paper

Recombinant DNA

pENTR/D-TOPO

Life Technologies

PB-TAC-ERN

Kim et al. (2016)

pCAG-PBase

Kim et al. (2016)

provided by Dr. Knut Woltjen

at Kyoto University

provided by Dr. Knut Woltjen

at Kyoto University

(Continued on next page)

iScience 25, 104314, May 20, 2022

15

iScience

ll

Article

OPEN ACCESS

Continued

REAGENT or RESOURCE

SOURCE

IDENTIFIER

Software and algorithms

Strand NGS software program

JSTAT 6.5

Agilent

http://toukeijstat.web.fc2.com/

RESOURCE AVAILABILITY

Lead contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by

the lead contact, Takashi Aoi (takaaoi@med.kobe-u.ac.jp).

Materials availability

This study did not generate new unique reagents.

Data and code availability

RNA-seq data have been deposited at GEO and are publicly available as of the date of publication.

This paper does not report original code.

Any additional information required to reanalyze the data reported in this paper is available from the

lead contact upon reasonable request.

EXPERIMENTAL MODEL AND SUBJECT DETAILS

iPSC culture

The validated iPSC line 201B7 was purchased from Riken Cell bank (Tsukuba, Japan) and transferred from

on-feeder to feeder-free conditions in our laboratory. We cultured the iPSC lines according to a previously

described method (Nakagawa et al., 2014). In brief, the culture plates were precoated with iMatrix-511

(0.5 mg/cm2), and the iPSC were maintained in Stem Fit medium (Ajinomoto) with penicillin (100 units/

mL) and streptomycin (100 mg/mL; Life Technologies, MA, USA) at 37 C with 5% CO2. The medium was

changed every other day and passaged every 7-10 days using 0.53 TrypLE Select (13 TrypLE Select diluted

1:1 with 0.5 mM EDTA/PBS [-]; Life Technologies) and Rho-associated kinase (Rock) inhibitor (Y-27632;

WAKO, Osaka, Japan).

METHOD DETAILS

Vector construction and generation of CDX2-iPSC

The cDNA encoding human CDX2 open reading frame (ORF) was amplified by polymerase chain reaction

(PCR) using the forward primer 50 -CACCATGTACGTGAGCTACCTCCTGGACAAGGAC-30 and reverse

primer 50 -TCACTGGGTGACGGTGGGGTTTAGCACCCCCCCAGTTG-30 , and the resulting PCR product

was cloned into pENTR/D-TOPO (Life Technologies) to generate pENTR-CDX2 clone according to the

manufacturer’s protocol. LR clonase II (Life Technologies) recombination was then performed using a

pENTR-CDX2 clone and the destination vector PB-TAC-ERN (Kim et al., 2016) to generate PB-TACCDX2-ERN.

To generate CDX2-iPSC, the dissociated single cells of FF-PB-3AB4 were seeded onto an iMatrix511coated 6-well plate at a density of 5 3 105 cells/well. The next day, the cells were transfected with

1.5 mg of pCAG-PBase (Kim et al., 2016) and 1.5 mg of PB-TAC-CDX2-ERN using Fugene HD (Promega,

WI, USA). Forty-eight hours after transfection, 100 mg/mL G418 (Nacalai Tesque, Kyoto, Japan) was added

to select transduced cells for 4 days. After 8 hiPSC colonies were isolated and expanded, the subclone

CDX2-iPSC that strongly expressed mCherry by DOX addition (1 mM for 3 days) was selected and used

in this paper. G418 (100 mg/mL) was continuously added during the maintenance culture of CDX2-iPSC

before differentiation into gastric organoids.

16

iScience 25, 104314, May 20, 2022

iScience

ll

Article

OPEN ACCESS

Gastric organoid differentiation

Differentiation into gastric organoids was performed according to a previously reported method

(McCracken et al., 2014). In brief, 2.5 3 105 to 4.0 3 105 iPSC were plated as single cells in a 12-well dish

(Nunc Cell-Culture Treated Multidishes; Thermo Scientific, MA, USA) in StemFit medium with 10 mM of

Rock inhibitor Y-27632 (WAKO). The next day, the medium was changed to RPMI-1640 medium (Nacalai

Tesque) supplemented with 100 ng/mL of Activin A (Peprotech, NJ, USA), 50 ng/mL of BMP4 (Peprotech),

B27 (Life Technologies), L-glutamine (Life Technologies), penicillin and streptomycin. Activin A and B27

and L-glutamine were added for three days, and BMP4 was added on the first day. FBS (BIOWEST, Nuaille´,

France) contained increasing concentrations of 0%, 0.2 and 2.0% to induce differentiation into DE by Day 4.

To generate foregut spheroids, RPMI1640 medium supplemented with 2% FBS, 2 mM of CHIR99021

(TOCRIS, Bristol, UK), 500 ng/mL of FGF4 (Cell Gaidance, MO, USA), 200 ng/mL of NOGGIN (R&D Systems,

MN, USA), penicillin and streptomycin was added for 3 days until Day 7. Retinoic acid (2 mM; WAKO) was

added on Day 6.

For three-dimensional culture of gastric organoids, we used Matrigel (354234; CORNING, NY, USA),

advanced DMEM/F12 medium (Life Technologies), N2 (Life Technologies), B27, L-glutamine, 10 mM of

HEPES (Invitrogen, MA, USA), penicillin/streptomycin and 100 ng/mL of EGF (R&D Systems) at Day 7.

