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 「Failure of DNA double-strand break repair by tau mediates Alzheimer's disease pathology in vitro」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

Failure of DNA double-strand break repair by tau mediates Alzheimer's disease pathology in vitro

ASADA-UTSUGI Megumi UEMURA Kengo Ayaki Takashi UEMURA Maiko T MINAMIYAMA Sumio HIKIAMI Ryota 10885354 MORIMURA Toshifumi 20333338 SHODAI Akemi UEKI Takatoshi TAKAHASHI Ryosuke 0000-0002-1407-9640 KINOSHITA Ayae URUSHITANI Makoto 60332326 0000-0003-2773-9836 滋賀医科大学

2022.04.13

概要

DNA double-strand break (DSB) is the most severe form of DNA damage and accumulates with age, in which cytoskeletal proteins are polymerized to repair DSB in dividing cells. Since tau is a microtubule-associated protein, we investigate whether DSB is involved in tau pathologies in Alzheimer's disease (AD). First, immunohistochemistry reveals the frequent coexistence of DSB and phosphorylated tau in the cortex of AD patients. In vitro studies using primary mouse cortical neurons show that non-p-tau accumulates perinuclearly together with the tubulin after DSB induction with etoposide, followed by the accumulation of phosphorylated tau. Moreover, the knockdown of endogenous tau exacerbates DSB in neurons, suggesting the protective role of tau on DNA repair. Interestingly, synergistic exposure of neurons to microtubule disassembly and the DSB strikingly augments aberrant p-tau aggregation and apoptosis. These data suggest that DSB plays a pivotal role in AD-tau pathology and that the failure of DSB repair leads to tauopathy.

この論文で使われている画像

参考文献

Electron microscopy. The cultures were fixed with 4% paraformaldehyde in

phosphate buffer saline (PBS) for 8 h., washed thrice with PBS, and permeabilized

with 0.1% Triton-X100 and 1% bovine serum albumin in PBS for 10 mins. After

rinsing thrice with PBS, the cultures were incubated in 1% bovine serum albumin

in PBS for 30 mins and in primary antibody solution (anti-Tau-1 ab, 1:2000, 1%

bovine serum albumin in PBS) for 8 h After rinsing thrice with PBS again, the

cultures were incubated in secondary antibody solution (1:100, Nanogold,

Nanoprobes Inc, NY, USA, with 1% bovine serum albumin in PBS) for 8 h., rinsed

with PBS and enhanced with silver acetate solution for 12 mins67. Following this,

the cultures were rinsed thrice with DW, immersed in 0.05% sodium acetate for

1 min, rinsed thrice with DW again, immersed in 0.05% gold chloride solution for

2 mins, and rinsed with DW once more. The cultures were postfixed with 0.1%

osmium tetroxide in PBS for 30 mins and rinsed with DW three times. After

dehydration of the cultures with graded ethanol and embedding in Epon 812

(TAAB Laboratories, UK), the culture dish was removed, and ultrathin sections

(70 nm) were cut parallelly to the cell layers using an ultra-microtome (ReichertJung). Sections on 150 square-mesh grids covered with formvar were stained with

uranyl acetate, followed by lead citrate, and examined under an electron microscope (JEOL, Japan) at 80 kV.

2.

shRNA knockdown of mouse tau. We used MISSION Lentiviral Transduction

Particles to knockdown mouse tau according to the target sequences defined in

Sigma-Aldrich (SHCLNV, NM_010838, Clone ID TRCN0000091300) and MISSION shRNA non-target shRNA control transduction particles (SHC016V-1EA).

Lentiviral particles were transduced 10 μl of 1.4 × 107 VP/mL on day 0 of primary

mouse cortical neuron, which was cultured for seven days.

Comet assay. Primary neurons treated with etoposide were performed according

to Trivigen’s instructions (Trevigen CometAssay, #4250-050-K). Cell susupensions

(1 × 105 cells / ml) in PBS mixed at at a1:10 ratio with Comet LMA agarose

(Trevigen) were pitetted onto CometSlidesTM (Trevigen) and placed at 4 °C in the

dark in the dark. Slides were immersed overnight in Lysis Solution (Trivegen) at

4 °C in the dark and then in Alkaline Unwinding solution (200 mM NaOH, 1 mM

EDTA, pH13) for 1 hr at 4 °C. Slides were electrophoresed in Alkaline Electrophoresis solution (300 mM NaOH, 1 mM EDTA, pH13) for 40 mins at 1 V/ cm,

300 mA, at 4 °C. Slides were incubated in 70% EtOH for 5 mins, dry at 37 °C and

were stained with 1 × SYBR Gold (invitrogen) at room temperature for 30 mins in

the dark. Comets were visualized on BIOREVO BZ-9000 (KEYENCE) and scored

using OpenComet (imageJ).

