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 「Parkinson’s disease-associated ATP13A2/PARK9 functions as a lysosomal H⁺,K⁺-ATPase」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

Parkinson’s disease-associated ATP13A2/PARK9 functions as a lysosomal H⁺,K⁺-ATPase

Fujii, Takuto Nagamori, Shushi Wiriyasermkul, Pattama Zheng, Shizhou Yago, Asaka Shimizu, Takahiro Tabuchi, Yoshiaki Okumura, Tomoyuki Fujii, Tsutomu Takeshima, Hiroshi Sakai, Hideki 京都大学 DOI:10.1038/s41467-023-37815-z

2023

概要

Mutations in the human ATP13A2 (PARK9), a lysosomal ATPase, cause Kufor-Rakeb Syndrome, an early-onset form of Parkinson’s disease (PD). Here, we demonstrate that ATP13A2 functions as a lysosomal H⁺, K⁺-ATPase. The K⁺-dependent ATPase activity and the lysosomal K⁺-transport activity of ATP13A2 are inhibited by an inhibitor of sarco/endoplasmic reticulum Ca²⁺-ATPase, thapsigargin, and K⁺-competitive inhibitors of gastric H⁺, K⁺-ATPase, such as vonoprazan and SCH28080. Interestingly, these H⁺, K⁺-ATPase inhibitors cause lysosomal alkalinization and α-synuclein accumulation, which are pathological hallmarks of PD. Furthermore, PD-associated mutants of ATP13A2 show abnormal expression and function. Our results suggest that the H⁺/K⁺-transporting function of ATP13A2 contributes to acidification and α-synuclein degradation in lysosomes.

この論文で使われている画像

参考文献

The membrane fractions of ATP13A2-transfected HEK293 cells and SHSY5Y cells were solubilized in lysis buffer supplemented with 1.5% DDM

and 0.3% CHS. Insoluble material was removed by ultracentrifugation

at 366,000 × g, 30 min at 4 °C. The samples were then injected into a

Superose 6 10/300 GL column (GE Life Sciences) connected to the

AKTAexplorer 10 XT FPLC system (GE Healthcare) at 4 °C, equilibrated

in a running buffer containing 50 mM Tris pH 7.5, 150 mM NaCl, 0.03%

DDM, and 0.0015% CHS. Fractions of 1 mL were collected, precipitated

with 10% trichloroacetic acid (TCA), and analyzed by Western blotting.

1.

Phosphorylation assay

Membrane fractions (50 μg) were added to the reaction buffer (2 mM

MgCl2, 5 mM NaN3, 17 mM HEPES, pH 6.5 or 7.4) with or without 20 mM

KCl. The reaction was initiated on ice by adding [γ-32P] ATP (2 μCi) and

stopped after 20 s with the stop solution (20% trichloroacetic acid and

10 mM phosphoric acid). After precipitation on ice for 20 min, samples

were centrifuged (20,000 × g,10 min, 4 °C). The pellet was washed with

ice-cold stop solution and distilled water and dissolved in sample

buffer comprising 2% SDS, 2.5% dithiothreitol, 10% glycerol, and

50 mM Tris-HCl, pH 6.8, and subjected to the 5% SDS-polyacrylamide

gel under acidic conditions at pH 6.057. The radioactivity was visualized

and quantified by digital autoradiography of the dried gel using

Typhoon FLA 9500.

2.

3.

4.

5.

6.

7.

8.

9.

Detection of phosphorylated α-synuclein in SH-SY5Y cells

The undifferentiated SH-SY5Y cells transiently and stably expressing αsynuclein and the differentiated SH-SY5Y cells by retinoic acid and BDNF

were treated with SCH28080, vonoprazan, and bafilomycin A1 for 48 h.

The cells were fixed with ice-cold methanol for 5 min, washed with Trisbuffered saline (TBS), and permeabilized with permeabilization buffer

containing 0.3% Triton X-100 and 0.1% BSA in TBS for 15 min at room

temperature. Non-specific binding of the antibody was blocked with 2%

BSA in TBS, and the cells were incubated with an anti-phosphorylated αsynuclein antibody (pSyn#64) (1:100) in Can Get Signal immunostain

(Toyobo) for 15 h at 4 °C. The cells were washed with TBS and incubated

with the Alexa Fluor 488-conjugated anti-mouse IgG antibody (1:100) for

1 h at room temperature. Immunofluorescence images were visualized

by using a Zeiss LSM 700 laser scanning confocal microscope. The area

(dimension) of the fluorescent signal due to phosphorylated α-synuclein

was measured by Zen3.3 software (Zeiss).

