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 「Human bone marrow-derived mesenchymal stromal cells cultured in serum-free media demonstrate enhanced antifibrotic abilities via prolonged survival and robust regulatory T cell induction in murine bleomycin-induced pulmonary 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

Human bone marrow-derived mesenchymal stromal cells cultured in serum-free media demonstrate enhanced antifibrotic abilities via prolonged survival and robust regulatory T cell induction in murine bleomycin-induced pulmonary fibrosis

髙尾 俊 広島大学

2022.03.23

概要

(2021) 12:506
Takao et al. Stem Cell Res Ther
https://doi.org/10.1186/s13287-021-02574-5

Open Access

RESEARCH

Human bone marrow‑derived mesenchymal
stromal cells cultured in serum‑free media
demonstrate enhanced antifibrotic abilities
via prolonged survival and robust regulatory
T cell induction in murine bleomycin‑induced
pulmonary fibrosis
Shun Takao1, Taku Nakashima1*  , Takeshi Masuda1, Masashi Namba2, Shinjiro Sakamoto1,
Kakuhiro Yamaguchi1, Yasushi Horimasu1, Shintaro Miyamoto1, Hiroshi Iwamoto1, Kazunori Fujitaka1,
Hironobu Hamada3, Shinya Takahashi4, Ayumu Nakashima5 and Noboru Hattori1 

Abstract 
Background:  Mesenchymal stromal cells (MSCs) are a potential therapeutic tool for pulmonary fibrosis. However,
ex vivo MSC expansion using serum poses risks of harmful immune responses or unknown pathogen infections in the
recipients. Therefore, MSCs cultured in serum-free media (SF-MSCs) are ideal for clinical settings; however, their efficacy in pulmonary fibrosis is unknown. Here, we investigated the effects of SF-MSCs on bleomycin-induced pulmonary inflammation and fibrosis compared to those of MSCs cultured in serum-containing media (S-MSCs).
Methods:  SF-MSCs and S-MSCs were characterized in vitro using RNA sequence analysis. The in vivo kinetics and
efficacy of SF-MSC therapy were investigated using a murine model of bleomycin-induced pulmonary fibrosis. For
normally distributed data, Student’s t test and one-way repeated measures analysis of variance followed by post hoc
Tukey’s test were used for comparison between two groups and multiple groups, respectively. For non-normally distributed data, Kruskal–Wallis and Mann–Whitney U tests were used for comparison between groups, using e Bonferroni’s correction for multiple comparisons. All tests were two-sided, and P < 0.05 was considered statistically significant.
Results:  Serum-free media promoted human bone marrow-derived MSC expansion and improved lung engraftment
of intravenously administered MSCs in recipient mice. SF-MSCs inhibited the reduction in serum transforming growth
factor-β1 and the increase of interleukin-6 in both the serum and the bronchoalveolar lavage fluid during bleomycininduced pulmonary fibrosis. SF-MSC administration increased the numbers of regulatory T cells (Tregs) in the blood
and lungs more strongly than in S-MSC administration. Furthermore, SF-MSCs demonstrated enhanced antifibrotic
effects on bleomycin-induced pulmonary fibrosis, which were diminished by antibody-mediated Treg depletion.

*Correspondence: tnaka@hiroshima-u.ac.jp
1
Department of Molecular and Internal Medicine, Graduate School
of Biomedical and Health Sciences, Hiroshima University, 1‑2‑3, Kasumi,
Minami‑ku, Hiroshima 734‑8551, Japan
Full list of author information is available at the end of the article
© The Author(s) 2021. 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 Creative Commons Public Domain Dedication waiver (http://​creat​iveco​
mmons.​org/​publi​cdoma​in/​zero/1.​0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Takao et al. Stem Cell Res Ther

(2021) 12:506

Page 2 of 16

Conclusions:  SF-MSCs significantly suppressed BLM-induced pulmonary inflammation and fibrosis through
enhanced induction of Tregs into the lungs and corrected the dysregulated cytokine balance. Therefore, SF-MSCs
could be a useful tool for preventing pulmonary fibrosis progression without the demerits of serum use.
Keywords:  Mesenchymal stromal cells, Serum-free, Bleomycin, Pulmonary fibrosis, Regulatory T cells, Interleukin-6,
Transforming growth factor-β

