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 「Interferon-γ enhances the therapeutic effect of mesenchymal stem cells on experimental renal fibrosis」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

Interferon-γ enhances the therapeutic effect of mesenchymal stem cells on experimental renal fibrosis

金井 亮 広島大学

2021.02.22

概要

Query Form
41598

Journal:

SREP

[Art. ID: 79664]

Journal: SREP

Author: The following queries have arisen during the editing of your manuscript. Please answer queries by making the
requisite corrections at the appropriate positions in the text.
Query

Details Required

AQ1

Please check your article carefully, coordinate with any co-authors and enter all final
edits clearly in the eproof, remembering to save frequently. Once corrections are
submitted, we cannot routinely make further changes to the article.

AQ2

Note that the eproof should be amended in only one browser window at any one time;
otherwise changes will be overwritten.

AQ3

Author surnames have been highlighted. Please check these carefully and adjust if
the first name or surname is marked up incorrectly. Note that changes here will affect
indexing of your article in public repositories such as PubMed. Also, carefully check
the spelling and numbering of all author names and affiliations, and the corresponding
email address(es).

AQ4

You cannot alter accepted Supplementary Information files except for critical changes
to scientific content. If you do resupply any files, please also provide a brief (but
complete) list of changes. If these are not considered scientific changes, any altered
Supplementary files will not be used, only the originally accepted version will be
published.

AQ5

Please confirm the section headings are correctly identified.

Journal : SREP 41598

Article No : 79664

Author’s Response

Pages : 1

MS Code : 79664

Dispatch : 12-12-2020

www.nature.com/scientificreports

Interferon‑γ enhances
the therapeutic effect
of mesenchymal stem cells
on experimental renal fibrosis

O
F

OPEN

PR
O

Ryo Kanai1, Ayumu Nakashima1,2*, Shigehiro Doi1, Tomoe Kimura1, Ken Yoshida1,
Satoshi Maeda2,3, Naoki Ishiuchi1, Yumi Yamada1, Takeshi Ike1, Toshiki Doi1, Yukio Kato2,3 &
Takao Masaki1*

AQ2

AQ3

AQ4

U

AQ1

N
CO
RR

EC

TE

D

Mesenchymal stem cells (MSCs) administered for therapeutic purposes can be activated by
interferon-γ (IFN-γ) secreted from natural killer cells in injured tissues and exert anti-inflammatory
effects. These processes require a substantial period of time, leading to a delayed onset of MSCs’
therapeutic effects. In this study, we investigated whether pretreatment with IFN-γ could potentiate
the anti-fibrotic ability of MSCs in rats with ischemia–reperfusion injury (IRI) and unilateral ureter
obstruction. Administration of MSCs treated with IFN-γ strongly reduced infiltration of inflammatory
cells and ameliorated interstitial fibrosis compared with control MSCs without IFN-γ treatment.
In addition, conditioned medium obtained from IFN-γ-treated MSCs decreased fibrotic changes in
cultured cells induced by transforming growth factor-β1 more efficiently than that from control MSCs.
Most notably, secretion of prostaglandin E2 from MSCs was significantly increased by treatment with
IFN-γ. Increased prostaglandin E2 in conditioned medium obtained from IFN-γ-treated MSCs induced
polarization of immunosuppressive CD163 and CD206-positive macrophages. In addition, knockdown
of prostaglandin E synthase weakened the anti-fibrotic effects of MSCs treated with IFN-γ in IRI rats,
suggesting the involvement of prostaglandin E2 in the beneficial effects of IFN-γ. Administration of
MSCs treated with IFN-γ might represent a promising therapy to prevent the progression of renal
fibrosis.

