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 「Pluripotent stem cell model of Shwachman-Diamond syndrome reveals apoptotic predisposition of hemoangiogenic progenitors」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

Pluripotent stem cell model of Shwachman-Diamond syndrome reveals apoptotic predisposition of hemoangiogenic progenitors

Hamabata, Takayuki 京都大学 DOI:10.14989/doctor.r13559

2023.05.23

概要

www.nature.com/scientificreports

OPEN

Pluripotent stem cell model
of Shwachman–Diamond syndrome
reveals apoptotic predisposition
of hemoangiogenic progenitors
Takayuki Hamabata1, Katsutsugu Umeda1*, Kagehiro Kouzuki1, Takayuki Tanaka1,
Tomoo Daifu1, Seishiro Nodomi1, Satoshi Saida1, Itaru Kato  1, Shiro Baba1,
Hidefumi Hiramatsu1, Mitsujiro Osawa2, Akira Niwa2, Megumu K. Saito  2,
Yasuhiko Kamikubo3, Souichi Adachi3, Yoshiko  Hashii4, Akira Shimada5,
Hiroyoshi Watanabe6, Kenji Osafune  7, Keisuke Okita  8, Tatsutoshi Nakahata9,
Kenichiro Watanabe10, Junko Takita1 & Toshio Heike1
Shwachman–Diamond syndrome (SDS), an autosomal recessive disorder characterized by bone
marrow failure, exocrine pancreatic insufficiency, and skeletal abnormalities, is caused by mutations in
the Shwachman–Bodian–Diamond syndrome (SBDS) gene, which plays a role in ribosome biogenesis.
Although the causative genes of congenital disorders frequently involve regulation of embryogenesis,
the role of the SBDS gene in early hematopoiesis remains unclear, primarily due to the lack of a
suitable experimental model for this syndrome. In this study, we established induced pluripotent
stem cells (iPSCs) from patients with SDS (SDS-iPSCs) and analyzed their in vitro hematopoietic and
endothelial differentiation potentials. SDS-iPSCs generated hematopoietic and endothelial cells less
efficiently than iPSCs derived from healthy donors, principally due to the apoptotic predisposition
of ­KDR+CD34+ common hemoangiogenic progenitors. By contrast, forced expression of SBDS gene
in SDS-iPSCs or treatment with a caspase inhibitor reversed the deficiency in hematopoietic and
endothelial development, and decreased apoptosis of their progenitors, mainly via p53-independent
mechanisms. Patient-derived iPSCs exhibited the hematological abnormalities associated with SDS
even at the earliest hematopoietic stages. These findings will enable us to dissect the pathogenesis of
multiple disorders associated with ribosomal dysfunction.
Shwachman–Diamond syndrome (SDS) is a rare autosomal recessive disorder characterized by hematological
abnormalities that manifest as cytopenia and progression to myelodysplastic syndrome and acute myeloid leukemia, exocrine pancreatic insufficiency, and skeletal a­ bnormalities1,2. Hematopoietic stem cell transplantation
is the sole curative treatment to correct the hematological defect of this syndrome, and mortality remains high
due to serious post-transplant c­ omplications3.

1

Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara‑cho, Shogoin, Sakyo‑ku,
Kyoto 606‑8507, Japan. 2Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto
University, Kyoto 606‑8507, Japan. 3Department of Human Health Sciences, Graduate School of Medicine, Kyoto
University, Kyoto 606‑8507, Japan. 4Department of Cancer Immunotherapy, Osaka University School of Medicine,
Suita 565‑0871, Japan. 5Department of Pediatric Hematology/Oncology, Okayama University, Okayama 700‑8558,
Japan. 6Department of Pediatrics, Graduate School of Biomedical Sciences, Tokushima University,
Tokushima  770‑8501, Japan. 7Department of Cell Growth and Differentiation, Center for iPS Cell Research and
Application, Kyoto University, Kyoto 606‑8507, Japan. 8Department of Life Science Frontiers, Center for iPS Cell
Research and Application, Kyoto University, Kyoto  606‑8507, Japan. 9Drug Discovery Technology Development
Office, Center for iPS Cell Research and Application, Kyoto University, Kyoto  606‑8507, Japan. 10Department
of Hematology and Oncology, Shizuoka Children’s Hospital, Shizuoka  420‑8660, Japan. *email: umeume@
kuhp.kyoto‑u.ac.jp
Scientific Reports |

