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 「Inhibition of microRNA-33b specifically ameliorates abdominal aortic aneurysm formation via suppression of inflammatory pathways」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

Inhibition of microRNA-33b specifically ameliorates abdominal aortic aneurysm formation via suppression of inflammatory pathways

Yamasaki, Tomohiro 京都大学 DOI:10.14989/doctor.k24484

2023.03.23

概要

www.nature.com/scientificreports

OPEN

Inhibition of microRNA‑33b
specifically ameliorates abdominal
aortic aneurysm formation
via suppression of inflammatory
pathways
Tomohiro Yamasaki1, Takahiro Horie1*, Satoshi Koyama1, Tetsushi Nakao1, Osamu Baba1,
Masahiro Kimura1, Naoya Sowa2, Kazuhisa Sakamoto3, Kazuhiro Yamazaki3,
Satoshi Obika4,5, Yuuya Kasahara4,5, Jun Kotera6, Kozo Oka6, Ryo Fujita6, Takashi Sasaki6,
Akihiro Takemiya6, Koji Hasegawa2, Kenji Minatoya3, Takeshi Kimura1 & Koh Ono1*
Abdominal aortic aneurysm (AAA) is a lethal disease, but no beneficial therapeutic agents have been
established to date. Previously, we found that AAA formation is suppressed in microRNA (miR)-33deficient mice compared with wild-type mice. Mice have only one miR-33, but humans have two miR33 s, miR-33a and miR-33b. The data so far strongly support that inhibiting miR-33a or miR-33b will be
a new strategy to treat AAA. We produced two specific anti-microRNA oligonucleotides (AMOs) that
may inhibit miR-33a and miR-33b, respectively. In vitro studies showed that the AMO against miR33b was more effective; therefore, we examined the in vivo effects of this AMO in a calcium chloride
­(CaCl2)-induced AAA model in humanized miR-33b knock-in mice. In this model, AAA was clearly
improved by application of anti-miR-33b. To further elucidate the mechanism, we evaluated AAA
1 week after ­CaCl2 administration to examine the effect of anti-miR-33b. Histological examination
revealed that the number of MMP-9-positive macrophages and the level of MCP-1 in the aorta of
mice treated with anti-miR-33b was significantly reduced, and the serum lipid profile was improved
compared with mice treated with control oligonucleotides. These results support that inhibition of
miR-33b is effective in the treatment for AAA.
Abbreviations
AAA​ Abdominal aortic aneurysm
ABCA1 ATP binding cassette transporter A1
ALT Alanine aminotransferase
AMO Anti-microRNA oligonucleotide
AmNA Amido-bridged nucleic acid
ANOVA Analysis of variance
AST Aspartate transaminase
CaCl2 Calcium chloride
CRE Creatinine

1

Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54
Shogoin‑kawahara‑cho, Sakyo‑ku, Kyoto  606‑8507, Japan. 2Division of Translational Research, National
Hospital Organization, Kyoto Medical Center, 1‑1 Fukakusa Mukaihata‑cho, Fushimi‑ku, Kyoto  612‑8555,
Japan. 3Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54
Shogoin‑kawahara‑cho, Sakyo‑ku, Kyoto 606‑8507, Japan. 4Graduate School of Pharmaceutical Sciences, Osaka
University, 1‑6 Yamadaoka, Suita‑shi, Osaka  565‑0871, Japan. 5Center for Drug Design Research, National
Institutes of Biomedical Innovation, Health and Nutrition, 7‑6‑8 Saito‑Asagi, Ibaraki‑shi, Osaka  567‑0085,
Japan. 6Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Shonan Health Innovation
Park, 2‑26‑1, Muraoka‑Higashi, Fujisawa‑shi, Kanagawa  251‑8555, Japan. *email: thorie@kuhp.kyoto-u.ac.jp;
kohono@kuhp.kyoto-u.ac.jp
Scientific Reports |

