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 「Engraftment of allogeneic iPS cell-derived cartilage organoid in a primate model of articular cartilage defect」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

Engraftment of allogeneic iPS cell-derived cartilage organoid in a primate model of articular cartilage defect

Abe, Kengo 大阪大学

2023.02.20

概要

Title

Engraftment of allogeneic iPS cell-derived
cartilage organoid in a primate model of
articular cartilage defect

Author(s)

Abe, Kengo; Yamashita, Akihiro; Morioka, Miho et
al.

Citation

Nature Communications. 2023, 14(1), p. 804

Version Type VoR
URL
rights

https://hdl.handle.net/11094/93295
This article is licensed under a Creative
Commons Attribution 4.0 International License.

Note

Osaka University Knowledge Archive : OUKA
https://ir.library.osaka-u.ac.jp/
Osaka University

Article

https://doi.org/10.1038/s41467-023-36408-0

Engraftment of allogeneic iPS cell-derived
cartilage organoid in a primate model
of articular cartilage defect
Received: 30 May 2022

Check for updates

1234567890():,;

1234567890():,;

Accepted: 31 January 2023

Kengo Abe1,2,3, Akihiro Yamashita1,3, Miho Morioka1, Nanao Horike1,3,
Yoshiaki Takei3,4, Saeko Koyamatsu1, Keisuke Okita 5, Shuichi Matsuda
Noriyuki Tsumaki 1,3,6

2

&

Induced pluripotent stem cells (iPSCs) are a promising resource for allogeneic
cartilage transplantation to treat articular cartilage defects that do not heal
spontaneously and often progress to debilitating conditions, such as
osteoarthritis. However, to the best of our knowledge, allogeneic cartilage
transplantation into primate models has never been assessed. Here, we show
that allogeneic iPSC-derived cartilage organoids survive and integrate as well
as are remodeled as articular cartilage in a primate model of chondral defects
in the knee joints. Histological analysis revealed that allogeneic iPSC-derived
cartilage organoids in chondral defects elicited no immune reaction and
directly contributed to tissue repair for at least four months. iPSC-derived
cartilage organoids integrated with the host native articular cartilage and
prevented degeneration of the surrounding cartilage. Single-cell RNAsequence analysis indicated that iPSC-derived cartilage organoids differentiated after transplantation, acquiring expression of PRG4 crucial for joint
lubrication. Pathway analysis suggested the involvement of SIK3 inactivation.
Our study outcomes suggest that allogeneic transplantation of iPSC-derived
cartilage organoids may be clinically applicable for the treatment of patients
with chondral defects of the articular cartilage; however further assessment of
functional recovery long term after load bearing injuries is required.

Articular cartilage covers the ends of bones and provides lubrication,
which is vital for smooth joint movement and shock absorption.
Articular cartilage is avascular and consists of chondrocytes embedded in the extracellular matrix (ECM), which enables the mechanical
functions necessary for joint motion and shock absorption. Cartilage
ECM consists of collagen fibrils composed of type II, IX, and XI collagen
molecules and proteoglycans composed of aggrecan, link protein, and
glycosaminoglycans.

As articular cartilage has a limited capacity for repair, and so far,
no drugs are available for cartilage repair, focal damage or erosion of
articular cartilage frequently leads to debilitating conditions, such as
osteoarthritis. Although cell-based therapies have been proposed, only
a limited number of autologous chondrocytes are generated since
expansion culture steers the chondrocyte character toward that of
fibroblastic cells1, as evidenced in autologous chondrocyte implantation (ACI), wherein more than 90% of the repaired tissue is

1

Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan. 2Department of Orthopaedic
Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan. 3Department of Clinical Application, Center for iPS Cell Research and Application,
Kyoto University, Kyoto, Japan. 4Regenerative Medicine Technology Department, Healthcare R&D Center, Asahi Kasei Corporation, Kyoto, Japan. 5Department
of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan. 6Premium Research Institute for Human Metaverse
e-mail: ntsumaki@tsu.med.osaka-u.ac.jp
Medicine (WPI-PRIMe), Osaka University, Osaka, Japan.