(2020) 10:14859

| https://doi.org/10.1038/s41598-020-71844-8

1
Vol.:(0123456789)

www.nature.com/scientificreports/
Mutations in the Shwachman–Bodian–Diamond (SBDS) gene, which is located on chromosome 7q11, are
present in approximately 90% of patients with SDS; these mutations commonly arise from gene conversion to
the highly similar pseudogene SBDSP4. SBDS mRNA is expressed in a broad range of t­ issues4. Multiple studies
have shown that SBDS protein has a primary function in ribosome a­ ssembly5–7. The additional proposed functions for SBDS, such as mitotic spindle stabilization, chemotaxis, cellular stress responses, and apoptosis, reflect
indirect downstream effects of perturbing ribosome a­ ssembly8–13.
Mutations in genes encoding transcription factors involved in regulating normal development are responsible
for a variety of inherited disorders. During embryogenesis, hematopoietic cells (HCs) and endothelial cells (ECs)
emerge from common hemoangiogenic progenitors that express vascular endothelial growth factor receptor
(VEGFR)-2 (also known as KDR in humans)14–16. Indeed, several HC- and/or EC-related transcriptional factors,
such as SCL and RUNX1, are associated with various congenital hematological d
­ isorders17. Multiple disorders
associated with ribosomal dysfunction (so-called ribosomopathies), including SDS and Diamond–Blackfan
anemia (DBA), also present with hematological ­defects18; however, the pathogenesis of ribosomopathies has
not been fully elucidated. Currently, the field lacks an adequate mouse model of the human disease because
the most analogous mutant in mouse fails to faithfully recapitulate all disease-associated p
­ henotype19–22: Sbds-/embryos fail to generate HCs and ECs due to early lethality prior to embryonic day (E) 6.5 before both lineages
have ­developed23.
Induced pluripotent stem cells (iPSCs) are pluripotent stem cell generated by enforced expression of specific
transcription ­factors24. Patient-specific iPSCs, in combination with directed cell differentiation, are a practical
source of human embryonic progenitors that can surpass the utility of murine models. Accordingly, these cells
have the potential to contribute enormously to patient-oriented research, including disease pathophysiology
and drug ­screening25. In this study, we generated iPSCs from three SDS patients and differentiate them into HCs
and ECs using our established differentiation system for human embryonic stem cells (ESCs) and ­iPSCs26–30.

Results

Generation of iPSCs from SDS patients.  Following transduction of peripheral blood cell derived from

SDS patients with an episomal plasmid vector encoding Oct3/4, Sox2, Klf4, L-Myc, Lin28, and shRNA against
TP53, four clones (SDS1-1 and SDS1-2 from patient 1 and SDS2 from patient 2, and SDS3 from patient 3)
were randomly selected for propagation and further analyses, as previously ­reported29,31. All patient-derived
SDS-iPSCs exhibited a characteristic human ESC-like morphology (Fig. 1a, Supplementary Fig. S1a), and were
capable of propagating in serial passage. DNA sequencing analysis verified an identical mutation in the SBDS
gene in all established SDS-iPSC clones (Fig. 1b, Supplementary Fig. S1b). Chromosomal analysis revealed that
all SDS-iPSC clones maintained a normal karyotype (Fig. 1c, Supplementary Fig. S1c). Expression levels of the
pluripotency markers Oct3/4, Sox2, Klf4, L-Myc, and Lin28 in all SDS-iPSCs were comparable to those in control
iPSCs, although transgene expression was rarely detected (Fig. 1d, Supplementary Fig.S1d). All three primary
germ-layer derivatives were detected in cystic teratomas formed after subcutaneous injection of undifferentiated
iPSCs into immunocompromised NOD/SCID/γcnull mice (Fig. 1e, Supplementary Fig. S1e).
To investigate the pathogenesis of this syndrome, SBDS cDNA and DsRed were transduced into SDS-iPSCs
using the PiggyBac transposon system (Fig. 2a). Western blotting revealed a reduction in SBDS protein expression
in SDS-iPSCs that was rescued in SBDS-overexpressed iPSCs (Fig. 2b). Polysome profiling demonstrated that
ribosomal assembly in SDS-iPSCs was reduced, as evidenced by a decrease in the 80S:40S ratio; this deficiency
was reversed by transduction of SBDS cDNA (Fig. 2c,d).