(2022) 12:11984

| https://doi.org/10.1038/s41598-022-16017-5

1
Vol.:(0123456789)

www.nature.com/scientificreports/
DAPI 4’,6-Diamidino-2-phenylindole
DMEM Dulbecco’s modified Eagle’s medium
EC Endothelial cell
EVG Elastica van Gieson
FBS Fetal bovine serum
HDL-C High-density lipoprotein cholesterol
HE Hematoxylin–eosin
IL Interleukin
JNK C-Jun N-terminal kinase
KI Knock-in
KO Knock-out
KOKI Knock-out, knock-in
LDL-C Low-density lipoprotein-cholesterol
MCP-1 Monocyte chemotactic protein-1
miR MicroRNA
MMP Matrix metalloproteinase
PBS Phosphate-buffered saline
PM Peritoneal macrophage
SEM Standard error of the mean
αSMA α-smooth muscle actin
SREBF Sterol regulatory element-binding transcription factor
T-BIL Total bilirubin
T-CHO Total cholesterol
TG Triglycerides
TNFα Tumor necrosis factor α
VSMC Vascular smooth muscle cell
WT Wild-type
The majority of aortic aneurysms in adults are essentially asymptomatic, but the risk of rupture increases as the
diameter of the aneurysm ­increases1. Of these, abdominal aortic aneurysms (AAA) have a reported incidence of
1.5 to 2 per 1,000 people per year in the United S­ tates2. Currently, surgical treatment is considered p
­ referable3,
4
whereas treatment with various drugs such as beta-blockers , angiotensin-converting enzyme i­nhibitors5, and
calcium channel b
­ lockers6 are being investigated. However, large-scale randomized controlled trials using these
drugs have not shown clear effects. Therefore, the development of therapeutic agents that can inhibit the expansion of aortic aneurysms is of great significance.
Using genetically engineered mice, we previously found that the absence of microRNA (miR)-33 suppressed
AAA progression via several anti-inflammatory p
­ athways7. Therefore, we decided to use these data to develop
a new therapeutic agent for aortic aneurysms. Nucleic acid medicine has been attracting attention as a new
therapeutic agent for hereditary and refractory diseases that have been difficult to ­treat8,9. In the development
of conventional oligonucleotide therapeutics, there have been problems with stability and efficacy in vivo10,
but advances in modified nucleic acid ­technology11 and Drug Delivery System ­technology12 have changed this
situation, and candidates that are highly effective not only after local administration but also after systemic
administration are being d
­ eveloped13. Oligonucleotide therapeutics are expected to have high specificity and
efficacy similar to antibody drugs and can be produced by chemical synthesis similar to small molecule ­drugs10.
In fact, oligonucleotides are designed based on the target RNA sequence, and highly effective oligonucleotides
can be obtained in a short ­time13.
In rodents, there is only one miR-33 (miR-33a) in the intron of sterol regulatory element binding transcription factor 2 (Srebf2), but in humans, in addition to miR-33a, there is another miR-33 (miR-33b) in the intron of
SREBF1. In this study, we developed bridged nucleic acid-modified anti-microRNA oligonucleotides (AMOs)
that individually target miR-33a and miR-33b, which differ by only two bases. As a result, we showed that it is
possible to target aortic aneurysms and large blood vessels and elucidated its mechanism of action in detail using
humanized miR-33b knock-in (KI) mice.

Results

miR‑33b KI mice showed severe calcium chloride‑induced AAA formation.  Previously, we cre-

ated a calcium chloride ­(CaCl2)-induced AAA model in miR-33-deficient (miR-33a−/− miR-33b−/−) and wildtype (WT) mice (miR-33a+/+ miR-33b−/−) and found that AAA formation was suppressed in miR-33-deficient
­mice7. This indicated that the amount of miR-33 is related to the worsening of AAA. In the present study, we
investigated the effects of miR-33a and miR-33b on AAA in order to clarify the pathogenesis in humans. We
previously generated miR-33b KI mice (miR-33a+/+ miR-33b+/+), which have human miR-33b in intron 16 of
Srebf114. miR-33b KI mice have both miR-33a and miR-33b as in humans. In these mice, miR-33b is physiologically co-expressed with Srebf1. We next crossed miR-33a knock-out (KO) mice with miR-33b KI mice to generate
mice with only miR-33b (KOKI) (miR-33a−/− miR-33b+/+)15. Then, we compared the severity of ­CaCl2-induced
AAA ­formation16 in the WT, KOKI, and KI mouse lines. As shown in Fig. 1a,b, the diameter of the aorta was
significantly increased in KOKI mice and further increased in KI mice compared with WT mice. In addition, the
lesion length of the aorta in KI mice was significantly larger than that in KOKI and WT mice. Next, we measured
the copy numbers of miR-33a and miR-33b in 1 µg of total RNA of the aorta (Fig. 1c). As expected, only miR-33a
was present in WT mice and only miR-33b was present in KOKI mice. miR-33b levels were higher than miR-33a
Scientific Reports |
Vol:.(1234567890)

