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 「Cellular senescence in white matter microglia is induced during ageing in mice and exacerbates the neuroinflammatory phenotype」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

Cellular senescence in white matter microglia is induced during ageing in mice and exacerbates the neuroinflammatory phenotype

Matsudaira, Tatsuyuki 大阪大学

2023.12.01

概要

Title

Cellular senescence in white matter microglia is
induced during ageing in mice and exacerbates
the neuroinflammatory phenotype

Author(s)

Matsudaira, Tatsuyuki; Nakano, Sosuke; Konishi,
Yusuke et al.

Citation

Communications Biology. 2023, 6(1), p. 665

Version Type VoR
URL
rights

https://hdl.handle.net/11094/92727
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/s42003-023-05027-2

OPEN

Cellular senescence in white matter microglia is
induced during ageing in mice and exacerbates the
neuroinflammatory phenotype

1234567890():,;

Tatsuyuki Matsudaira 1,7 ✉, Sosuke Nakano1,7, Yusuke Konishi1, Shimpei Kawamoto 1, Ken Uemura1,
Tamae Kondo1, Koki Sakurai 2, Takaaki Ozawa 2, Takatoshi Hikida 2, Okiru Komine 3,
Koji Yamanaka 3, Yuki Fujita4, Toshihide Yamashita 4, Tomonori Matsumoto1 & Eiji Hara 1,5,6 ✉

Cellular senescence, a state of irreversible cell-cycle arrest caused by a variety of cellular
stresses, is critically involved in age-related tissue dysfunction in various organs. However,
the features of cells in the central nervous system that undergo senescence and their role in
neural impairment are not well understood as yet. Here, through comprehensive investigations utilising single-cell transcriptome analysis and various mouse models, we show that
microglia, particularly in the white matter, undergo cellular senescence in the brain and spinal
cord during ageing and in disease models involving demyelination. Microglial senescence is
predominantly detected in disease-associated microglia, which appear in ageing and neurodegenerative diseases. We also find that commensal bacteria promote the accumulation of
senescent microglia and disease-associated microglia during ageing. Furthermore, knockout
of p16INK4a, a key senescence inducer, ameliorates the neuroinflammatory phenotype in
damaged spinal cords in mice. These results advance our understanding of the role of cellular
senescence in the central nervous system and open up possibilities for the treatment of agerelated neural disorders.

1 Research Institute for Microbial Diseases (RIMD), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan. 2 Laboratory for Advanced Brain
Functions, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan. 3 Department of Neuroscience and
Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan. 4 Department of
Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan. 5 Immunology Frontier
Research Center (IFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan. 6 Center for Infectious Diseases Education and Research
(CiDER), Osaka University, 2-8 Yamadaoka, Suita, Osaka 565-0871, Japan. 7These authors contributed equally: Tatsuyuki Matsudaira and Sosuke
Nakano. ✉email: matsudaira@biken.osaka-u.ac.jp; ehara@biken.osaka-u.ac.jp

COMMUNICATIONS BIOLOGY | (2023)6:665 | https://doi.org/10.1038/s42003-023-05027-2 | www.nature.com/commsbio