Impaired granulopoiesis during in  vitro differentiation of SDS‑iPSCs.  First, using a previously
reported in vitro culture s­ ystem26,29, we investigated whether generated SDS-iPSCs recapitulated the hematological phenotype of the syndrome (Fig. 3a). Floating HCs, which mainly consisted of mature neutrophils, first
appeared on day 15 of differentiation of SDS and control iPSCs (Fig. 3b,c, Supplementary Fig. S2a). The remaining HCs consisted of immature myeloid cells and a small number of macrophages. Serial analyses revealed that
floating HCs generated from SDS-iPSCs were less abundant than those from control iPSCs (Fig. 3c). Positivity
for myeloperoxidase and lactoferrin, the constituent proteins of neutrophil-specific granules, was comparable in
neutrophils obtained from SDS-iPSCs and control iPSCs (Supplementary Fig. S2b,c). Similarly, the bactericidal
activity of neutrophils from SDS-iPSCs and control iPSCs did not significantly differ (Supplementary Fig. S2d).
HC production was comparable between SBDS-overexpressing SDS-iPSCs and control iPSCs (Fig. 3c,d, Supplementary Fig.  S1e,f), with no attendant morphological changes (Supplementary Fig.  S2g,h). As reported
­previously9,32, the chemotactic activity of SDS-iPSC–derived neutrophils was severely impaired, and this deficiency was reversed by overexpression of SBDS (Fig. 3e).
We then examined the hematological defects of SDS-iPSCs at the clonogenic progenitor level. In methylcellulose colony-forming assays, SDS-iPSCs formed significantly fewer HC colonies, a defect that was rescued by
SBDS overexpression (Fig. 3f). The decreased size of HC colonies was also reversed by SBDS overexpression,
although the size is not somehow comparable to that of control iPSCs (Fig. 3g). Collectively, these data demonstrated that SDS-iPSCs exhibited reduced HC production, accompanied by impaired neutrophil chemotaxis and
limited colony-forming potential, all of which are typical hematological abnormalities of SDS patients. ...

この論文で使われている画像

参考文献

1. Shwachman, H., Diamond, L., Oski, F. & Khaw, K. T. The syndrome of pancreatic insufficiency and bone marrow dysfunction. J.

Pediatr. 65, 645–663 (1964).

2. Smith, O. P., Hann, I. M., Chessells, J. M., Reeves, B. R. & Milla, P. Haematological abnormalities in Shwachmann-Diamond

syndrome. Br. J. Haematol. 94, 279–284 (1996).

3. Cesaro, R. et al. Haematopoietic stem cell transplantation for Shwachman-Diamond disease: a study from the European Group

for blood and marrow transplantation. Br. J. Haematol. 131, 231–236 (2005).

4. Boocock, G. R. B. et al. Mutations in SBDS are associated with Shwachman-Diamond syndrome. Nat. Genet. 33, 97–101 (2003).

5. Menne, T. F. et al. The Shwachman-Boidan-Diamond syndrome protein mediates translational activation of ribosomes in yeast.

Nat. Genet. 39, 486–495. https​://doi.org/10.1038/ng199​4 (2007).

6. Finch, A. J. et al. Uncoupling of GTP hydrolysis from elF6 release on the ribosome causes Shwachman-Diamond syndrome. Genes

Dev. 25, 917–929. https​://doi.org/10.1101/gad.62301​1 (2011).

7. Weis, F. et al. Mechanism of elF6 release from the nascent 60S ribosomal subunit. Nat. Struct. Mol. Biol. 22, 914–919. https​://doi.

org/10.1038/nsmb.3112 (2015).

8. Wessels, D. et al. The Shwachman-Bodian-Diamond syndrome gene encodes an RNA-binding protein that localizes to the pseudopod of Dictyostelium amoebae during chemotaxis. J. Cell Sci. 119, 370–379 (2006).

9. Austin, K. M. et al. Mitotic spindle destabilization and genomic instability in Shwachman-Diamond syndrome. J. Clin. Invest. 118,

1511–1518. https​://doi.org/10.1172/JCI33​764 (2008).

10. Rujkijyanont, P. et al. SBDS-deficient cells undergo accelerated apoptosis through the Fas-pathway. Haematologica 93, 363–371.

https​://doi.org/10.3324/haema​tol.11579​ (2008).

11. Watanabe, K. et al. SBDS-deficiency results in specific hypersensitivity to Fas stimulation and accumulation of Fas at the plasma

membrane. Apoptosis 14, 77–89. https​://doi.org/10.1007/s1049​5-008-0275-9 (2009).

12. Ball, J. L. et al. Shwachman-Bodian Diamond syndrome is a multi-functional protein implicated in cellular stress responses. Hum.

Mol. Genet. 18, 3684–3695. https​://doi.org/10.1093/hmg/ddp31​6 (2009).

13. Warren, A. J. Molecular basis of the human ribosomopathy Shwachman-Diamond syndrome. Adv. Biol. Regul. 67, 109–127. https​

://doi.org/10.1016/j.jbior​.2017.09.002 (2018).