Background
Idiopathic pulmonary fibrosis (IPF) is a severe pulmonary fibrotic disease that presents with short life expectancy and a high mortality rate [1]. Since patients with
IPF who are treated with antifibrotic agents show inhibition of forced vital capacity (FVC) decline and improved
survival, antifibrotic agents such as pirfenidone and nintedanib have been conditionally recommended for IPF
treatment [2]. However, these agents cannot halt disease
progression; moreover, they have adverse effects, including gastrointestinal disorders, skin-related problems,
and liver damage [3]. Thus, considering the lack of more
effective options for treating IPF, further therapeutic
approaches are being explored.
Mesenchymal stromal cells (MSCs) are pluripotent
cells in the bone marrow and are now known to be isolated from various sources, including adipose tissue,
umbilical cord, peripheral blood, and muscle tissue [4].
Systemically administered MSCs home to the site of
injury and exert anti-inflammatory effects by modulating various immune cells. Furthermore, MSCs secrete
cytokines and growth factors with proliferative and
angiogenic effects and support tissue repair through
paracrine effects [5]. With the expectation of applying
these favorable effects to therapy, several preclinical and
clinical studies are ongoing using human MSCs to treat
chronic diseases including autoimmune, inflammatory,
degenerative, and cardiovascular diseases [6]. Regarding
lung diseases, MSC-based therapies have been reported
as effective in preventing experimental models of pulmonary fibrosis [7]. Based on successful studies with animal
models, clinical trials of MSC-based therapies for interstitial lung diseases, mainly human IPF, are underway
worldwide [8]. Some clinical trials have shown that MSCbased cell therapy provides a protective effect against
FVC decline over time in patients with IPF [9, 10]. In
addition, no serious adverse events related to MSC-based
cell therapy have been reported in these trials, suggesting the safety and tolerance of this therapy [8, 10]. These
findings suggest that MSC-based cell therapy could be a
new potential therapeutic option for treating IPF.
In vitro expansion of MSCs is necessary before their
transplantation into recipients of MSC-based therapies.
MSC growth in  vitro generally requires culture media
supplemented with fetal bovine serum (FBS) or human

serum to provide the factors essential for cell growth. In
fact, most studies on MSCs have used serum-containing media [11]. However, using serum in culture media
poses various potential disadvantages including pathogen contamination (e.g., unknown viruses, mycoplasma,
and prions), harmful immunizing effects [11, 12], inhibition of cell growth [13], uneven quality between lots,
global shortage of supply, and high costs [14]. Therefore,
defined culture conditions without sera are ideal as a tool
for MSC therapy in humans. Recently, several chemically
defined serum-free media for experimental MSC cultures
have been commercialized [11, 14]. Wu et  al. showed
that human MSCs cultured in serum-free medium (SFMSCs) exhibited strong immunomodulatory activity and
secreted higher levels of immunoregulatory factors compared with MSCs cultured in serum-containing medium
(S-MSCs) in  vitro, and showing improved therapeutic
activity in a rat model of pulmonary arterial hypertension
in  vivo [15]. Moreover, other animal studies have also
reported better therapeutic efficacy of SF-MSCs compared with that of S-MSCs in a mouse model of acute
colitis [16], a rat model of renal fibrosis [17] and peritoneal fibrosis [18]. Thus, using SF-MSCs not only avoids
the disadvantages of using sera, but SF-MSCs may also
be more useful as a therapeutic tool than S-MSCs. Previous preclinical studies of SF-MSCs used for lung disease
reported that xenogeneic SF-MSCs demonstrated therapeutic effects in a rat model of Escherichia coli-induced
[19] and ventilator-induced lung injury [20]. However,
the therapeutic effects of SF-MSCs on pulmonary fibrosis
have not been investigated. Considering the progress of
IPF clinical trials with MSCs, clarifying the efficacy of SFMSCs in experimental pulmonary fibrosis is essential for
developing MSC-based therapies in human IPF. Therefore, in the current study, we investigated the effects of
SF-MSCs on an experimental mouse model of lung fibrosis and compared these effects with those of S-MSCs.