The morbidity rate of chronic kidney disease (CKD) is estimated to be 8%–16% w
­ orldwide1. Etiological studies of CKD have reported multiple causes of disease initiation including hypertension, diabetes mellitus, and
­glomerulonephritis2. Despite differences in disease initiation, renal fibrosis exacerbated by persistent inflammation is a histological change common to all these ­etiologies3,4. Currently there are few effective treatments
that prevent the progression of CKD and many patients eventually develop renal failure, which requires renal
replacement therapy, resulting in a heavy social and economic burden. Therefore, there is an urgent need for the
development of novel therapeutic strategies to treat renal fibrosis associated with CKD.
The pathogenesis of CKD is mediated by inflammatory cells, which cause renal fibrosis via fibroblast activation and increased extracellular matrix ­deposition5. Damage-associated molecular patterns (DAMPs) released
from damaged tissues activate the local immune system and several studies reported DAMPs were involved in
promoting renal ­fibrosis6,7. In CKD patients, the inflammatory microenvironment is maintained by infection,
uremic toxins, or tissue i­ schemia9, and this chronic inflammation contributes to the sustained release of DAMPs
from injured kidney tissues, which induces further inflammation and fibrosis. Therefore, the inhibition of inflammation is expected to ameliorate renal fibrosis.
Mesenchymal stem cells (MSCs) isolated from various tissues including bone marrow, blood, and adipose ­tissue10, have multipotency and self-renewal ­ability11,12. They exert their beneficial effects by suppressing

1

Department of Nephrology, Hiroshima University Hospital, 1‑2‑3 Kasumi, Minami‑ku, Hiroshima,
Hiroshima  734‑8551, Japan. 2Department of Stem Cell Biology and Medicine, Graduate School of Biomedical
and Health Sciences, Hiroshima University, 1‑2‑3 Kasumi, Minami‑ku, Hiroshima, Hiroshima  734‑8553,
Japan. 3TWOCELLS Company, Limited, 16‑35 Hijiyama‑honmachi, Minami‑ku, Hiroshima  732‑0816,
Japan. *email: ayumu@hiroshima‑u.ac.jp; masakit@hiroshima‑u.ac.jp
Scientific Reports | _#####################_ | https://doi.org/10.1038/s41598-020-79664-6
Journal : SREP 41598

Article No : 79664

Pages : 14

MS Code : 79664

1
Dispatch : 12-12-2020

Vol.:(0123456789)

www.nature.com/scientificreports/

O
F

inflammation and fibrosis via a paracrine ­mechanism13,14. Several studies have reported that MSCs or extracellular vesicles derived from MSCs have beneficial effects for renal fi
­ brosis15–18.
High mobility group box-1 protein (HMGB1) and interleukin-18 (IL-18) are members of DAMPs— HMGB1
was reported to promote the migration of M
­ SCs19,20, whereas IL-18 contributed to the secretion of interferon-γ
21
(IFN-γ) from natural killer c­ ells . Furthermore, IFN-γ released from immune cells at sites of damaged tissues
stimulated MSCs to secrete anti-inflammatory mediators including prostaglandin E2 (PGE2)22–26. PGE2 has been
reported to induce the polarization of immunosuppressive M2 macrophages that produce anti-inflammatory
cytokines and inhibit the persistence of ­inflammation27,28. In addition, we previously reported that MSCs promoted macrophage differentiation from an M1 pro-inflammatory phenotype to an immunosuppressive M2
phenotype, and that the administration of ex vivo-expanded MSCs suppressed the progression of fi
­ brosis29,30.
Taken together, this suggests that IFN-γ-preconditioned MSCs have a strong immunosuppressive effect and
therefore might ameliorate renal fibrosis. However, the utility of MSCs cultured with IFN-γ for the treatment of
kidney disease has not been investigated.
This study investigated the therapeutic effect of MSCs cultured in IFN-γ-containing medium on inflammation and fibrosis using rat ischemia–reperfusion injury (IRI) and unilateral ureter obstruction (UUO) models.

Results

D

PR
O

Expression of HMGB1, IL‑18, and IFN‑γ in the kidney after the IRI procedure.  HMGB1 and
IL-18, which are known as DAMPs, are released from damaged tissues. Released IL-18, which is also induced
by ­HMGB18, contributes to secretion of IFN-γ19. To confirm expression of HMGB1, IL-18, and IFN-γ induced
by IRI, rats were sacrificed to evaluate their expression in the kidney at 1 day (Post IRI Day 1) and 7 days (Post
IRI Day 7) after the IRI procedure. As shown in a previous s­ tudy31, the protein level of HMGB1 was increased
by the IRI procedure (Fig. 1a). Immunostaining revealed that IL-18 and IFN-γ-positive areas were increased
strongly in the kidney at 1 day after IRI. However, their increase was attenuated at 7 days after IRI (Fig. 1b,c).
These changes are similar to those seen in a previous ­study32. Furthermore, because IL-18 promotes IFN-γ secretion from natural killer cells, IFN-γ expression might be associated with IL-18 secretion. MSCs are activated by
released IFN-γ and exert immunosuppressive ­effects22–26. Therefore, administration of MSCs cultured in IFN-γcontaining medium might have beneficial effects on preventing the progression of renal fibrosis.
IFN‑γ enhances the ability of MSCs to attenuate fibrosis induced by IRI.  To evaluate anti-fibrotic