14. Shalaby, F. et al. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 376, 62–66 (1995).

15. Shalaby, F. et al. A requirement for Flk1 in primitive and definitive hematopoiesis and vasculogenesis. Cell 89, 981–990 (1997).

16. Nishikawa, S. I., Nishikawa, S., Hirashima, M., Matsuyoshi, N. & Kodama, H. Progressive lineage analysis by cell sorting and culture

identifies FLK1+ VE-cadherin+ cells at diverging point of endothelial and hematopoietic lineages. Development 125, 1747–1757

(1998).

17. Wlison, N. K., Calero-Nieto, F. J., Ferreira, R. & Göttgens, B. Transcriptional regulation of haematopoietic transcription factors.

Stem Cell Res. Ther. 2, 6. https​://doi.org/10.1186/scrt4​7 (2011).

Scientific Reports |

Vol:.(1234567890)

(2020) 10:14859 |

https://doi.org/10.1038/s41598-020-71844-8

10

www.nature.com/scientificreports/

18. Narla, A. & Ebert, B. L. Ribosomopathies: human disorders of ribosome dysfunction. Blood 115, 3196–3205. https​://doi.

org/10.1182/blood​-2009-10-17812​9 (2010).

19. Tourlakis, M. E. et al. Deficiency of Sbds in the mouse pancreas leads to features of Shwachman-Diamond syndrome, with loss of

zymogen granules. Gastroenterology 143, 481–492. https​://doi.org/10.1053/j.gastr​o.2012.04.012 (2012).

20. Tourlakis, M. E. et al. In vivo senescence in the Sbds-deficient murine pancreas: cell-type specific consequences of translation in

sufficiency. PLoS Genet. 11, e1005288. https​://doi.org/10.1371/journ​al.pgen.10052​88 (2015).

21. Zambetti, N. A. et al. Deficiency of the ribosome biogenesis gene Sbds in hematopoietic stem and progenitor cells causes neutropenia in mice by attenuating lineage progression in myelocytes. Haematologica 100, 1285–1293. https​://doi.org/10.3324/haema​

tol.2015.13157​3 (2015).

22. Zambetti, N. A. et al. Mesenchymal inflammation drives genotoxic stress in hematopoietic stem cells and predicts disease evolution

in human pre-leukemia. Cell Stem Cell 19, 613–627. https​://doi.org/10.1016/j.stem.2016.08.021 (2016).

23. Zhang, S., Shi, M., Hui, C. C. & Rommens, J. M. Loss of the mouse ortholog of the Shwachman-diamond syndrome gene (Sbds)

results in early embryonic lethality. Mol. Cell Biol. 26, 6656–6663 (2006).

24. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

25. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917–1920 (2007).

26. Niwa, A. et al. A novel serum-free monolayer culture for orderly hematopoietic differentiation of human pluripotent cells via

mesodermal progenitors. PLoS ONE 6, e22261. https​://doi.org/10.1371/journ​al.pone.00222​61 (2011).

27. Morishima, T. et al. Neutrophil differentiation from human-induced pluripotent stem cells. J Cell Physiol 226, 1283–1291. https​

://doi.org/10.1002/jcp.22456​(2011).

28. Tanaka, T. et al. Induced pluripotent stem cells from CINCA syndrome patients as a model for dissecting somatic mosaicism and

drug discovery. Blood 120, 1299–1308. https​://doi.org/10.1182/blood​-2012-03-41788​1 (2012).

29. Morishima, T. et al. Genetic correction of HAX1 in induced pluripotent stem cells from a patient with severe congenital neutropenia

improves defective granulopoiesis. Haematologica 99, 19–27. https​://doi.org/10.3324/haema​tol.2013.08387​3 (2014).

30. Suzuki, N. M. et al. Pluripotent cell models of fanconi anemia identify the early pathological defect in human hemoangiogenic

progenitors. Stem Cells Trans. Med. 4, 333–338. https​://doi.org/10.5966/sctm.2013-0172 (2015).

31. Okita, K. et al. An efficient nonviral method to generated integration-free human-induced pluripotent stem cells from cord blood

and peripheral blood cells. Stem Cells 31, 458–466. https​://doi.org/10.1002/stem.1293 (2013).

32. Orelio, C. & Kuijpers, T. W. Shwachman-Diamond syndrome neutrophils have altered chemoattractant-induced F-actin polymerization and polarization characteristics. Haematologica 94, 409–413. https​://doi.org/10.3324/haema​tol.13733​ (2009).

33. Dror, Y. & Freedman, M. H. Shwachman-Diamond syndrome marrow cells show abnormally increased apoptosis mediated through

the Fas pathway. Blood 97, 3011–3301 (2001).