Nature Communications | (2023)14:804

1

Article
fibrocartilaginous2. Allogeneic cartilage has been transplanted clinically without matching human leukocyte antigen (HLA) types and
without the use of immunosuppressive drugs3–5. However, whether the
transplanted allogeneic cartilage causes an immune reaction remains
controversial. Some reports suggest low immunogenicity of
chondrocytes6,7, whereas others show that chondrocytes are antigenic
and elicit varying degrees of immune reactions8,9. To the best of our
knowledge, the regenerative mechanisms following allogeneic cartilage transplantation have not yet been reported. Whether transplanted
cartilage achieves engraftment (survives and constitutes repaired tissue directly) or only transiently remains and secretes growth factors to
stimulate recipient progenitor cells has not been analyzed.
Induced pluripotent stem (iPS) cells are a promising source for the
regenerative treatment of articular cartilage damage10,11. Cartilage
consisting of chondrocytes and ECM has been successfully created
from iPS cells by differentiating them into chondrocytes, which are
subsequently transferred into a three-dimensional culture to make iPS
cell-derived chondrocytes produce and accumulate ECM around
themselves to form cartilaginous tissue particles12,13. Owing to the selfrenewal activity of iPS cells, allogeneic iPS cell-derived cartilage organoids can theoretically be produced inexhaustibly and transplanted
into an unlimited number of patients, solving the issues associated
with allogenic cartilage, such as scarcity of donors, risk of disease
transmission, and variations in cartilage qualities between donors.
In this study, we analyzed the allogenic transplantation of major
histocompatibility complex (MHC)-mismatched iPS cell-derived cartilage organoids in a primate animal model without the use of immunosuppressive drugs. We differentiated cynomolgus monkey iPS cells
(cyiPSCs) into chondrocytes to create cyiPSC-derived cartilage organoids (cyiPS-Cart). We then transplanted cyiPS-Cart into chondral
defects on the knee joint surface of cynomolgus monkeys in an allogeneic manner. Single-cell RNA-sequencing (scRNA-seq) and molecular analysis of the cyiPS-Cart graft revealed molecular pathways
involved in cell differentiation that remodeled the cyiPS-Cart toward
articular cartilage after transplantation.

Results
Preparation of cyiPS cell-derived cartilage organoid (cyiPS-Cart)
1466A1 cyiPSCs expressing enhanced green fluorescent protein (EGFP)
under a constitutive promoter were used. Cartilage was created from
cyiPSCs14 and human iPSCs12,13 using modified protocols. Briefly,
chondrocytes were induced from cyiPSCs in a chondrogenic medium
for two weeks and transferred to a three-dimensional culture, where
they produced and accumulated ECM to form cartilaginous particles
(Supplementary Fig. 1a). The cyiPSC-derived cartilage organoid (cyiPSCart) particles were 1–3 mm in diameter (Supplementary Fig. 1b).
Histological analysis showed that the particles consisted of cells, and
the ECM was stained positively with safranin O. Immunohistochemical
analysis revealed that the ECM contained type II collagen (Supplementary Fig. 1c). Type I collagen was not detectable except at the
periphery of a particle.

Allogeneic transplantation of cyiPS-Cart in primate chondral
defect model
Cartilage defects can be classified into two categories based on their
depth: chondral defects extending down to but not through the subchondral bone, and osteochondral defects extending down through
the subchondral bone (Fig. 1a). Chondral defects are the most common
in patients with articular cartilage damage or erosion, including during
the early stages of osteoarthritis.
We created chondral defects in the femoral trochlear ridge of the
right knee joints of 12 cynomolgus monkeys and transplanted cyiPSCart (transplantation group) in six monkeys or nothing (empty group)
in the remaining monkeys (Fig. 1b). MHC typing revealed a mismatch
between cyiPSCs and recipient monkeys (Supplementary Table 1).