N
CO
RR

EC

TE

effects of MSCs, we injected PBS, rat MSCs (rMSCs) treated with IFN-γ (IFN-γ rMSCs) or untreated rMSCs
(control rMSCs) into the abdominal aorta of rats after ischemic reperfusion. Twenty-one days later, the rats were
sacrificed, and injured kidneys were collected to evaluate the degree of fibrosis by western blotting. The protein
levels of α-smooth muscle actin (α-SMA) and transforming growth factor-β1 (TGF-β1), markers for drivers of
fibrosis, were increased in the kidney of PBS-injected rats and their levels were suppressed by injection of control
rMSCs (Fig. 2a). Furthermore, injection of IFN-γ rMSCs decreased IRI-induced fibrotic changes more significantly than that of control rMSCs (Fig. 2a). Immunostaining of α-SMA, collagen type I (Col-I), and collagen
type III (Col-III) (extracellular matrix proteins) was also performed to assess renal fibrosis. α-SMA, Col-I, and
Col-III-positive areas were increased in the PBS group. Similar to the results from western blotting, administration of IFN-γ rMSCs reduced α-SMA, Col-I, and Col-III-positive areas more strongly compared with that of
control rMSCs (Fig. 2b,c). These results suggest that IFN-γ-preconditioned rMSCs have a strong anti-fibrotic
effect. ...

この論文で使われている画像

参考文献

1. Jha, V. et al. Chronic kidney disease: global dimension and perspectives. Lancet 382, 260–272 (2013).

2. Xie, Y. et al. Analysis of the global burden of disease study highlights the global, regional, and national trends of chronic kidney

disease epidemiology from 1990 to 2016. Kidney Int. 94, 567–581 (2018).

3. Lee, S. B. & Kalluri, R. Mechanistic connection between inflammation and fibrosis. Kidney Int. 78, S22–S26 (2010).

4. Meng, X. M., Nikolic-Paterson, D. J. & Lan, H. Y. Inflammatory processes in renal fibrosis. Nat. Rev. Nephrol. 10, 493–503 (2014).

5. Zeisberg, M. & Neilson, E. G. Mechanisms of tubulointerstitial fibrosis. J. Am. Soc. Nephrol. 21, 1819–1834 (2010).

6. Rosin, D. L. & Okusa, M. D. Dangers within: DAMP responses to damage and cell death in kidney disease. J. Am. Soc. Nephrol.

22, 416–425 (2011).

7. Anders, H. J. & Schaefer, L. Beyond tissue injury-damage-associated molecular patterns, toll-like receptors, and inflammasomes

also drive regeneration and fibrosis. J. Am. Soc. Nephrol. 25, 1387–1400 (2014).

8. He, Q. et al. HMGB1 promotes the synthesis of pro-IL-1β and pro-IL-18 by activation of p38 MAPK and NF-κB through receptors

for advanced glycation end-products in macrophages. Asian Pac. J. Cancer Prev. 13, 1365–1370 (2012).

9. Zewinger, S., Schumann, T., Fliser, D. & Speer, T. Innate immunity in CKD-associated vascular diseases. Nephrol. Dial. Transplant.

31, 1813–1821 (2016).

10. Pittenger, M. F. et al. Multilineage potential of adult human mesenchymal stem cells. Science 284, 143–147 (1999).

11. Caplan, A. I. & Dennis, J. E. Mesenchymal stem cells as trophic mediators. J. Cell Biochem. 98, 1076–1084 (2006).

Scientific Reports | _#####################_ | https://doi.org/10.1038/s41598-020-79664-6

Vol:.(1234567890)

Journal : SREP 41598

Article No : 79664

Pages : 14

MS Code : 79664

12

Dispatch : 12-12-2020

www.nature.com/scientificreports/

CO

RR

EC

TE

PR

12. Chen, Y., Shao, J. Z., Xiang, L. X., Dong, X. J. & Zhang, G. R. Mesenchymal stem cells: a promising candidate in regenerative

medicine. Int. J. Biochem. Cell Biol. 40, 815–820 (2008).