Statistics and reproducibility. For most analyses, data is shown as the mean ±

standard error (SEM) from 2 to 4 independent experiments. Results were analyzed

using one-way or two-way ANOVA, Tukey-Kramer HSD test, and Student’s t test. The

statistical analyses were performed using JMP® 16 (SAS Institute Inc., Cary, NC, USA).

The level of significance was set at P < 0.05 (*P < 0.05, **P < 0.01, ***P < 0.001, and

ns = P ≥ 0.05). The making graph were used by GraphPad Prism (GraphPad software

9.0 (https://www.graphpad.com/scientific-software/prism) and JMP® 16 (SAS Institute

Inc., Cary, NC, USA).

Ethics approval and consent to participate. The human samples were taken for

the project ‘Pathological and biochemical studies of neurodegenerative diseases

using human autopsy brain and spinal cord’ (No. R1038) by the Kyoto University

Ethics Committee. Informed and written consents were obtained from all individuals or their guardians, before the autopsy analysis according to the Declaration of

Helsinki. All animal experiments were approved by the Animal Care and Use

Committee of Shiga University of Medical Science (2020-5-8) and Kyoto University of Medical Science (Med Kyo 20017).

1.

3.

4.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

Reporting summary. Further information on research design is available in the Nature

Research Reporting Summary linked to this article.

26.

Data availability

27.

All data in this study is available at Mendeley Data with https://doi.org/10.17632/

cy5y2wncbz.1. Uncut Western blot images are provided in Supplementary Figures. All

relevant data including the numerical and statistical source data that underlie the graphs

in figures are provided as Supplementary Data 1(as an Excel file). Imformation of human

brain samples is provided in Suppl. Tabel 1.

28.

29.

Received: 6 June 2021; Accepted: 24 March 2022;

30.

Chow, H. M. & Herrup, K. Genomic integrity and the ageing brain. Nat. Rev.

Neurosci. 16, 672–684 (2015).

Vilela, T. C., de Andrade, V. M., Radak, Z. & de Pinho, R. A. The role of

exercise in brain DNA damage. Neural Regen. Res. 15, 1981–1985 (2020).

Kirkwood, T. B. Understanding the odd science of aging. Cell 120, 437–447

(2005).

Zhu, L. S., Wang, D. Q., Cui, K., Liu, D. & Zhu, L. Q. Emerging perspectives

on DNA double-strand breaks in neurodegenerative diseases. Curr.

Neuropharmacol. 17, 1146–1157 (2019).

Lodato, M. A. et al. Aging and neurodegeneration are associated with

increased mutations in single human neurons. Science 359, 555–559 (2018).

Pao, P. C. et al. HDAC1 modulates OGG1-initiated oxidative DNA damage

repair in the aging brain and Alzheimer’s disease. Nat. Commun. 11, 2484

(2020).

Shanbhag, N. M. et al. Early neuronal accumulation of DNA double strand

breaks in Alzheimer’s disease. Acta Neuropathol. Commun. 7, 77 (2019).

O’Connell, B. C. et al. A genome-wide camptothecin sensitivity screen

identifies a mammalian MMS22L-NFKBIL2 complex required for genomic

stability. Mol. Cell 40, 645–657 (2010).

Suberbielle, E. et al. Physiologic brain activity causes DNA double-strand

breaks in neurons, with exacerbation by amyloid-beta. Nat. Neurosci. 16,

613–621 (2013).

Jackson, S. P. & Bartek, J. The DNA-damage response in human biology and

disease. Nature 461, 1071–1078 (2009).

Aziz, K. et al. Targeting DNA damage and repair: embracing the

pharmacological era for successful cancer therapy. Pharmacol. Ther. 133,

334–350 (2012).

Zada, D., Bronshtein, I., Lerer-Goldshtein, T., Garini, Y. & Appelbaum, L.

Sleep increases chromosome dynamics to enable reduction of accumulating

DNA damage in single neurons. Nat. Commun. 10, 895 (2019).