1

ARTICLE

W

COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-023-05027-2

hile recent medical advances have increased life
expectancy, many people suffer from age-related
neural dysfunction and neurodegenerative diseases.
Microglia, tissue-resident macrophages in the central nervous
system (CNS), play important roles in maintaining homoeostasis
by regulating neuronal activity and phagocytosing unwanted
substances1–4. Recently, disease-associated microglia (DAM) with
specific transcriptional signatures have been detected in the
brains of elderly individuals, patients with various neurodegenerative diseases and neurodegenerative disease mouse models5–9,
suggesting DAM’s involvement in age-related neural disease
pathogenesis1,3. Furthermore, a recent analysis of the enhancerpromoter interactome using human brain cells revealed that
variants related to sporadic Alzheimer’s disease (AD) were primarily restricted to microglia enhancers10. Therefore, microglial
abnormalities may be critically involved in degenerative CNS
diseases.
Accumulating evidence indicates that cellular senescence leads
to a decline in tissue function, associated with ageing and various
diseases11,12. Cellular senescence is defined as irreversible cellcycle arrest caused by a persistent DNA damage response, activated by various cellular stresses, such as telomere shortening,
oncogene activation, excessive oxidative stress and radiation13,14.
In addition to irreversible cell-cycle arrest, mainly coordinated by
p16INK4a and p21Waf1/Cip1, senescent cells exhibit a secretory
phenotype that releases various inflammatory cytokines, chemokines and growth factors into their extracellular fluid15–18. This
phenotype, now called senescence-associated secretory phenotype
(SASP), plays beneficial and detrimental roles, depending on the
biological context14,19. The accumulation of senescent cells can
cause localised inflammation in the surrounding tissues, leading
to cancer promotion and tissue dysfunction20–22. Furthermore,
senescent-like signatures in the context of neurodegenerative
diseases, including AD, multiple sclerosis (MS), Parkinson’s disease, and amyotrophic lateral sclerosis, have been detected in
various cell types in the CNS, including neural cells23–25, cerebrovascular cells26, and glial cells23,27–32. Importantly, removing
senescent cells or inhibiting their accumulation reduces neuroinflammation and ameliorates cognitive functions27–30,33,34,
demonstrating that cellular senescence may contribute to CNS
tissue dysfunction. In contrast, a comprehensive study by singlecell analysis of the hippocampus of aged mice revealed that p16positive senescent cells primarily accumulated in oligodendrocyte
progenitor cells (OPCs) and microglia during the natural ageing
process35. Moreover, single-cell analysis in mice that can detect
and remove p16-expressing cells36 showed that microglia is one
of the main populations expressing p16 in the aged brain37,
suggesting that glial cells, including microglia, are major cell types
that experience senescence during the natural aging process.
However, little is known about the regional specificity of senescent cell accumulation, mechanism of accumulation, or the outcome of the cell type-specific inhibition of senescent cells in
the CNS.
In this study, we began by confirming senescent cells in the
CNS during natural aging processes in detail and investigate their
role in neural pathogenesis. We demonstrated that senescent cells
accumulate in both the brain and spinal cord with age using p16
reporter (p16-luc) mice38, which can help visualise senescent cells
in vivo. Subsequent single-cell RNA sequencing (scRNA-seq) and
detailed histological analysis revealed that senescent cells were
primarily detected in microglia in the CNS white matter regions.
Microglial senescence was prominently identified in DAM and
further increased during ageing and in pathological conditions
involving demyelination and neurodegeneration. Furthermore,
we found that microglial senescence suppression attenuated the
neuroinflammatory phenotype, and commensal bacteria partially
2