13. daSilva, M. L., Fontes, A. M., Covas, D. T. & Caplan, A. Mechanisms involved in the therapeutic properties of mesenchymal stem

cells. Cytokine Growth Factor Rev. 20, 419–427 (2009).

14. Hocking, A. M. & Gibran, N. S. Mesenchymal stem cells: paracrine signaling and differentiation during cutaneous wound repair.

Exp Cell Res. 316, 2213–2219 (2011).

15. Gregorini, M. et al. Mesenchymal stromal cells prevent renal fibrosis in a rat model of unilateral ureteral obstruction by suppressing

the renin-angiotensin system via HuR. PLoS ONE 11, e0148542. https​://doi.org/10.1371/journ​al.pone.01485​42 (2016).

16. Matsui, F. et al. Mesenchymal stem cells protect against obstruction-induced renal fibrosis by decreasing STAT3 activation and

STAT3-dependent MMP-9 production. Am. J. Physiol. Renal. Physiol. 312, F25–F32 (2017).

17. Asanuma, H. et al. Arterially delivered mesenchymal stem cells prevent obstruction-induced renal fibrosis. J. Surg. Res. 168, e51–e59

(2011).

18. Grange, C. et al. Stem cell-derived extracellular vesicles inhibit and revert fibrosis progression in a mouse model of diabetic

nephropathy. Sci. Rep. 9, 4468. https​://doi.org/10.1038/s4159​8-019-41100​-9 (2019).

19. Meng, E. et al. High mobility group box 1 protein inhibits the proliferation of human mesenchymal stem cells and promotes their

migration and differentiation along osteoblastic pathway. Stem Cell Dev. 17, 805–813 (2008).

20. Lin, F. et al. Signaling pathways involved in the effects of HMGB1 on mesenchymal stem cell migration and osteoblastic differentiation. Int. J. Mol. Med. 37, 789–797 (2016).

21. Thomas, H. et al. (2014) Interaction with mesenchymal stem cells provokes natural killer cells for enhanced IL-12/IL-18-induced

interferon-gamma secretion. Mediat. Inflamm. https​://doi.org/10.1155/2014/14346​3 (2014).

22. Lin, T. et al. Preconditioning of murine mesenchymal stem cells synergistically enhanced immunomodulation and osteogenesis.

Stem Cell Res Ther. 8, 277. https​://doi.org/10.1186/s1328​7-017-0730-z (2017).

23. English, K. Mechanisms of mesenchymal stromal cell immunomodulation. Immunol. Cell Biol. 91, 19–26 (2013).

24. English, K., Barry, F. P., Field-Corbett, C. P. & Mahon, B. P. IFN-gamma and TNF-alpha differentially regulate immunomodulation

by murine mesenchymal stem cells. Immunol. Lett. 110, 91–100 (2007).

25. Chen, K. et al. Human umbilical cord mesenchymal stem cells hUC-MSCs exert immunosuppressive activities through a PGE2dependent mechanism. Clin. Immunol. 135, 448–458 (2010).

26. Wei, Y. et al. High-efficient generation of VCAM-1+ mesenchymal stem cells with multidimensional superiorities in signatures

and efficacy on aplastic anaemia mice. Cell Prolif. 53, e12862. https​://doi.org/10.1111/cpr.12862​ (2020).

27. Luan, B. et al. CREB pathway links PGE2 signaling with macrophage polarization. Proc. Natl. Acad. Sci. USA 112, 15642–15647

(2015).

28. Jin, L. et al. Mesenchymal stem cells promote type 2 macrophage polarization to ameliorate the myocardial injury caused by diabetic

cardiomyopathy. J. Transl. Med. 17, 251. https​://doi.org/10.1186/s1296​7-019-1999-8 (2019).

29. Yoshida, K. et al. Serum-free medium enhances the immunosuppressive and antifibrotic abilities of mesenchymal stem cells utilized

in experimental renal fibrosis. Stem Cells Transl. Med. 7, 893–905 (2018).