Forth, S. & Kapoor, T. M. The mechanics of microtubule networks in cell

division. J. Cell Biol. 216, 1525–1531 (2017).

Welte, M. A. Bidirectional transport along microtubules. Curr. Biol. 14,

R525–R537 (2004).

Monda, J. K. et al. Microtubule tip tracking by the spindle and kinetochore

protein Ska1 requires diverse tubulin-interacting surfaces. Curr. Biol. 27,

3666–3675.e3666 (2017).

Shokrollahi M., Mekhail K. Interphase microtubules in nuclear organization

and genome maintenance. Trends Cell Biol. 31, 721–731(2021).

Starr, D. A. & Fridolfsson, H. N. Interactions between nuclei and the

cytoskeleton are mediated by SUN-KASH nuclear-envelope bridges. Annu.

Rev. Cell Dev. Biol. 26, 421–444 (2010).

Oshidari, R. et al. Nuclear microtubule filaments mediate non-linear

directional motion of chromatin and promote DNA repair. Nat. Commun. 9,

2567 (2018).

Lottersberger, F., Karssemeijer, R. A., Dimitrova, N. & de Lange, T. 53BP1 and

the LINC complex promote microtubule-dependent DSB mobility and DNA

repair. Cell 163, 880–893 (2015).

Poruchynsky, M. S. et al. Microtubule-targeting agents augment the toxicity of

DNA-damaging agents by disrupting intracellular trafficking of DNA repair

proteins. Proc. Natl Acad. Sci. USA 112, 1571–1576 (2015).

Caridi, C. P. et al. Nuclear F-actin and myosins drive relocalization of

heterochromatic breaks. Nature 559, 54–60 (2018).

Madabhushi, R., Pan, L. & Tsai, L. H. DNA damage and its links to

neurodegeneration. Neuron 83, 266–282 (2014).

Brochier, C. & Langley, B. Chromatin modifications associated with DNA

double-strand breaks repair as potential targets for neurological diseases.

Neurotherapeutics 10, 817–830 (2013).

Folch, J. et al. Role of cell cycle re-entry in neurons: a common apoptotic

mechanism of neuronal cell death. Neurotox. Res. 22, 195–207 (2012).

Enriquez-Rios, V. et al. DNA-PKcs, ATM, and ATR interplay maintains

genome integrity during neurogenesis. J. Neurosci. 37, 893–905 (2017).

Kanungo, J. DNA-dependent protein kinase and DNA repair: relevance to

Alzheimer’s disease. Alzheimers Res. Ther. 5, 13 (2013).

Schaser, A. J. et al. Alpha-synuclein is a DNA binding protein that modulates

DNA repair with implications for Lewy body disorders. Sci. Rep. 9, 10919

(2019).

Mitra, J. et al. Motor neuron disease-associated loss of nuclear TDP-43 is

linked to DNA double-strand break repair defects. Proc. Natl Acad. Sci. USA

116, 4696–4705 (2019).

Wang, H. et al. Mutant FUS causes DNA ligation defects to inhibit oxidative

damage repair in Amyotrophic Lateral Sclerosis. Nat. Commun. 9, 3683

(2018).

Guerrero, E. N. et al. TDP-43/FUS in motor neuron disease: complexity and

challenges. Prog. Neurobiol. 145-146, 78–97 (2016).

COMMUNICATIONS BIOLOGY | (2022)5:358 | https://doi.org/10.1038/s42003-022-03312-0 | www.nature.com/commsbio

11

ARTICLE

COMMUNICATIONS BIOLOGY | https://doi.org/10.1038/s42003-022-03312-0

31. Thadathil N. et al. DNA double-strand break accumulation in Alzheimer’s

disease: evidence from experimental models and postmortem human brains.

Mol. Neurobiol. 58, 118–131 (2021).

32. Colnaghi L., Rondelli D., Muzi-Falconi M., Sertic S. Tau and DNA damage in

neurodegeneration. Brain Sci. 10, 946 (2020).

33. Mano, T. et al. Neuron-specific methylome analysis reveals epigenetic

regulation and tau-related dysfunction of BRCA1 in Alzheimer’s disease. Proc.

Natl Acad. Sci. USA 114, E9645–E9654 (2017).

34. Benhelli-Mokrani, H. et al. Genome-wide identification of genic and

intergenic neuronal DNA regions bound by Tau protein under physiological

and stress conditions. Nucleic Acids Res. 46, 11405–11422 (2018).