Methods
MSCs

Human bone marrow-derived MSCs were collected
from the sternum of consenting patients during thoracic
surgery with the approval of the Ethics Committee of
Hiroshima University Hospital (E-1089). MSCs were cultured in Dulbecco’s modified Eagle’s medium (DMEM)

Takao et al. Stem Cell Res Ther

(2021) 12:506

(Sigma-Aldrich, St. Louis, MO, USA) supplemented with
10% FBS (Sigma-Aldrich) for S-MSCs, or with serumfree STK2 medium (KBDSTC102; DS Pharma Biomedical) for SF-MSCs. The MSCs were dissociated with
Accutase (Innovative Cell Tech, San Diego, CA, USA)
and passaged 5–6 times before use. For the proliferation
assay, MSCs were seeded into 24-well plates at a density
of 5 × ­103 cells/well and cultured as S-MSCs or SF-MSCs.
After staining with Trypan blue (Sigma, St. Louis, MO,
USA), the cell number per well was counted at 24, 48,
72, 96, and 120 h after the start of the cell culture using an
automated cell counter (TC-20, Bio-Rad, Hercules, CA,
USA). Cell morphologies were also recorded at the indicated time points using a Nikon Diaphot 300 microscope
(Nikon, Tokyo, Japan). ...

この論文で使われている画像

参考文献

1. Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, et al. An

official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis:

evidence-based guidelines for diagnosis and management. Am J Respir

Crit Care Med. 2011;183(6):788–824.

2. Raghu G, Rochwerg B, Zhang Y, Garcia CA, Azuma A, Behr J, et al. An official ATS/ERS/JRS/ALAT clinical practice guideline: treatment of idiopathic

pulmonary fibrosis an update of the 2011 clinical practice guideline. Am J

Respirat Crit Care Med. 2015;192(2):e3-19.

3. Rogliani P, Calzetta L, Cavalli F, Matera MG, Cazzola M. Pirfenidone, nintedanib and N-acetylcysteine for the treatment of idiopathic pulmonary

fibrosis: a systematic review and meta-analysis. Pulm Pharmacol Ther.

2016;40:95–103.

4. Hass R, Kasper C, Böhm S, Jacobs R. Different populations and sources

of human mesenchymal stem cells (MSC): a comparison of adult and

neonatal tissue-derived MSC. Cell Commun Signal. 2011;9:12.

5. Lou S, Duan Y, Nie H, Cui X, Du J, Yao Y. Mesenchymal stem cells:

biological characteristics and application in disease therapy. Biochimie.

2021;185:9–21.

6. Ullah I, Subbarao RB, Rho GJ. Human mesenchymal stem cells—current

trends and future prospective. Biosci Rep. 2015;35(2):e00191.

7. Behnke J, Kremer S, Shahzad T, Chao CM, Böttcher-Friebertshäuser E,

Morty RE, et al. MSC based therapies-new perspectives for the injured

lung. J Clin Med. 2020;9(3):682.

Page 15 of 16

8. Lu Q, El-Hashash AHK. Cell-based therapy for idiopathic pulmonary

fibrosis. Stem cell investigation. 2019;6:22.

9. Fishman JE, Kim GJ, Kyeong NY, Goldin JG, Glassberg MK. Intravenous stem cell dose and changes in quantitative lung fibrosis and

DLCO in the AETHER trial: a pilot study. Eur Rev Med Pharmacol Sci.

2019;23(17):7568–72.

10. Averyanov A, Koroleva I, Konoplyannikov M, Revkova V, Lesnyak V, Kalsin

V, et al. First-in-human high-cumulative-dose stem cell therapy in idiopathic pulmonary fibrosis with rapid lung function decline. Stem Cells

Transl Med. 2020;9(1):6–16.

11. Karnieli O, Friedner OM, Allickson JG, Zhang N, Jung S, Fiorentini D, et al. A

consensus introduction to serum replacements and serum-free media for

cellular therapies. Cytotherapy. 2017;19(2):155–69.

12. Shih DT, Burnouf T. Preparation, quality criteria, and properties of human

blood platelet lysate supplements for ex vivo stem cell expansion. New

Biotechnol. 2015;32(1):199–211.

13. Shahdadfar A, Frønsdal K, Haug T, Reinholt FP, Brinchmann JE. In vitro

expansion of human mesenchymal stem cells: choice of serum is a

determinant of cell proliferation, differentiation, gene expression, and

transcriptome stability. Stem cells (Dayton, Ohio). 2005;23(9):1357–66.