For the first 3 days, 2 mM retinoic acid and 200 ng/mL NOGGIN were added to the media. The media

were replaced every three to four days, as necessary.

A semi-quantitative or real-time quantitative reverse-transcriptase (RT)-PCR analysis

Total RNA was isolated using Trizol (Life Technologies) and treated with the TURBO DNA-free kit (Thermo

Fisher Scientific). For the extraction of RNA from CDX2-iPSC derived DOX(+)/() gastric organoids, Invitrogen Phasemaker Tubes (Thermo Fischer Scientific) were used with Trizol, and DNase treatment was not

performed. The Prime Script II 1st strand cDNA Synthesis Kit (Takara, Shiga, Japan) was used to synthesize

cDNA from 200-500 ng of total RNA.

For semi-quantitative RT-PCR analysis (Figures 1C, 2B, 3D, and S1B), the resulting cDNA was subjected to

PCR with a TaKaRa Ex Taq PCR kit (Takara). For the quantitative PCR analysis (Figures 4C and S2B), we

used a Light Cycler480 Real time PCR system (Roche) with TB Green Premix Ex Taq II (Takara). The

PCR primers used are listed in Table S1.

Western blotting

After 1 mM DOX treatment of CDX2-iPSC for 2 days, the cells were washed once with PBS, lysed with the

M-PER Mammalian Protein Extraction Reagent (Thermo Fisher Scientific) and subjected to SDS-polyacrylamide gel electrophoresis (SDS-PAGE). After the electrophoretic transfer of the proteins to the PVDF membranes, immunoblotting with mouse anti-CDX2 (Invitrogen) and mouse anti-b-actin (Sigma Aldrich, MO,

USA) followed by horseradish peroxidase (HRP)-conjugated secondary antibodies at a 1:3000 dilution in

Can Get Signal immunoreaction enhancer solution (TOYOBO, Osaka, Japan) was performed. The

Amersham Imager 600 imagers (Cytiva, Tokyo, Japan) was used to detect signals.

Immunocytochemistry

Cells were fixed with PBS containing 4% paraformaldehyde for 10 min at room temperature. After washing with

PBS, the cells were treated with 0.3% Triton X-100 in PBS for 45 min at room temperature and blocked with 2%

skim-milk in PBS for 1 h. The cells were incubated with primary antibodies at 4 C overnight and then stained with

secondary antibodies. The primary antibodies were mouse anti-OCT3/4 (611202, dilution 1:200; BD transduction

Laboratories, New Jersey, USA), rabbit anti-SOX2 (ab97959, dilution 1:100; abcam, Cambridge, UK), rabbit antiSOX2 (ab93689, dilution 1:100; abcam), mouse anti-CDX2 (39-7800, dilution 1:100; Invitrogen), mouse anti-MUC5AC (CLH2, dilution 1:100; Leica, Nussloch, Germany), anti-MUC2 mouse monoclonal antibody (Ccp58, dilution 1:200; Leica), rat anti-E-cadherin (M108, dilution 1:500; Takara), goat anti-NANOG (AF1997, dilution

1:200; R&D Systems), goat anti-SOX17 (AF1924, dilution 1:200, R&D Systems), mouse anti-a-SMA (M0851, dilution 1:200; Dako, Glostrup, Denmark), mouse anti-b-III tubulin (MAB1637, dilution 1:200; Millipore, MA, USA) and

anti-GATA4 mouse monoclonal antibody (sc-25310, dilution 1:200; Santa Cruz, TX, USA). All of the secondary

antibodies (Alexa Fluor 594-conjugated anti-mouse or anti-goat IgG, Alexa Fluor 488-conjugated anti-mouse,

anti-goat or anti-rat; or anti-rabbit IgG, Cy5 647-conjugated anti-mouse IgG) were obtained from Life

iScience 25, 104314, May 20, 2022

17

ll

OPEN ACCESS

Technologies. Hoechst 33342 (WAKO) was used for nuclear staining. In Figures 2D, 3B, 3E, 4E, 4F, 6B, S7B and

S11, representative data of two or three independent experiments are shown.

In vitro spontaneous differentiation via embryoid body formation

For embryoid body (EB) formation, undifferentiated iPSC were dissociated into single cells, resuspended in

Primate ES medium (Reprocell) containing 20 mM Rock inhibitor Y-27632 (WAKO) and seeded on low-celladhesion 96-well spindle-bottom plates (PrimeSurface, Sumitomo Bakelite, MS-9096M; Tokyo, Japan) at

1 3 104 cells per well. After 7 days of culture, the EBs were transferred to gelatin-coated 24-well plates

and cultured in the same medium for another 7 days. The differentiated cells were immune-stained with

the indicated antibodies.

Karyotype analyses

The G-band karyotype analysis for FF-PB-3AB4 was performed at Chromocenter, Inc. (Yonago, Japan).

Frozen section samples

The cultured organoids were fixed with 4% paraformaldehyde and then embedded in Tissue-Tek O.C.T

Compound (Sakura Finetek Japan, Tokyo, Japan) and frozen at 80 C. The frozen samples were sectioned

at 5-8 mm on a cryostat.

Histological and immunohistochemical analyses of t ...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る