35. Bou Samra, E. et al. A role for Tau protein in maintaining ribosomal DNA

stability and cytidine deaminase-deficient cell survival. Nat. Commun. 8, 693

(2017).

36. Wei, Y. et al. Binding to the minor groove of the double-strand, tau protein

prevents DNA from damage by peroxidation. PLoS ONE 3, e2600 (2008).

37. Thurston, V. C., Zinkowski, R. P. & Binder, L. I. Tau as a nucleolar protein in

human nonneural cells in vitro and in vivo. Chromosoma 105, 20–30 (1996).

38. Loomis, P. A., Howard, T. H., Castleberry, R. P. & Binder, L. I. Identification

of nuclear tau isoforms in human neuroblastoma cells. Proc. Natl Acad. Sci.

USA 87, 8422–8426 (1990).

39. Eftekharzadeh, B. et al. Tau protein disrupts nucleocytoplasmic transport in

Alzheimer’s disease. Neuron 99, 925–940.e927 (2018).

40. Goedert, M., Ghetti, B. & Spillantini, M. G. Frontotemporal dementia:

implications for understanding Alzheimer disease. Cold Spring Harb. Perspect.

Med. 2, a006254 (2012).

41. Hutton, M. et al. Association of missense and 5’-splice-site mutations in tau

with the inherited dementia FTDP-17. Nature 393, 702–705 (1998).

42. D’Souza, I. et al. Missense and silent tau gene mutations cause frontotemporal

dementia with parkinsonism-chromosome 17 type, by affecting multiple

alternative RNA splicing regulatory elements. Proc. Natl Acad. Sci. USA 96,

5598–5603 (1999).

43. Hou, Y. et al. NAD(+) supplementation normalizes key Alzheimer’s features

and DNA damage responses in a new AD mouse model with introduced DNA

repair deficiency. Proc. Natl Acad. Sci. USA 115, E1876–E1885 (2018).

44. Zheng, J. et al. Hippocampal tau oligomerization early in tau pathology

coincides with a transient alteration of mitochondrial homeostasis and DNA

repair in a mouse model of tauopathy. Acta Neuropathol. Commun. 8, 25

(2020).

45. Mah, L. J., El-Osta, A. & Karagiannis, T. C. gammaH2AX: a sensitive

molecular marker of DNA damage and repair. Leukemia 24, 679–686 (2010).

46. Thadathil, N. et al. DNA double-strand break accumulation in Alzheimer’s

disease: evidence from experimental models and postmortem human brains.

Mol. Neurobiol. 58, 118–131 (2021).

47. Amaral, N., Ryu, T., Li, X. & Chiolo, I. Nuclear dynamics of heterochromatin

repair. Trends Genet. 33, 86–100 (2017).

48. Caridi P. C., Delabaere L., Zapotoczny G., Chiolo I. And yet, it moves: nuclear

and chromatin dynamics of a heterochromatic double-strand break. Philos.

Trans. R. Soc. Lond. B Biol. Sci. 372, (2017).

49. Chiolo, I. et al. Double-strand breaks in heterochromatin move outside of a

dynamic HP1a domain to complete recombinational repair. Cell 144, 732–744

(2011).

50. Ulrich, G. et al. Phosphorylation of nuclear Tau is modulated by distinct

cellular pathways. Sci. Rep. 8, 17702 (2018).

51. Shevelyov Y. Y., Ulianov S. V. The nuclear lamina as an organizer of

chromosome architecture. Cells 8, 136 (2019).

52. Kadavath, H. et al. Tau stabilizes microtubules by binding at the interface

between tubulin heterodimers. Proc. Natl Acad. Sci. USA 112, 7501–7506

(2015).

53. Bramblett, G. T. et al. Abnormal tau phosphorylation at Ser396 in alzheimer’s

disease recapitulates development and contributes to reduced microtubule

binding. Neuron 10, 1089–1099 (1993).

54. Ludwig P. E. & Das J. M. Histology, Glial Cells. StatPearls Publishing LLC

(2021).

55. Kashiwagi, H., Shiraishi, K., Sakaguchi, K., Nakahama, T. & Kodama, S. Repair

kinetics of DNA double-strand breaks and incidence of apoptosis in mouse

neural stem/progenitor cells and their differentiated neurons exposed to

ionizing radiation. J. Radiat. Res. 59, 261–271 (2018).