(2022) 12:11984 |

https://doi.org/10.1038/s41598-022-16017-5

2

www.nature.com/scientificreports/

Figure 1.  Genetic phenotypes of abdominal aortic aneurysm (AAA) formation in wild-type (WT) mice,
miR-33a knock-out and miR-33b knock-in (KOKI) mice, and miR-33b knock-in (KI) mice. (a) Representative
photographs of sham controls and calcium chloride ­(CaCl2)-induced AAA in WT, KOKI, and KI mice. White
bars indicate 1 mm. (b) Maximum diameter and lesion length of the abdominal aorta between the left renal
artery and the terminal aorta of ­CaCl2-induced AAA, n = 10 mice in WT, n = 9 mice in KOKI, and n = 7 mice
in KI mice. One-way ANOVA with Holm–Sidak’s multiple comparisons test. *P < 0.05 and ***P < 0.001. (c)
Absolute copy numbers of miR-33a and miR-33b in sham (left), ­CaCl2-induced AAA (mid), and integrated
graphs, n = 5 mice in each sham, n = 7 mice in WT and KOKI for ­CaCl2-induced AAA, and n = 7 mice in KI for
­CaCl2-induced AAA. One-way ANOVA with Holm–Sidak’s multiple comparisons test (left and mid) and twoway ANOVA with Holm–Sidak’s multiple comparisons test (right). *P < 0.05 and **P < 0.01. All data represent
mean ± standard error of the mean (SEM).
Scientific Reports |

(2022) 12:11984 |

https://doi.org/10.1038/s41598-022-16017-5

3
Vol.:(0123456789)

www.nature.com/scientificreports/

Figure 2.  Selection of effective and specific anti-microRNA oligonucleotide (AMOs) against miR-33a and
miR-33b. (a) Experimental scheme for reporter assay. In the absence of anti-microRNA (miR), intrinsic miR
inhibits luciferase expression (left). Adding anti-miR inhibits intrinsic miR, and the expression of luciferase will
increase. Luciferase intensity is proportional with inhibition efficiency of anti-miRs (right). (b) Reactivities of
miR-33a perfect match (33a PM) and miR-33b perfect match (33b PM) reporter vectors in HepG2 cells (human
hepatocellular carcinoma cell line), n = 3. One-way analysis of variance (ANOVA) with Dunnett’s multiple
comparison test. ***P < 0.001. ...

この論文で使われている画像

参考文献

1. Gadowski, G. R., Pilcher, D. B. & Ricci, M. A. Abdominal aortic aneurysm expansion rate: Effect of size and beta-adrenergic

blockade. J. Vasc. Surg. 19, 727–731. https://​doi.​org/​10.​1016/​s0741-​5214(94)​70048-6 (1994).

2. Folsom, A. R. et al. Circulating biomarkers and abdominal aortic aneurysm incidence: The Atherosclerosis Risk in Communities

(ARIC) Study. Circulation 132, 578–585. https://​doi.​org/​10.​1161/​circu​latio​naha.​115.​016537 (2015).

3. Soden, P. A. et al. Outcomes for symptomatic abdominal aortic aneurysms in the American College of Surgeons National Surgical

Quality Improvement Program. J. Vasc. Surg. 64, 297–305. https://​doi.​org/​10.​1016/j.​jvs.​2016.​02.​055 (2016).