10.

11.

12.

13.

14.

15.

16.

Human tissue procurement

Human normal gastric mucosa was obtained from the dissected stomach of a Japanese gastric cancer patient (Female, 67 years) in

accordance with the recommendations of the Declaration of Helsinki

and with ethics committee approval of the University of Toyama (No.

R2017085). Informed consent was obtained from the patient at

Toyama University Hospital.

17.

18.

19.

Statistical analysis

Results are shown as mean ± SEM. Differences between groups were

analyzed by one-way analysis of variance, and correction for multiple

comparisons was made by using Tukey’s multiple comparison test. A

comparison between the two groups was made by using two-tailed

unpaired Student’s t test. Statistically significant differences were

assumed at P < 0.05.

Reporting summary

20.

21.

22.

Further information on research design is available in the Nature

Portfolio Reporting Summary linked to this article.

23.

Data availability

Source data are available as a Source Data file. Source data are provided with this paper.

Nature Communications | (2023)14:2174

Chu, Y.-T., Tai, C.-H., Lin, C.-H. & Wu, R.-M. Updates on the genetics

of Parkinson’s disease: clinical implications and future treatment.

Acta Neurol. Taiwan. 30, 83–93 (2021).

Srinivasan, E. et al. Alpha-synuclein aggregation in Parkinson’s

disease. Front. Med. 8, 736978 (2021).

Yang, X. & Xu, Y. Mutations in the ATP13A2 gene and Parkinsonism: a

preliminary review. Biomed. Res. Int. 2014, 371256 (2014).

Park, J.-S., Blair, N. F. & Sue, C. M. The role of ATP13A2 in Parkinson’s

disease: clinical phenotypes and molecular mechanisms. Mov.

Disord. 30, 770–779 (2015).

Li, W., Fu, Y., Halliday, G. M. & Sue, C. M. PARK genes link mitochondrial dysfunction and alpha-synuclein pathology in sporadic

Parkinson’s disease. Front. Cell Dev. Biol. 9, 612476 (2021).

Ramonet, D. et al. PARK9-associated ATP13A2 localizes to intracellular acidic vesicles and regulates cation homeostasis and neuronal

integrity. Hum. Mol. Genet. 21, 1725–1743 (2012).

Usenovic, M., Tresse, E., Mazzulli, J. R., Taylor, J. P. & Krainc, D.

Deficiency of ATP13A2 leads to lysosomal dysfunction, α-synuclein

accumulation, and neurotoxicity. J. Neurosci. 32, 4240–4246 (2012).

Palmgren, M. G. & Nissen, P. P-type ATPases. Annu. Rev. Biophys.

40, 243–266 (2011).

Dehay, B. et al. Loss of P-type ATPase ATP13A2/PARK9 function

induces general lysosomal deficiency and leads to Parkinson disease neurodegeneration. Proc. Natl. Acad. Sci. USA 109,

9611–9616 (2012).

Lopes da Fonseca, T., Pinho, R. & Outeiro, T. F. A familial ATP13A2

mutation enhances alpha-synuclein aggregation and promotes cell

death. Hum. Mol. Genet. 25, 2959–2971 (2016).

Thomas, T. & Thomas, T. J. Polyamine metabolism and cancer. J.

Cell. Mol. Med. 7, 113–126 (2003).

van Veen, S. et al. ATP13A2 deficiency disrupts lysosomal polyamine export. Nature 578, 419–424 (2020).

Li, P., Wang, K., Salustros, N., Grønberg, C. & Gourdon, P. Structure

and transport mechanism of P5B-ATPases. Nat. Commun. 12,

1–8 (2021).

Antony, T. et al. Cellular polyamines promote the aggregation of

alpha-synuclein. J. Biol. Chem. 278, 3235–3240 (2003).

Si, J. et al. ATP13A2 regulates cellular α-synuclein multimerization,

membrane association, and externalization. Int. J. Mol. Sci. 22,

2689 (2021).

Sim, S. I., von Bülow, S., Hummer, G. & Park, E. Structural basis of

polyamine transport by human ATP13A2 (PARK9). Mol. Cell 81,

4635–4649.e8 (2021).