promoted microglial senescence. These findings provide further
insights into CNS cellular senescence and shed light on the role of
microglial senescence in age-related and neurodegenerative CNS
impairment progression.
Results
Senescent microglia expressing p16INK4a accumulate in the
white matter of the brains of old mice. To determine whether
senescent cells accumulate in the ageing brain, we first analysed
young (2–3 months old) and old (over 18 months old) p16-luc
mouse brains, in which the expression of p16INK4a, a key cellular
senescence inducer, can be visualised as a bioluminescence
signal38. Old p16-luc mice exhibited a marked increase in bioluminescence signal, especially in the pons and medulla oblongata
regions, compared to young mice (Fig. 1a, b, Supplementary
Fig. 1a). Further evaluation by immunoblotting and RT-qPCR
revealed that p16INK4a mRNA and protein were highly expressed
in the brain’s white matter regions, such as the corpus callosum
and medulla oblongata in old mice (Fig. 1c, d). Furthermore, the
expression of SASP factor genes, including Il1b and Cxcl10, was
also increased, especially in the old mice’s brain regions where
p16INK4a was highly expressed (Supplementary Fig. 1b, c).
p16INK4a immunohistochemical staining using a specific antibody
(Supplementary Fig. 1d, e) confirmed that the p16INK4a-expressing cells were predominantly accumulated in the white matter of
old mice but not in that of young mice (Fig. 1e, Supplementary
Fig. 1f, g).
To characterise p16INK4a-expressing cells, the corpus callosum
was collected from young and old mice and subjected to scRNAseq analysis (Fig. 2a). Cell-type classification based on their
specific markers39 and their uniform manifold approximation
and projection (UMAP) plots (young, 4,657 cells; old, 6,789 cells)
revealed that cells expressing Cdkn2a, encoding p16INK4a, were
abundant in the white matter of old mice, mainly in the microglia
(Fig. 2b–e, Supplementary Fig. 2a–h), which is consistent with
previous scRNA-seq analyses showing high p16 expression in
microglia from the hippocampus or the whole brain of old
mice35,37. Consistent with this data, microglia sorted from the old
mice’s brains showed higher p16INK4a mRNA and protein
expression than those of young mice (Fig. 2f, g, Supplementary
Fig. 3a–c). Moreover, immunostaining analysis confirmed that
the p16INK4a-expressing cells were positive for Iba1, a microglial
marker (Fig. 2h, i). These results, together with the observation
that the γH2AX levels (another cellular senescence marker)
increased in the old mice’s microglia (Supplementary Fig. ...

この論文で使われている画像

参考文献

1.

2.

3.

4.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

17.

12

Colonna, M. & Butovsky, O. Microglia function in the central nervous system

during health and neurodegeneration. Annu. Rev. Immunol. 35, 441–468

(2017).

Nayak, D., Roth, T. L. & McGavern, D. B. Microglia development and

function. Annu. Rev. Immunol. 32, 367–402 (2014).

Prinz, M., Masuda, T., Wheeler, M. A. & Quintana, F. J. Microglia and central

nervous system-associated macrophages—from origin to disease modulation.

Annu. Rev. Immunol. 39, 251–277 (2021).

Prinz, M., Jung, S. & Priller, J. Microglia biology: onecentury of evolving

concepts. Cell 179, 292–311 (2019).

Krasemann, S. et al. The TREM2-APOE pathway drives the transcriptional

phenotype of dysfunctional microglia in neurodegenerative diseases.

Immunity 47, 566–581.e9 (2017).

Masuda, T. et al. Spatial and temporal heterogeneity of mouse and human

microglia at single-cell resolution. Nature 566, 388–392 (2019).

Keren-Shaul, H. et al. A unique microglia type associated with restricting

development of Alzheimer’s disease. Cell 169, 1276–1290.e17 (2017).

Sankowski, R. et al. Mapping microglia states in the human brain through the

integration of high-dimensional techniques. Nat. Neurosci. 22, 2098–2110

(2019).

Jordão, M. J. C. et al. Neuroimmunology: Single-cell profiling identifies

myeloid cell subsets with distinct fates during neuroinflammation. Science 363,

eaat7554 (2019).

Alexi, N. et al. Brain cell type—specific enhancer—promoter interactome

maps and disease-risk association. Science 1139, 1134–1139 (2019).

Muñoz-Espín, D. & Serrano, M. Cellular senescence: from physiology to

pathology. Nat. Rev. Mol. Cell Biol. 15, 482–496 (2014).

Campisi, J. Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 75,

685–705 (2013).

Di Micco, R., Krizhanovsky, V., Baker, D. & d’Adda di Fagagna, F. Cellular

senescence in ageing: from mechanisms to therapeutic opportunities. Nat. Rev.

Mol. Cell Biol. 22, 75–95 (2021).

Gorgoulis, V. et al. Cellular senescence: defining a path forward. Cell 179,

813–827 (2019).

Coppé, J. P. et al. Senescence-associated secretory phenotypes reveal cellnonautonomous functions of oncogenic RAS and the p53 tumor suppressor.