4. Lindholt, J. S., Henneberg, E. W., Juul, S. & Fasting, H. Impaired results of a randomised double blinded clinical trial of propranolol

versus placebo on the expansion rate of small abdominal aortic aneurysms. Int. Angiol. 18, 52–57 (1999).

5. Sweeting, M. J., Thompson, S. G., Brown, L. C., Greenhalgh, R. M. & Powell, J. T. Use of angiotensin converting enzyme inhibitors

is associated with increased growth rate of abdominal aortic aneurysms. J. Vasc. Surg. 52, 1–4. https://​doi.​org/​10.​1016/j.​jvs.​2010.​

02.​264 (2010).

6. Brady, A. R., Thompson, S. G., Fowkes, F. G., Greenhalgh, R. M. & Powell, J. T. Abdominal aortic aneurysm expansion: Risk factors

and time intervals for surveillance. Circulation 110, 16–21. https://​doi.​org/​10.​1161/​01.​Cir.​00001​33279.​07468.​9f (2004).

7. Nakao, T. et al. Genetic ablation of MicroRNA-33 attenuates inflammation and abdominal aortic aneurysm formation via several

anti-inflammatory pathways. Arterioscler. Thromb. Vasc. Biol. 37, 2161–2170. https://​doi.​org/​10.​1161/​atvba​ha.​117.​309768 (2017).

8. Finkel, R. S. et al. Nusinersen versus sham control in infantile-onset spinal muscular atrophy. N. Engl. J. Med. 377, 1723–1732.

https://​doi.​org/​10.​1056/​NEJMo​a1702​752 (2017).

9. Cideciyan, A. V. et al. Effect of an intravitreal antisense oligonucleotide on vision in Leber congenital amaurosis due to a photoreceptor cilium defect. Nat. Med. 25, 225–228. https://​doi.​org/​10.​1038/​s41591-​018-​0295-0 (2019).

10. Crooke, S. T., Baker, B. F., Crooke, R. M. & Liang, X. H. Antisense technology: An overview and prospectus. Nat. Rev. Drug. Discov.

20, 427–453. https://​doi.​org/​10.​1038/​s41573-​021-​00162-z (2021).

11. Morihiro, K., Kasahara, Y. & Obika, S. Biological applications of xeno nucleic acids. Mol. Biosyst. 13, 235–245. https://​doi.​org/​10.​

1039/​c6mb0​0538a (2017).

12. Gökirmak, T. et al. Overcoming the challenges of tissue delivery for oligonucleotide therapeutics. Trends Pharmacol. Sci. 42,

588–604. https://​doi.​org/​10.​1016/j.​tips.​2021.​04.​010 (2021).

13. Kim, J. et al. Patient-customized oligonucleotide therapy for a rare genetic disease. N. Engl. J. Med. 381, 1644–1652. https://​doi.​

org/​10.​1056/​NEJMo​a1813​279 (2019).

14. Horie, T. et al. MicroRNA-33b knock-in mice for an intron of sterol regulatory element-binding factor 1 (Srebf1) exhibit reduced

HDL-C in vivo. Sci. Rep. 4, 5312. https://​doi.​org/​10.​1038/​srep0​5312 (2014).

15. Koyama, S. et al. Identification of differential roles of microRNA-33a and -33b during atherosclerosis progression with genetically

modified mice. J. Am. Heart Assoc. 8, e012609. https://​doi.​org/​10.​1161/​jaha.​119.​012609 (2019).

16. Daugherty, A. & Cassis, L. A. Mouse models of abdominal aortic aneurysms. Arterioscler. Thromb. Vasc. Biol. 24, 429–434. https://​

doi.​org/​10.​1161/​01.​ATV.​00001​18013.​72016.​ea (2004).

17. Yahara, A. et al. Amido-bridged nucleic acids (AmNAs): Synthesis, duplex stability, nuclease resistance, and in vitro antisense

potency. ChemBioChem 13, 2513–2516. https://​doi.​org/​10.​1002/​cbic.​20120​0506 (2012).

18. Yamamoto, T. et al. Amido-bridged nucleic acids with small hydrophobic residues enhance hepatic tropism of antisense oligonucleotides in vivo. Org. Biomol. Chem. 13, 3757–3765. https://​doi.​org/​10.​1039/​c5ob0​0242g (2015).