Nature Communications | (2023)14:804

https://doi.org/10.1038/s41467-023-36408-0

Computed tomography (CT) imaging analysis of the knee joints indicated that bone structures were normal immediately after surgery and
at 4 and 12 weeks after surgery (Supplementary Fig. 2), suggesting that
the defects were chondral and did not extend through the subchondral
bone throughout the experiment.
Three monkeys from each group were sacrificed 4 and 17 weeks
after surgery (n = 3). The gross appearance of the joint surface indicated that chondral defects in the empty group were filled with brown
tissue at 4 and 17 weeks (Fig. 1c). On the other hand, chondral defects in
the transplantation group were filled with transparent tissue at 4 weeks
(Fig. 1c) which later turned white as articular cartilage, making it difficult to distinguish the area where cyiPS-Cart were transplanted from
the surrounding articular cartilage area at 17 weeks after transplantation (Fig. 1c). In each monkey, one transplant site was harvested and
subjected to histological analysis and other two sites were combined
and used for scRNA-seq analysis.

Allogeneic transplantation of cyiPS-Cart did not elicit an
immune reaction in primate chondral defects
We recently reported that allogeneic cyiPS-Cart elicited an immune
reaction when transplanted into osteochondral defects14. However, no
scientific evidence exists on whether allogeneic cartilage elicits an
immune reaction when implanted in chondral defects. To answer this
question, we analyzed histological sections of the transplanted sites.
For control, we created osteochondral defects in additional three
monkeys, transplanted cyiPS-Cart, and sacrificed them 4 weeks later. ...

この論文で使われている画像

参考文献

1.

Huey, D. J., Hu, J. C. & Athanasiou, K. A. Unlike bone, cartilage

regeneration remains elusive. Science 338, 917–921 (2012).

2. Roberts, S., Menage, J., Sandell, L. J., Evans, E. H. & Richardson, J. B.

Immunohistochemical study of collagen types I and II and procollagen IIA in human cartilage repair tissue following autologous

chondrocyte implantation. Knee 16, 398–404 (2009).

3. Wang, T. et al. Patellofemoral cartilage lesions treated with particulated juvenile allograft cartilage: a prospective study with minimum 2-year clinical and magnetic resonance imaging outcomes.

Arthroscopy 34, 1498–1505 (2018).

4. Farr, J., Tabet, S. K., Margerrison, E. & Cole, B. J. Clinical, radiographic, and histological outcomes after cartilage repair with particulated juvenile articular cartilage: a 2-year prospective study.

Am. J. Sports Med. 42, 1417–1425 (2014).

5. Dawkins, B. J. et al. Patellofemoral joint cartilage restoration with

particulated juvenile allograft in patients under 21 years old. Knee

https://doi.org/10.1016/j.knee.2021.07.006 (2021).

6. Adkisson, H. D. et al. Immune evasion by neocartilage-derived

chondrocytes: Implications for biologic repair of joint articular

cartilage. Stem Cell Res. 4, 57–68 (2010).

7. Kimura, T., Yamashita, A., Ozono, K. & Tsumaki, N. Limited immunogenicity of human induced pluripotent stem cell-derived cartilages. Tissue Eng. Part A 22, 1367–1375 (2016).

8. Malejczyk, J., Osiecka, A., Hyc, A. & Moskalewski, S. Effect of

immunosuppression on rejection of cartilage formed by transplanted allogeneic rib chondrocytes in mice. Clin. Orthop. Relat.

Res. 269, 266–273 (1991).

9. Romaniuk, A. et al. Rejection of cartilage formed by transplanted

allogeneic chondrocytes: evaluation with monoclonal antibodies.

Transpl. Immunol. 3, 251–257 (1995).

10. Yamashita, A. & Tsumaki, N. Recent progress of animal transplantation studies for treating articular cartilage damage using pluripotent stem cells. Dev. Growth Differ. 63, 72–81 (2021).

11. Castro-Vinuelas, R. et al. Induced pluripotent stem cells for cartilage repair: current status and future perspectives. Eur. Cell Mater.