PLoS Biol. 6, 2853–2868 (2008).

Rodier, F. et al. Persistent DNA damage signalling triggers senescenceassociated inflammatory cytokine secretion. Nat. Cell Biol. 11, 973–979 (2009).

Acosta, J. C. et al. A complex secretory program orchestrated by the

inflammasome controls paracrine senescence. Nat. Cell Biol. 15, 978–990

(2013).

18. Kuilman, T. et al. Oncogene-induced senescence relayed by an interleukindependent inflammatory network. Cell 133, 1019–1031 (2008).

19. Chan, A. S. L. & Narita, M. Short-term gain, long-term pain: the senescence

life cycle and cancer. Genes Dev. 33, 127–143 (2019).

20. Okumura, S. et al. Gut bacteria identified in colorectal cancer patients

promote tumourigenesis via butyrate secretion. Nat. Commun. 12, 1–14

(2021).

21. Tsuji, S. et al. SARS-CoV-2 infection triggers paracrine senescence and leads

to a sustained senescence-associated inflammatory response. Nat. Aging 2,

115–124 (2022).

22. Yoshimoto, S. et al. Obesity-induced gut microbial metabolite promotes liver

cancer through senescence secretome. Nature 499, 97–101 (2013).

23. Vazquez-Villaseñor, I. et al. Expression of p16 and p21 in the frontal

association cortex of ALS/MND brains suggests neuronal cell cycle

dysregulation and astrocyte senescence in early stages of the disease.

Neuropathol. Appl. Neurobiol. 46, 171–185 (2020).

24. Dehkordi, S. K. et al. Profiling senescent cells in human brains reveals neurons

with CDKN2D/p19 and tau neuropathology. Nat. Aging 1, 1107–1116 (2021).

25. Nicaise, A. M. et al. Cellular senescence in progenitor cells contributes to

diminished remyelination potential in progressive multiple sclerosis. Proc.

Natl. Acad. Sci. 116, 9030–9039 (2019).

26. Bryant, A. G. et al. Cerebrovascular senescence is associated with tau

pathology in Alzheimer’s disease. Front. Neurol. 11, 1–16 (2020).

27. Zhang, P. et al. Senolytic therapy alleviates Aβ-associated oligodendrocyte

progenitor cell senescence and cognitive deficits in an Alzheimer’s disease

model. Nat. Neurosci. 22, 719–728 (2019).

28. Bussian, T. J. et al. Clearance of senescent glial cells prevents tau-dependent

pathology and cognitive decline. Nature 562, 578–582 (2018).

29. Gaikwad, S. et al. Tau oligomer induced HMGB1 release contributes to

cellular senescence and neuropathology linked to Alzheimer’s disease and

frontotemporal dementia. Cell Rep. 36, 109419 (2021).

30. Chinta, S. J. et al. Cellular senescence is induced by the environmental

neurotoxin paraquat and contributes to neuropathology linked to Parkinson’s

disease. Cell Rep. 22, 930–940 (2018).

31. Brelstaff, J. H. et al. Microglia become hypofunctional and release

metalloproteases and tau seeds when phagocytosing live neurons with P301S

tau aggregates. Sci. Adv. 7, 1–16 (2021).

32. Hu, Y. et al. Replicative senescence dictates the emergence of diseaseassociated microglia and contributes to Aβ pathology. Cell Rep. 35, 109228

(2021).

33. Musi, N. et al. Tau protein aggregation is associated with cellular senescence in

the brain. Aging Cell 17, e12840 (2018).

34. Zhang, X. et al. Rejuvenation of the aged brain immune cell landscape in

mice through p16-positive senescent cell clearance. Nat. Commun. 13, 5–10

(2022).

35. Ogrodnik, M. et al. Whole-body senescent cell clearance alleviates age-related

brain inflammation and cognitive impairment in mice. Aging Cell 20, 1–16

(2021).