14. Gottipamula S, Muttigi MS, Kolkundkar U, Seetharam RN. Serum-free

media for the production of human mesenchymal stromal cells: a review.

Cell Prolif. 2013;46(6):608–27.

15. Wu M, Han ZB, Liu JF, Wang YW, Zhang JZ, Li CT, et al. Serum-free media

and the immunoregulatory properties of mesenchymal stem cells in vivo

and in vitro. Cell Physiol Biochem. 2014;33(3):569–80.

16. Wu X, Wu D, Mu Y, Zhao Y, Ma Z. Serum-free medium enhances the therapeutic effects of umbilical cord mesenchymal stromal cells on a murine

model for acute colitis. Front Bioeng Biotechnol. 2020;8:586.

17. Yoshida K, Nakashima A, Doi S, Ueno T, Okubo T, Kawano KI, et al. Serumfree medium enhances the immunosuppressive and antifibrotic abilities

of mesenchymal stem cells utilized in experimental renal fibrosis. Stem

Cells Transl Med. 2018;7(12):893–905.

18. Nagasaki K, Nakashima A, Tamura R, Ishiuchi N, Honda K, Ueno T, et al.

Mesenchymal stem cells cultured in serum-free medium ameliorate

experimental peritoneal fibrosis. Stem Cell Res Ther. 2021;12(1):203.

19. Curley GF, Jerkic M, Dixon S, Hogan G, Masterson C, O’Toole D, et al.

Cryopreserved, xeno-free human umbilical cord mesenchymal stromal

cells reduce lung injury severity and bacterial burden in rodent Escherichia coli-induced acute respiratory distress syndrome. Crit Care Med.

2017;45(2):e202–12.

20. Horie S, Gaynard S, Murphy M, Barry F, Scully M, O’Toole D, et al. Cytokine

pre-activation of cryopreserved xenogeneic-free human mesenchymal

stromal cells enhances resolution and repair following ventilator-induced

lung injury potentially via a KGF-dependent mechanism. Intensive Care

Med Exp. 2020;8(1):8.

21. Egger C, Cannet C, Gerard C, Jarman E, Jarai G, Feige A, et al. Administration of bleomycin via the oropharyngeal aspiration route leads to

sustained lung fibrosis in mice and rats as quantified by UTE-MRI and

histology. PLoS ONE. 2013;8(5):e63432.

22. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause

D, et al. Minimal criteria for defining multipotent mesenchymal stromal

cells. The international society for cellular therapy position statement.

Cytotherapy. 2006;8(4):315–7.

23. Wang L, Wang Y, Yang T, Guo Y, Sun T. Angiotensin-converting enzyme

2 attenuates bleomycin-induced lung fibrosis in mice. Cell Physiol Biochem. 2015;36(2):697–711.

24. Hsu HS, Liu CC, Lin JH, Hsu TW, Hsu JW, Su K, et al. Involvement of ER

stress, PI3K/AKT activation, and lung fibroblast proliferation in bleomycininduced pulmonary fibrosis. Sci Rep. 2017;7(1):14272.

25. Goodwin J, Choi H, Hsieh MH, Neugent ML, Ahn JM, Hayenga HN, et al.

Targeting hypoxia-inducible factor-1α/pyruvate dehydrogenase kinase

1 axis by dichloroacetate suppresses bleomycin-induced pulmonary

fibrosis. Am J Respir Cell Mol Biol. 2018;58(2):216–31.

26. Chen L, Tredget EE, Liu C, Wu Y. Analysis of allogenicity of mesenchymal stem cells in engraftment and wound healing in mice. PLoS ONE.

2009;4(9):e7119.

27. Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: immune evasive,

not immune privileged. Nat Biotechnol. 2014;32(3):252–60.

Takao et al. Stem Cell Res Ther

(2021) 12:506

28. Kabat M, Bobkov I, Kumar S, Grumet M. Trends in mesenchymal stem cell

clinical trials 2004–2018: is efficacy optimal in a narrow dose range? Stem

Cells Transl Med. 2020;9(1):17–27.