34. Sieff, C. A. et al. Pathogenesis of the erythroid failure in Diamond Blackfan anaemia. Br. J. Haematol. 148, 611–622. https​://doi.

org/10.1111/j.1365-2141.2009.07993​.x (2010).

35. Tulpule, A. et al. Pluripotent stem cell models of Shwachman-Diamond syndrome reveal a common mechanism for pancreatic

and hematopoietic dysfunction. Cell Stem Cell 12, 727–736. https​://doi.org/10.1016/j.stem.2013.04.002 (2013).

36. Donati, G., Montanaro, L. & Drerenzini, M. Ribosome biogenesis and control of cell proliferation: p53 is not alone. Cancer Res 72,

1602–1607. https​://doi.org/10.1158/0008-5472.CAN-11-3992 (2009).

37. Shimamura, A. & Alter, B. P. Pathophysiology and management of inherited bone marrow failure syndromes. Blood Rev. 24,

101–122. https​://doi.org/10.1016/j.blre.2010.03.002 (2010).

38. Austin, K. M., Leary, R. J. & Shimamura, A. The Shwachman-Diamond SBDS protein localizes to the nucleolus. Blood 106,

1253–1258 (2005).

39. Zhang, Y. et al. Control of hematopoietic stem cell emergence by antagonistic functions of ribosomal protein paralogs. Dev. Cell

24, 411–425. https​://doi.org/10.1016/j.devce​l.2013.01.018 (2013).

40. Wan, Y. et al. Transcriptome analysis reveal a ribosome constitutes disorder involved in the RPL5 downregulated zebrafish model

of Diamond-Blackfan anemia. BMC Med. Genomics 9, 13. https​://doi.org/10.1186/s1292​0-016-0174-9 (2016).

41. Garçon, L. et al. Ribosomal and hematopoietic defects in induced pluripotent stem cells derived from Diamond Blackfan anemia

patients. Blood 122, 912–921. https​://doi.org/10.1182/blood​-2013-01-47832​1 (2013).

42. Mccann, K. L. & Baserga, S. J. Genetics. Mysterious ribosomopathies. Science 341, 849–850. https​://doi.org/10.1126/scien​ce.12441​

56 (2013).

43. Shammas, C. et al. Structural and mutational analysis of the SBDS protein family. J. Biol. Chem. 280, 19221–19229 (2005).

44. Umeda, K. et al. Identification and characterization of hemoangiogenic progenitors during cynomolgus monkey embryonic stem

cell differentiation. Stem Cells 24, 1348–1358 (2006).

45. Choi, K. D. et al. Hematopoietic and endothelial differentiation of human induced pluripotent stem cells. Stem Cells 27, 559–567.

https​://doi.org/10.1634/stemc​ells.2008-0922 (2009).

Acknowledgments

We thank Ms. Tomomi Sudo, Center for iPS Cell Research and Application, Kyoto University, for her excellent

technical assistance, and the Center for Anatomical Studies, Kyoto University Graduate School of Medicine, for

immunocytochemical analysis.

Author contributions

T.H. performed experiments; M.O., K.O., K.O., and K.W. established iPS cell lines; T.T., S.N., S.S., S.B., A.N.,

and M.K.S. assisted with teratoma formation analysis; K.K., T.D., I.K., Y.K., Y.H., A.S., and H.W. assisted with

lentiviral transduction of iPS cells; T.H., K.U., and K.W. analyzed and interpreted data; H.H., S.A., T.N., J.T., and

T.H. assisted with interpretation of data and provided insightful comments; T.H. prepared figures; T.H., K.U.,

and K.W. wrote the manuscript; T.H., K.U., K.O., K.W., and T.H. designed the research.

Funding

This study is supported by a Grant-in-Aid for Scientific Research (C) (15K09651).

Competing interests The authors declare no competing interests.

Additional information

Supplementary information is available for this paper at https​://doi.org/10.1038/s4159​8-020-71844​-8.

Correspondence and requests for materials should be addressed to K.U.

Scientific Reports |

(2020) 10:14859 |

https://doi.org/10.1038/s41598-020-71844-8

11

Vol.:(0123456789)

www.nature.com/scientificreports/

Reprints and permissions information is available at 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 licence, and indicate if changes were made. The images or other third party material in this

article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the

material. If material is not included in the article’s Creative Commons licence 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 licence, visit http://creat​iveco​mmons​.org/licen​ses/by/4.0/.

© The Author(s) 2020

Scientific Reports |

Vol:.(1234567890)

(2020) 10:14859 |

https://doi.org/10.1038/s41598-020-71844-8

12

...

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