36. Demaria, M. et al. An essential role for senescent cells in optimal wound

healing through secretion of PDGF-AA. Dev. Cell 31, 722–733 (2014).

37. Talma, N., Gerrits, E., Wang, B., Eggen, B. J. L. & Demaria, M. Identification

of distinct and age-dependent p16High microglia subtypes. Aging Cell 20,

1–13 (2021).

38. Yamakoshi, K. et al. Real-time in vivo imaging of p16Ink4a reveals cross talk

with p53. J. Cell Biol. 186, 393–407 (2009).

39. Ximerakis, M. et al. Single-cell transcriptomic profiling of the aging mouse

brain. Nat. Neurosci. 22, 1696–1708 (2019).

40. Elmore, M. R. P. et al. Replacement of microglia in the aged brain reverses

cognitive, synaptic, and neuronal deficits in mice. Aging Cell 17, e12832

(2018).

41. Kohlmeier, F., Maya-Mendoza, A. & Jackson, D. A. EdU induces DNA

damage response and cell death in mESC in culture. Chromosom. Res. 21,

87–100 (2013).

42. Elmore, M. R. P. et al. Colony-stimulating factor 1 receptor signaling is

necessary for microglia viability, unmasking a microglia progenitor cell in the

adult brain. Neuron 82, 380–397 (2014).

43. Marsh, S. E. et al. Dissection of artifactual and confounding glial signatures by

single-cell sequencing of mouse and human brain. Nat. Neurosci. 25, 306–316

(2022).

44. Erny, D. et al. Host microbiota constantly control maturation and function of

microglia in the CNS. Nat. Neurosci. 18, 965–977 (2015).

45. Erny, D. et al. Microbiota-derived acetate enables the metabolic fitness of the

brain innate immune system during health and disease. Cell Metab. 33,

2260–2276.e7 (2021).

46. Mossad, O. et al. Gut microbiota drives age-related oxidative stress and

mitochondrial damage in microglia via the metabolite N6-carboxymethyllysine.

Nat. Neurosci. 25, 295–305 (2022).

47. Gurney, M. E. et al. Motor neuron degeneration in mice that express a human

Cu,Zn superoxide dismutase mutation. Science (80-.). 264, 1772–1775 (1994).

COMMUNICATIONS BIOLOGY | (2023)6:665 | https://doi.org/10.1038/s42003-023-05027-2 | www.nature.com/commsbio

COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-023-05027-2

48. Constantinescu, C. S., Farooqi, N., O’Brien, K. & Gran, B. Experimental

autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS).

Br. J. Pharmacol. 164, 1079–1106 (2011).

49. Takeuchi, S. et al. Intrinsic cooperation between p16INK4a and p21 Waf1/Cip1

in the onset of cellular senescence and tumor suppression in vivo. Cancer Res.

70, 9381–9390 (2010).

50. Ritzel, R. M. et al. Old age increases microglial senescence, exacerbates

secondary neuroinflammation, and worsens neurological outcomes after acute

traumatic brain injury in mice. Neurobiol. Aging 77, 194–206 (2019).

51. Trias, E. et al. Emergence of microglia bearing senescence markers during

paralysis progression in a rat model of inherited ALS. Front. Aging Neurosci.

10, 1–14 (2019).

52. Safaiyan, S. et al. White matter aging drives microglial diversity. Neuron 109,

1100–1117.e10 (2021).

53. Cudejko, C. et al. p16INK4a deficiency promotes IL-4-induced polarization

and inhibits proinflammatory signaling in macrophages. Blood 118,

2556–2566 (2011).

54. Falcão, A. M. et al. Disease-specific oligodendrocyte lineage cells arise in

multiple sclerosis. Nat. Med. 24, 1837–1844 (2018).

55. Gaublomme, J. T. et al. Single-cell genomics unveils critical regulators of Th17

cell pathogenicity. Cell 163, 1400–1412 (2015).