30. Ishiuchi, N. et al. Hypoxia-preconditioned mesenchymal stem cells prevent renal fibrosis and inflammation in ischemia-reperfusion

rats. Stem Cell Res. Ther. 11, 130. https​://doi.org/10.1186/s1328​7-020-01642​-6 (2020).

31. Huiling, W. et al. HMGB1 contributes to kidney ischemia reperfusion injury. J. Am. Soc. Nephrol. 21, 1878–1890 (2010).

32. Sakai, K. et al. Protective effect and mechanism of IL-10 on renal ischemia–reperfusion injury. Lab Invest. 99, 671–683 (2019).

33. Scaffidi, P., Misteli, T. & Bianchi, M. E. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 418,

191–195 (2002).

34. Lotze, M. T. & Tracey, K. J. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat. Rev.

Immunol. 5, 331–342 (2005).

35. Lu, B. et al. Molecular mechanism and therapeutic modulation of high mobility group box 1 release and action: an updated review.

Expert Rev. Clin. Immunol. 10, 713–727 (2014).

36. Phanish, M. K., Wahab, N. A., Colville-Nash, P., Hendry, B. M. & Dockrell, M. E. C. The differential role of Smad2 and Smad3 in

the regulation of pro-fibrotic TGFbeta1 responses in human proximal-tubule epithelial cells. Biochem J. 393, 601–607 (2006).

37. Yoshimura, A., Wakabayashi, Y. & Mori, T. Cellular and molecular basis for the regulation of inflammation by TGF-β. J Biochem.

147, 781–792 (2010).

38. Ueno, T. et al. Mesenchymal stem cells ameliorate experimental peritoneal fibrosis by suppressing inflammation and inhibiting

TGF-β1 signaling. Kidney Int. 84, 297–307 (2013).

39. Nakashima, A. et al. DEC1 modulates the circadian phase of clock gene expression. Mol. Cell Biol. 28, 4080–4092 (2008).

Acknowledgements

We would like to express our gratitude to Dr Mikihito Kajiya of the Department of Periodontal Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University for his insightful advice on the experimental

design. We would like to thank Prof. Takeshi Kawamoto of the Writing Center, Hiroshima University and Mitchell Arico from Edanz Group (www.edanz​editi​ng.com/ac) for editing a draft of this manuscript. We also thank

Ms. Miki Kagiya for technical assistance. This study was supported in part by JSPS KAKENHI Grant Number

JP17K09699. A part of this work was carried out at the Analysis Center of Life Science, Natural Science Center

for Basic Research and Development, Hiroshima University.

Author contributions

A.N. and T.M. designed the study; R.K., A.N., T.K., K.Y., S.M., N.I, Y.Y., T.I., and T.D. carried out experiments;

R.K., S.D., N.I. and Y.K. analyzed the data; R.K. and A.N. drafted and revised the paper. All authors read and

approved the final manuscript.

Competing interests The Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University is a collaborative research laboratory funded by TWOCELLS Company, Limited. Dr. Maeda

is the Deputy Division Manager of R&D Division II, Head Office of Research and Development of TWOCELLS

Company, Limited. Emeritus Prof. Kato is the Vice President of TWOCELLS Company, Limited. Except for the

abovementioned disclosures, all authors have declared that no conflict of interest exists.

Additional information

Supplementary Information The online version contains supplementary material available at https​://doi.

Scientific Reports | _#####################_ | https://doi.org/10.1038/s41598-020-79664-6

Journal : SREP 41598

Article No : 79664

Pages : 14

MS Code : 79664

13

Dispatch : 12-12-2020

Vol.:(0123456789)

www.nature.com/scientificreports/

org/10.1038/s4159​8-020-79664​-6.

Correspondence and requests for materials should be addressed to A.N. or T.M.

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/.

CO

RR

EC

TE

PR

© The Author(s) 2020

Scientific Reports | _#####################_ | https://doi.org/10.1038/s41598-020-79664-6

Vol:.(1234567890)

Journal : SREP 41598

Article No : 79664

Pages : 14

MS Code : 79664

14

Dispatch : 12-12-2020

...

参考文献をもっと見る