36, 96–109 (2018).

12. Yamashita, A., Yoshitomi, H., Kihara, S., Toguchida, J. & Tsumaki, N.

Culture substrate-associated YAP inactivation underlies chondrogenic differentiation of human induced pluripotent stem cells.

Stem Cells Transl. Med. https://doi.org/10.1002/sctm.200058 (2020).

13. Yamashita, A. et al. Generation of scaffoldless hyaline cartilaginous

tissue from human iPSCs. Stem Cell Rep. 4, 404–418 (2015).

14

Article

14. Okutani, Y. et al. Generation of monkey induced pluripotent stem

cell-derived cartilage lacking major histocompatibility complex

class I molecules on the cell surface. Tissue Eng. Part A 28,

94–106 (2022).

15. Stuart, T. et al. Comprehensive integration of single-cell data. Cell

177, 1888–1902.e1821 (2019).

16. Ji, Q. et al. Single-cell RNA-seq analysis reveals the progression of

human osteoarthritis. Ann. Rheum. Dis. 78, 100–110 (2019).

17. Swann, D. A., Silver, F. H., Slayter, H. S., Stafford, W. & Shore, E. The

molecular structure and lubricating activity of lubricin isolated from

bovine and human synovial fluids. Biochem. J. 225, 195–201 (1985).

18. Rhee, D. K. et al. The secreted glycoprotein lubricin protects cartilage surfaces and inhibits synovial cell overgrowth. J. Clin. Invest.

115, 622–631 (2005).

19. Ruan, M. Z. et al. Proteoglycan 4 expression protects against the

development of osteoarthritis. Sci. Transl. Med. 5, 176ra134 (2013).

20. Zhang, C. H. et al. Creb5 establishes the competence for Prg4

expression in articular cartilage. Commun. Biol. 4, 332 (2021).

21. Flannery, C. R. et al. Articular cartilage superficial zone protein (SZP)

is homologous to megakaryocyte stimulating factor precursor and

Is a multifunctional proteoglycan with potential growth-promoting,

cytoprotective, and lubricating properties in cartilage metabolism.

Biochem. Biophys. Res. Commun. 254, 535–541 (1999).

22. Ogawa, H., Kozhemyakina, E., Hung, H. H., Grodzinsky, A. J. & Lassar, A. B. Mechanical motion promotes expression of Prg4 in

articular cartilage via multiple CREB-dependent, fluid flow shear

stress-induced signaling pathways. Genes Dev. 28, 127–139 (2014).

23. Altarejos, J. Y. & Montminy, M. CREB and the CRTC co-activators:

sensors for hormonal and metabolic signals. Nat. Rev. Mol. Cell Biol.

12, 141–151 (2011).

24. Yahara, Y. et al. Pterosin B prevents chondrocyte hypertrophy and

osteoarthritis in mice by inhibiting Sik3. Nat. Commun. 7,

10959 (2016).

25. Sasagawa, S. et al. SIK3 is essential for chondrocyte hypertrophy

during skeletal development in mice. Development 139,

1153–1163 (2012).

26. Csukasi, F. et al. The PTH/PTHrP-SIK3 pathway affects skeletogenesis through altered mTOR signaling. Sci. Transl. Med. https://doi.

org/10.1126/scitranslmed.aat9356 (2018).

27. Chesterman, P. J. & Smith, A. U. Homotransplantation of articular

cartilage and isolated chondrocytes. An experimental study in

rabbits. J. Bone Jt. Surg. Br. 50, 184–197 (1968).

28. Ao, Y. et al. The use of particulated juvenile allograft cartilage for

the repair of porcine articular cartilage defects. Am. J. Sports Med.

47, 2308–2315 (2019).

29. Caplan, A. I. Adult mesenchymal stem cells: when, where, and how.

Stem Cells Int. 2015, 628767 (2015).

30. Caplan, A. I. M. S. Cs The sentinel and safe-guards of injury. J. Cell.

Physiol. 231, 1413–1416 (2016).