56. Mezö, C. et al. Different effects of constitutive and induced microbiota

modulation on microglia in a mouse model of Alzheimer’s disease. Acta

Neuropathol. Commun. 8, 1–19 (2020).

57. Miyauchi, E. et al. Gut microorganisms act together to exacerbate

inflammation in spinal cords. Nature 585, 102–106 (2020).

58. Robbins, P. D. et al. Senolytic drugs: reducing senescent cell viability to extend

health span. Annu. Rev. Pharmacol. Toxicol. 61, 779–803 (2021).

59. Kirkland, J. L. & Tchkonia, T. Senolytic drugs: from discovery to translation. J.

Intern. Med. 288, 518–536 (2020).

60. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and

tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

61. Liu, Y. et al. Expression of p16INK4a prevents cancer and promotes aging in

lymphocytes. Blood 117, 3257–3267 (2011).

62. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177,

1888–1902.e21 (2019).

63. McGinnis, C. S., Murrow, L. M. & Gartner, Z. J. DoubletFinder: Doublet

detection in single-cell RNA sequencing data using artificial nearest neighbors.

Cell Syst. 8, 329–337.e4 (2019).

64. Finak, G. et al. MAST: A flexible statistical framework for assessing

transcriptional changes and characterizing heterogeneity in single-cell RNA

sequencing data. Genome Biol. 16, 1–13 (2015).

65. Aibar, S. et al. SCENIC: Single-cell regulatory network inference and

clustering. Nat. Methods 14, 1083–1086 (2017).

Acknowledgements

We acknowledge the NGS core facility of the Genome Information Research Center at

the RIMD (Osaka University) for the support in RNA sequencing and data analysis. We

also thank Ms. Masae Suzuki and Ms. Yuko Wakabayashi for caring for the experimental

mice and other members in our laboratory for discussion during the preparation of this

manuscript (Osaka University). We thank Dr Anaelle Dumas for her help in creating the

figures using elements from BioRender (Fig. 2a, Supplementary Figs. 4a, 4e, 11c and 12).

This work was supported in part by Japan Agency of Medical Research and Development

ARTICLE

(AMED) (grant JP21gm5010001, JP22gm1710004 and JP22zf0127008), Japan Society for

the Promotion of Science (JSPS) (grant JP22H00457) and the Japan Science and Technology Agency (grant JPMJMS2022-15) to E.H., and JSPS (20J01264) to T.Matsudaira.

This work was also supported in part by grants from AMED (grants JP21wm0425010

and JP21gm1510006), JSPS (grants JP21H05694 and JP22H02944) to T.H. and the

Collaborative Research Program of Institute for Protein Research, Osaka University,

ICR-21-3.

Author contributions

Ta.M. designed experiments, performed most of the experiments, analysed the data and

wrote the manuscript. S.N. helped the experiments related to EAE experiments and GF

mice. Y.K. analysed the scRNA-seq data. S.K. and K.U. established the p16 staining

system and helped with the experiment using p16-luc mice and GF mice. T.K. helped

with the sample collection and experiments related to immunofluorescence. K.S., T.O.,

T.H., Y.F. and T.Y. analysed the data. O.K. and K.Y. provided the samples from SOD1

G93A mice and analysed the data. To.M. analysed the data and helped write the

manuscript. E.H. helped write the manuscript and supervised the project.

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/s42003-023-05027-2.

Correspondence and requests for materials should be addressed to Tatsuyuki Matsudaira

or Eiji Hara.

Peer review information Communications Biology thanks Diego Gomez-Nicola, Marta

Olah and the other, anonymous, reviewer(s) for their contribution to the peer review of

this work. Primary Handling Editors: Mireya Plass and George Inglis. A peer review file is

available.

Reprints and permission 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

COMMUNICATIONS BIOLOGY | (2023)6:665 | https://doi.org/10.1038/s42003-023-05027-2 | www.nature.com/commsbio

13

...

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