56. Dinant, C., Houtsmuller, A. B. & Vermeulen, W. Chromatin structure and

DNA damage repair. Epigenet. Chromatin 1, 9 (2008).

57. Casafont, I., Palanca, A., Lafarga, V., Berciano, M. T. & Lafarga, M. Effect of

ionizing radiation in sensory ganglion neurons: organization and dynamics of

nuclear compartments of DNA damage/repair and their relationship with

transcription and cell cycle. Acta Neuropathol. 122, 481–493 (2011).

58. Tomashevski, A., Webster, D. R., Grammas, P., Gorospe, M. & Kruman, I. I.

Cyclin-C-dependent cell-cycle entry is required for activation of nonhomologous end joining DNA repair in postmitotic neurons. Cell Death

Differ. 17, 1189–1198 (2010).

12

59. Schwartz, E. I. et al. Cell cycle activation in postmitotic neurons is essential for

DNA repair. Cell Cycle 6, 318–329 (2007).

60. Yang, Y., Mufson, E. J. & Herrup, K. Neuronal cell death is preceded by cell cycle

events at all stages of Alzheimer’s disease. J. Neurosci. 23, 2557–2563 (2003).

61. Hoglinger, G. U. et al. The pRb/E2F cell-cycle pathway mediates cell death in

Parkinson’s disease. Proc. Natl Acad. Sci. USA 104, 3585–3590 (2007).

62. Ranganathan, S. & Bowser, R. Alterations in G1 to S Phase Cell-Cycle Regulators

during Amyotrophic Lateral Sclerosis. Am. J. Pathol. 162, 823–835 (2003).

63. Inoue, H. et al. Site-specific phosphorylation of Tau protein is associated with

deacetylation of microtubules in mouse spermatogenic cells during meiosis.

FEBS Lett. 588, 2003–2008 (2014).

64. Takemura, R. et al. Increased microtubule stability and alpha tubulin

acetylation in cells transfected with microtubule-associated proteins MAP1B,

MAP2 or tau. J. Cell Sci. 103, 953–964 (1992).

65. Tian, B., Yang, Q. & Mao, Z. Phosphorylation of ATM by Cdk5 mediates DNA

damage signalling and regulates neuronal death. Nat. Cell Biol. 11, 211–218 (2009).

66. Iijima-Ando, K., Zhao, L., Gatt, A., Shenton, C. & Iijima, K. A. DNA damageactivated checkpoint kinase phosphorylates tau and enhances tau-induced

neurodegeneration. Hum. Mol. Genet. 19, 1930–1938 (2010).

67. Hagiwara, H., Aoki, T., Suzuki, T. & Takata, K. Pre-embedding

immunoelectron microscopy of chemically fixed mammalian tissue culture

cells. Methods Mol. Biol. 657, 145–154 (2010).

Acknowledgements

We thank Prof. S. Takeda (Kyoto University) for DSB induction method, K. Asamoto

(Kyoto University) for immunohistochemistry and K. Tanigawa (Kyoto University) for

creating graphs. We thank the Central Research Laboratory, Shiga University of Medical

Science (CRL), and the Medical Research Support Center, Kyoto University Graduate

School of Medicine, for technical support. This work was supported by the intramural

research grant of Shiga University of Medical Science and by a research fund from Kim’s

Korean Ginseng CO. LTD.

Author contributions

M.A.-U. and M.U. gratefully contributed to the study design. M.A.-U. performed

experiments, analyzed data, and wrote a first draft of the manuscript. M.T.U., S.M., R.H.,

T.M., R.T., and A.K. gave critical advice throughout the experiments. T.A. contributed to

human immunohistochemistry. A.S. contributed to primary mouse cortical neuron

culture. T.U. conducted serial section transmission electron microscopy analysis. M.U.

and U.K. contributed to study concept and edited the manuscript. All authors read and

approved the final 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/s42003-022-03312-0.

Correspondence and requests for materials should be addressed to Makoto Urushitani.

Peer review information Communications Biology thanks Mohammad Moshahid Khan,

Luca Colnaghi and the other, anonymous, reviewer(s) for their contribution to the peer

review of this work. Primary Handling Editors: Krishnananda Chattopadhyay and

Manuel Breuer. Peer reviewer reports are 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) 2022

COMMUNICATIONS BIOLOGY | (2022)5:358 | https://doi.org/10.1038/s42003-022-03312-0 | www.nature.com/commsbio

...

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