19. Satoh, K. et al. Cyclophilin A enhances vascular oxidative stress and the development of angiotensin II-induced aortic aneurysms.

Nat. Med. 15, 649–656. https://​doi.​org/​10.​1038/​nm.​1958 (2009).

20. Ouimet, M. et al. MicroRNA-33-dependent regulation of macrophage metabolism directs immune cell polarization in atherosclerosis. J. Clin. Invest. 125, 4334–4348. https://​doi.​org/​10.​1172/​jci81​676 (2015).

21. Ouimet, M. et al. Mycobacterium tuberculosis induces the miR-33 locus to reprogram autophagy and host lipid metabolism. Nat.

Immunol. 17, 677–686. https://​doi.​org/​10.​1038/​ni.​3434 (2016).

22. Afonso, M. S. et al. miR-33 silencing reprograms the immune cell landscape in atherosclerotic plaques. Circ. Res. 128, 1122–1138.

https://​doi.​org/​10.​1161/​circr​esaha.​120.​317914 (2021).

23. Im, S.-S. et al. Linking lipid metabolism to the innate immune response in macrophages through sterol regulatory element binding

protein-1a. Cell Metab. 13, 540–549. https://​doi.​org/​10.​1016/j.​cmet.​2011.​04.​001 (2011).

24. Reboldi, A. et al. 25-Hydroxycholesterol suppresses interleukin-1–driven inflammation downstream of type I interferon. Science

345, 679–684. https://​doi.​org/​10.​1126/​scien​ce.​12547​90 (2014).

25. Yvan-Charvet, L. et al. Increased inflammatory gene expression in ABC transporter-deficient macrophages. Circulation 118,

1837–1847. https://​doi.​org/​10.​1161/​CIRCU​LATIO​NAHA.​108.​793869 (2008).

26. Westerterp, M. et al. Deficiency of ATP-binding cassette transporters A1 and G1 in macrophages increases inflammation and

accelerates atherosclerosis in mice. Circ. Res. 112, 1456–1465. https://​doi.​org/​10.​1161/​CIRCR​ESAHA.​113.​301086 (2013).

27. Oishi, Y. et al. SREBP1 contributes to resolution of pro-inflammatory TLR4 signaling by reprogramming fatty acid metabolism.

Cell Metab. 25, 412–427. https://​doi.​org/​10.​1016/j.​cmet.​2016.​11.​009 (2017).

28. Ardans, J. A., Economou, A. P., Martinson, J. M. Jr., Zhou, M. & Wahl, L. M. Oxidized low-density and high-density lipoproteins

regulate the production of matrix metalloproteinase-1 and -9 by activated monocytes. J. Leukoc. Biol. 71, 1012–1018 (2002).

29. Barter, P. J. et al. Antiinflammatory properties of HDL. Circ. Res. 95, 764–772. https://​doi.​org/​10.​1161/​01.​RES.​00001​46094.​59640.​

13 (2004).

Scientific Reports |

Vol:.(1234567890)

(2022) 12:11984 |

https://doi.org/10.1038/s41598-022-16017-5

18

www.nature.com/scientificreports/

30. Murphy, A. J. et al. High-density lipoprotein reduces the human monocyte inflammatory response. Arterioscler. Thromb. Vasc.

Biol. 28, 2071–2077. https://​doi.​org/​10.​1161/​ATVBA​HA.​108.​168690 (2008).

31. Wang, L. et al. Triglyceride-rich lipoprotein lipolysis releases neutral and oxidized FFAs that induce endothelial cell inflammation.

J. Lipid Res. 50, 204–213. https://​doi.​org/​10.​1194/​jlr.​M7005​05-​JLR200 (2009).

32. Jacobson, T. A. et al. National lipid association recommendations for patient-centered management of dyslipidemia: Part 1—full

report. J. Clin. Lipidol. 9, 129–169. https://​doi.​org/​10.​1016/j.​jacl.​2015.​02.​003 (2015).