29. Muroi K, Miyamura K, Okada M, Yamashita T, Murata M, Ishikawa T, et al.

Bone marrow-derived mesenchymal stem cells (JR-031) for steroid-refractory grade III or IV acute graft-versus-host disease: a phase II/III study. Int J

Hematol. 2016;103(2):243–50.

30. Choudhery MS, Khan M, Mahmood R, Mehmood A, Khan SN, Riazuddin

S. Bone marrow derived mesenchymal stem cells from aged mice have

reduced wound healing, angiogenesis, proliferation and anti-apoptosis

capabilities. Cell Biol Int. 2012;36(8):747–53.

31. Cheng RJ, Xiong AJ, Li YH, Pan SY, Zhang QP, Zhao Y, et al. Mesenchymal

stem cells: allogeneic MSC may be immunosuppressive but autologous

MSC are dysfunctional in lupus patients. Front Cell Dev Biol. 2019;7:285.

32. Silva JD, Paredes BD, Araújo IM, Lopes-Pacheco M, Oliveira MV, Suhett GD,

et al. Effects of bone marrow-derived mononuclear cells from healthy

or acute respiratory distress syndrome donors on recipient lung-injured

mice. Crit Care Med. 2014;42(7):e510–24.

33. Akiyama K, Chen C, Wang D, Xu X, Qu C, Yamaza T, et al. Mesenchymalstem-cell-induced immunoregulation involves FAS-ligand-/FAS-mediated

T cell apoptosis. Cell Stem Cell. 2012;10(5):544–55.

34. Wang D, Huang S, Yuan X, Liang J, Xu R, Yao G, et al. The regulation of the

Treg/Th17 balance by mesenchymal stem cells in human systemic lupus

erythematosus. Cell Mol Immunol. 2017;14(5):423–31.

35. Li J, Tan J, Martino MM, Lui KO. Regulatory T-cells: potential regulator of

tissue repair and regeneration. Front Immunol. 2018;9:585.

36. Boveda-Ruiz D, D’Alessandro-Gabazza CN, Toda M, Takagi T, Naito M,

Matsushima Y, et al. Differential role of regulatory T cells in early and late

stages of pulmonary fibrosis. Immunobiology. 2013;218(2):245–54.

37. Kamio K, Azuma A, Matsuda K, Usuki J, Inomata M, Morinaga A, et al.

Resolution of bleomycin-induced murine pulmonary fibrosis via a splenic

lymphocyte subpopulation. Respir Res. 2018;19(1):71.

38. Jenkins RG, Moore BB, Chambers RC, Eickelberg O, Königshoff M, Kolb

M, et al. An official American thoracic society workshop report: use of

animal models for the preclinical assessment of potential therapies for

pulmonary fibrosis. Am J Respir Cell Mol Biol. 2017;56(5):667–79.

39. Mock JR, Garibaldi BT, Aggarwal NR, Jenkins J, Limjunyawong N, Singer

BD, et al. Foxp3+ regulatory T cells promote lung epithelial proliferation.

Mucosal Immunol. 2014;7(6):1440–51.

40. Moyé S, Bormann T, Maus R, Sparwasser T, Sandrock I, Prinz I, et al. Regulatory T cells limit pneumococcus-induced exacerbation of lung fibrosis in

mice. J Immunol (Baltimore Md 1950). 2020;204(9):2429–38.

41. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal

developmental pathways for the generation of pathogenic effector TH17

and regulatory T cells. Nature. 2006;441(7090):235–8.

42. Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J. Adenovector-mediated

gene transfer of active transforming growth factor-beta1 induces prolonged severe fibrosis in rat lung. J Clin Investig. 1997;100(4):768–76.

43. Tatler AL, Jenkins G. TGF-β activation and lung fibrosis. Proc Am Thorac

Soc. 2012;9(3):130–6.

44. Araujo GR, Aglas L, Vaz ER, Machado Y, Huber S, Himly M, et al. TGFβ1

mimetic peptide modulates immune response to grass pollen allergens

in mice. Allergy. 2020;75(4):882–91.

45. Cui L, Chen SY, Lerbs T, Lee JW, Domizi P, Gordon S, et al. Activation of JUN

in fibroblasts promotes pro-fibrotic programme and modulates protective immunity. Nat Commun. 2020;11(1):2795.