31. Ansboro, S., Roelofs, A. J. & De Bari, C. Mesenchymal stem cells for

the management of rheumatoid arthritis: immune modulation,

repair or both? Curr. Opin. Rheumatol. 29, 201–207 (2017).

32. Petrigliano, F. A. et al. Long-term repair of porcine articular cartilage

using cryopreservable, clinically compatible human embryonic

stem cell-derived chondrocytes. NPJ Regen. Med. 6, 77 (2021).

33. Kimura, T. et al. Proposal of patient-specific growth plate cartilage

xenograft model for FGFR3 chondrodysplasia. Osteoarthr. Cartil.

26, 1551–1561 (2018).

34. Tognana, E. et al. Adjacent tissues (cartilage, bone) affect the

functional integration of engineered calf cartilage in vitro.

Osteoarthr. Cartil. 13, 129–138 (2005).

35. Lee, M. C., Sung, K. L., Kurtis, M. S., Akeson, W. H. & Sah, R. L.

Adhesive force of chondrocytes to cartilage. Effects of chondroitinase ABC. Clin. Orthop. Relat. Res. https://doi.org/10.1097/

00003086-200001000-00029 (2000).

Nature Communications | (2023)14:804

https://doi.org/10.1038/s41467-023-36408-0

36. Chen, X. et al. Integration capacity of human induced pluripotent stem cell-derived cartilage. Tissue Eng. Part A 25,

437–445 (2019).

37. Shiina, T. et al. Discovery of novel MHC-class I alleles and haplotypes in Filipino cynomolgus macaques (Macaca fascicularis) by

pyrosequencing and Sanger sequencing: Mafa-class I polymorphism. Immunogenetics 67, 563–578 (2015).

38. Yamashita, A., Takada, T., Yamamoto, G. & Torii, R. Stable maintenance of monkey embryonic stem cells in the absence of bFGF.

Transpl. Proc. 38, 1614–1615 (2006).

39. Morizane, A. et al. MHC matching improves engraftment of iPSCderived neurons in non-human primates. Nat. Commun. 8,

385 (2017).

40. Wang, Y. et al. Endogenous regeneration of critical-size chondral

defects in immunocompromised rat xiphoid cartilage using decellularized human bone matrix scaffolds. Tissue Eng. Part A 18,

2332–2342 (2012).

41. Wakitani, S. et al. Mesenchymal cell-based repair of large, fullthickness defects of articular cartilage. J. Bone Jt. Surg. Am. 76,

579–592 (1994).

42. Sugimoto, M. et al. Universal surface biotinylation: a simple, versatile and cost-effective sample multiplexing method for single-cell

RNA-seq analysis. DNA Res. https://doi.org/10.1093/dnares/

dsac017 (2022).

43. Shichino, S. et al. TAS-Seq is a robust and sensitive amplification

method for bead-based scRNA-seq. Commun. Biol. 5, 602 (2022).

44. Yates, A. D. et al. Ensembl 2020. Nucleic Acids Res. 48,

D682–D688 (2020).

45. Lun, A. T. L. et al. EmptyDrops: distinguishing cells from empty

droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 20, 63 (2019).

46. Finak, G., Manuel. -Perez, J. & Gottardo, R. flowTrans: parameter

optimization for flow cytometry data transformation. R package

version 1.36.0. (2019).

47. Scrucca, L., Fop, M., Murphy, T. B. & Raftery, A. E. mclust 5: clustering, classification and density estimation using Gaussian finite

mixture models. R. J. 8, 289–317 (2016).

48. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell

184, 3573–3587.e3529 (2021).

49. The Tabula Muris Consortium, Single-cell transcriptomics of 20

mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

50. Kamatani, T. et al. Human iPS cell-derived cartilaginous tissue

spatially and functionally replaces nucleus pulposus. Biomaterials

284, 121491 (2022).

51. Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graphbased genome alignment and genotyping with HISAT2 and HISATgenotype. Nat. Biotechnol. 37, 907–915 (2019).

52. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general

purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

53. La Manno, G. et al. RNA velocity of single cells. Nature 560,

494–498 (2018).

54. Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing

RNA velocity to transient cell states through dynamical modeling.

Nat. Biotechnol. 38, 1408–1414 (2020).

55. Wolf, F. A. et al. PAGA: graph abstraction reconciles clustering with

trajectory inference through a topology preserving map of single

cells. Genome Biol. 20, 59 (2019).

56. Iwai, T., Murai, J., Yoshikawa, H. & Tsumaki, N. Smad7 inhibits

chondrocyte differentiation at multiple steps during endochondral

bone formation and down-regulates p38 MAPK pathways. J. Biol.

Chem. 283, 27154–27164 (2008).

57. Gosset, M., Berenbaum, F., Thirion, S. & Jacques, C. Primary culture

and phenotyping of murine chondrocytes. Nat. Protoc. 3,

1253–1260 (2008).

15

Article

https://doi.org/10.1038/s41467-023-36408-0

Acknowledgements

Additional information

We thank Chieko Matsuda, Masumi Sanada, Hiroki Hagizawa, and Yuya

Nishijima for their assistance and helpful discussion. We thank the iPS

Cell Research Fund for its research support. This study was supported by

JSPS KAKENHI Grant No. 18H02923 (to N.T.) and WPI Premium Research

Institute for Human Metaverse Medicine (PRIMe) (to N.T.) from the Japan

Society for the Promotion of Science. This study was also supported by

the Center for Clinical Application Research on Specific Disease/

Organ (type B) Grant No. 21bm0304004h0009 (to N.T.); Research

Project for Practical Applications of Regenerative Medicine Grant No.

21bk0104079h0003 (to N.T.); Practical Research Project for Rare/

Intractable Diseases (step 1) Grant No. 21ek0109452h0002 (to N.T.);

Core Center for iPS Cell Research Grant No. 20bm0104001h0008 (to

N.T.); and the Acceleration Program for Intractable Diseases Research

utilizing disease-specific iPS cells Grant No. 20bm0804006h0004

(to N.T.) from the Japan Agency for Medical Research and Development (AMED).

Supplementary information The online version contains

supplementary material available at

https://doi.org/10.1038/s41467-023-36408-0.

Author contributions

K.A., S.M., and N.T. designed experiments. K.O. prepared cyiPSCs. A.Y.

created the cyiPS-cart. K.A. transplanted cyiPS-Carts into monkeys. K.A.

performed CT and histological analyses. K.A., M.M., S.K., and N.T. performed scRNA-seq analysis. N.H. and Y.T. performed experiments

regarding Sik3. K.A. and N.T. wrote the manuscript.

Competing interests

N.T. is an inventor and Kyoto University is a holder of the patent on

“An efficient chondrocyte induction method” (PCT/JP2014/079117).

This patent is licensed to Asahi KASEI corporation. Y.T. is an

employee of Asahi KASEI. The remaining authors declare no

competing interests.

Nature Communications | (2023)14:804

Correspondence and requests for materials should be addressed to

Noriyuki Tsumaki.

Peer review information Nature Communications thanks Denis

Evseenko and the other, anonymous, reviewer(s) for their contribution to

the peer review of this work. Peer reviewer reports are available.

Reprints and permissions information is available at

http://www.nature.com/reprints

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

Open Access This article is licensed under a Creative Commons

Attribution 4.0 International License, which permits use, sharing,

adaptation, distribution and reproduction in any medium or format, as

long as you give appropriate credit to the original author(s) and the

source, provide a link to the Creative Commons license, and indicate if

changes were made. The images or other third party material in this

article are included in the article’s Creative Commons license, unless

indicated otherwise in a credit line to the material. If material is not

included in the article’s Creative Commons license and your intended

use is not permitted by statutory regulation or exceeds the permitted

use, you will need to obtain permission directly from the copyright

holder. To view a copy of this license, visit http://creativecommons.org/

licenses/by/4.0/.

© The Author(s) 2023

16

...

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