33. Liu, Q. K. Triglyceride-lowering and anti-inflammatory mechanisms of omega-3 polyunsaturated fatty acids for atherosclerotic

cardiovascular risk reduction. J. Clin. Lipidol. 15, 556–568. https://​doi.​org/​10.​1016/j.​jacl.​2021.​05.​007 (2021).

34. Daugherty, A., Manning, M. W. & Cassis, L. A. Angiotensin II promotes atherosclerotic lesions and aneurysms in apolipoprotein

E–deficient mice. J. Clin. Investig. 105, 1605–1612. https://​doi.​org/​10.​1172/​JCI78​18 (2000).

35. Lareyre, F. et al. TGFβ (Transforming Growth Factor-β) blockade induces a human-like disease in a nondissecting mouse model

of abdominal aortic aneurysm. Arterioscler. Thromb. Vasc. Biol. 37, 2171–2181. https://​doi.​org/​10.​1161/​ATVBA​HA.​117.​309999

(2017).

36. Lu, G. et al. A novel chronic advanced stage abdominal aortic aneurysm murine model. J. Vasc. Surg. 66, 232-242.e234. https://​

doi.​org/​10.​1016/j.​jvs.​2016.​07.​105 (2017).

37. Murali Krishna, S., Morton, S. K., Li, J. & Golledge, J. Risk factors and mouse models of abdominal aortic aneurysm rupture. Int.

J. Mol. Sci. 21, 7250 (2020).

38. Clark, J. & Montori, V. In patients with ASCVD and elevated LDL-C with maximal statin therapy, inclisiran reduced LDL-C levels

at 18 months. Ann. Intern. Med. 173, 18. https://​doi.​org/​10.​7326/​acpj2​02008​180-​018 (2020).

39. Raal, F. J. et al. Inclisiran for the treatment of heterozygous familial hypercholesterolemia. N. Engl. J. Med. 382, 1520–1530. https://​

doi.​org/​10.​1056/​NEJMo​a1913​805 (2020).

40. Ray, K. K. et al. Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N. Engl. J. Med. 382, 1507–1519. https://​

doi.​org/​10.​1056/​NEJMo​a1912​387 (2020).

41. Kobayashi, M., Inoue, K., Warabi, E., Minami, T. & Kodama, T. A simple method of isolating mouse aortic endothelial cells. J.

Atheroscler. Thromb. 12, 138–142. https://​doi.​org/​10.​5551/​jat.​12.​138 (2005).

42. Horie, T. et al. MicroRNA-33 regulates sterol regulatory element-binding protein 1 expression in mice. Nat. Commun. 4, 2883.

https://​doi.​org/​10.​1038/​ncomm​s3883 (2013).

Acknowledgements

This work was supported by the Ministry of Education, Culture, Sports, Science and Technology (MEXT) and

Japan Society for the Promotion of Science (JSPS) KAKENHI Grants 17K09860 and 20K08904 (T.H.), 1605297

(T.K.), 17H04177, 17H05599, and 20H03675 (K.O.). This work was also supported by AMED under grant

JP19fk0210112, 21ym0126013h0001, and 22ym0126074h0001 (K.O.), as well as grants from the Mochida Memorial Foundation for Medical and Pharmaceutical Research, Suzuken Memorial Foundation, Bristol Myers Squibb,

Fujiwara Memorial Foundation (T.H.) and The Vehicle Racing Commemorative Foundation (K.O.).

Author contributions

T.Y., T.H. and K.O. designed the project; T.Y., T.H., S.K., T.N., O.B., M.K. and N.S. performed experiments; T.Y.,

T.H., S.K., K.S., K.Y., S.O., Y.K., J.K., K.O., R.F., T.S., A.T., K.H., K.M., T.K. and K.O. analyzed and interpreted

data; and T.Y., T.H. and K.O. prepared the manuscript.

Competing interests The authors declare no competing interests.

Additional information

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

10.​1038/​s41598-​022-​16017-5.

Correspondence and requests for materials should be addressed to T.H. or K.O.

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) 2022

Scientific Reports |

(2022) 12:11984 |

https://doi.org/10.1038/s41598-022-16017-5

19

Vol.:(0123456789)

...

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