46. Kobayashi T, Tanaka K, Fujita T, Umezawa H, Amano H, Yoshioka K, et al.

Bidirectional role of IL-6 signal in pathogenesis of lung fibrosis. Respir Res.

2015;16(1):99.

Page 16 of 16

47. Di Rocco G, Gentile A, Antonini A, Truffa S, Piaggio G, Capogrossi MC, et al.

Analysis of biodistribution and engraftment into the liver of genetically

modified mesenchymal stromal cells derived from adipose tissue. Cell

Transplant. 2012;21(9):1997–2008.

48. Eggenhofer E, Benseler V, Kroemer A, Popp FC, Geissler EK, Schlitt HJ, et al.

Mesenchymal stem cells are short-lived and do not migrate beyond the

lungs after intravenous infusion. Front Immunol. 2012;3:297.

49. Shen Q, Chen B, Xiao Z, Zhao L, Xu X, Wan X, et al. Paracrine factors from

mesenchymal stem cells attenuate epithelial injury and lung fibrosis. Mol

Med Rep. 2015;11(4):2831–7.

50. Yamada Y, Nakashima A, Doi S, Ishiuchi N, Kanai R, Miyasako K, et al.

Localization and maintenance of engrafted mesenchymal stem cells

administered via renal artery in kidneys with ischemia-reperfusion injury.

Int J Mol Sci. 2021;22(8).

51. Toma C, Wagner WR, Bowry S, Schwartz A, Villanueva F. Fate of cultureexpanded mesenchymal stem cells in the microvasculature: in vivo

observations of cell kinetics. Circ Res. 2009;104(3):398–402.

52. Liao L, Shi B, Chang H, Su X, Zhang L, Bi C, et al. Heparin improves BMSC

cell therapy: anticoagulant treatment by heparin improves the safety

and therapeutic effect of bone marrow-derived mesenchymal stem cell

cytotherapy. Theranostics. 2017;7(1):106–16.

53. Srour N, Thébaud B. Mesenchymal stromal cells in animal bleomycin

pulmonary fibrosis models: a systematic review. Stem Cells Transl Med.

2015;4(12):1500–10.

54. Laffey JG, Matthay MA. Fifty years of research in ARDS. Cell-based therapy

for acute respiratory distress syndrome. Biology and potential therapeutic

value. Am J Respirat Crit Care Med. 2017;196(3):266–73.

55. Wise J. Covid-19: arthritis drug tocilizumab reduces deaths in hospitalised

patients, study shows. BMJ. 2021;372:n433.

56. D’Alessio FR, Tsushima K, Aggarwal NR, West EE, Willett MH, Britos MF,

et al. CD4+CD25+Foxp3+ Tregs resolve experimental lung injury in

mice and are present in humans with acute lung injury. J Clin Investig.

2009;119(10):2898–913.

57. Halter S, Aimade L, Barbié M, Brisson H, Rouby JJ, Langeron O, et al. T

regulatory cells activation and distribution are modified in critically

ill patients with acute respiratory distress syndrome: a prospective

single-centre observational study. Anaesthesia Crit Care Pain Med.

2020;39(1):35–44.

58. Mohebbi SR, Baghaei K, Rostami-Nejad M, Nazemalhosseini Mojarad E,

Mirjalali H, Yadegar A, et al. Significant changes of CD4, FOXP3, CD25, and

IL6 expression level in Iranian COVID-19 patients. Gastroenterol Hepatol

Bed Bench. 2020;13(4):388–92.

59. Sadeghi A, Tahmasebi S, Mahmood A, Kuznetsova M, Valizadeh H,

Taghizadieh A, et al. Th17 and Treg cells function in SARS-CoV2 patients

compared with healthy controls. J Cell Physiol. 2021;236(4):2829–39.

60. Bluestone JA, Trotta E, Xu D. The therapeutic potential of regulatory T

cells for the treatment of autoimmune disease. Expert Opin Ther Targets.

2015;19(8):1091–103.

61. Lanzoni G, Linetsky E, Correa D, Messinger Cayetano S, Alvarez RA, Kouroupis D, et al. Umbilical cord mesenchymal stem cells for COVID-19 acute

respiratory distress syndrome: A double-blind, phase 1/2a, randomized

controlled trial. Stem Cells Transl Med. 2021:1–14.

Publisher’s Note

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

...

参考文献をもっと見る