Tillinghast, J., Drury, S., Bowser, D., Benn, A. & Lee, K. P. K. Structural

mechanisms for gating and ion selectivity of the human polyamine

transporter ATP13A2. Mol. Cell 81, 4650–4662.e4 (2021).

Chen, X. et al. Cryo-EM structures and transport mechanism of

human P5B type ATPase ATP13A2. Cell Discov. 7, 106 (2021).

Tomita, A. et al. Cryo-EM reveals mechanistic insights into lipidfacilitated polyamine export by human ATP13A2. Mol. Cell 81,

4799–4809.e5 (2021).

Holemans, T. et al. A lipid switch unlocks Parkinson’s diseaseassociated ATP13A2. Proc. Natl. Acad. Sci. USA 112,

9040–9045 (2015).

Gitler, A. D. et al. Alpha-synuclein is part of a diverse and highly

conserved interaction network that includes PARK9 and manganese toxicity. Nat. Genet. 41, 308–315 (2009).

Tan, J. et al. Regulation of intracellular manganese homeostasis by

Kufor-Rakeb syndrome-associated ATP13A2 protein. J. Biol. Chem.

286, 29654–29662 (2011).

Kong, S. M. Y. et al. Parkinson’s disease-linked human PARK9/

ATP13A2 maintains zinc homeostasis and promotes α-Synuclein

externalization via exosomes. Hum. Mol. Genet. 23,

2816–2833 (2014).

Article

24. Tsunemi, T. & Krainc, D. Zn2+ dyshomeostasis caused by loss of

ATP13A2/PARK9 leads to lysosomal dysfunction and alphasynuclein accumulation. Hum. Mol. Genet. 23, 2791–2801 (2014).

25. Tsunemi, T. et al. Increased lysosomal exocytosis induced by lysosomal Ca2+ channel agonists protects human dopaminergic neurons

from α-synuclein toxicity. J. Neurosci. 39, 5760–5772 (2019).

26. Wang, X. et al. TPC proteins are phosphoinositide-activated

sodium-selective ion channels in endosomes and lysosomes. Cell

151, 372–383 (2012).

27. Djarmati, A. et al. ATP13A2 variants in early-onset Parkinson’s disease patients and controls. Mov. Disord. 24, 2104–2111 (2009).

28. Fujii, T. et al. Inhibition of gastric H+,K+-ATPase by new dihydropyrazole derivative KYY-008. Biochem. Biophys. Res. Commun. 567,

177–182 (2021).

29. Inatomi, N., Matsukawa, J., Sakurai, Y. & Otake, K. Potassiumcompetitive acid blockers: Advanced therapeutic option for acidrelated diseases. Pharmacol. Ther. 168, 12–22 (2016).

30. Ward, R. M. & Kearns, G. L. Proton pump inhibitors in pediatrics:

mechanism of action, pharmacokinetics, pharmacogenetics, and

pharmacodynamics. Paediatr. Drugs 15, 119–131 (2013).

31. Morii, M., Takata, H. & Takeguchi, N. Acid activation of omeprazole

in isolated gastric vesicles, oxyntic cells, and gastric glands. Gastroenterology 96, 1453–1461 (1989).

32. Hersey, S. J., Perez, A., Matheravidathu, S. & Sachs, G. Gastric H+-K

-ATPase in situ: evidence for compartmentalization. Am. J. Physiol.

257, G539–G547 (1989).

33. Asano, S., Furumoto, R., Tega, Y., Matsuda, S. & Takeguchi, N.

Mutational analysis of the putative K+-binding site on the fourth

transmembrane segment of the gastric H+,K+-ATPase. J. Biochem.

127, 993–1000 (2000).

34. Xicoy, H., Wieringa, B. & Martens, G. J. M. The SH-SY5Y cell line in

Parkinson’s disease research: a systematic review. Mol. Neurodegener. 12, 10 (2017).

35. Dehay, B. et al. Lysosomal dysfunction in Parkinson disease:

ATP13A2 gets into the groove. Autophagy 8, 1389–1391 (2012).

36. Tsunemi, T., Hamada, K. & Krainc, D. ATP13A2/PARK9 regulates

secretion of exosomes and α-synuclein. J. Neurosci. 34,

15281–15287 (2014).

37. Fujiwara, H. et al. alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nat. Cell Biol. 4, 160–164 (2002).

38. Lee, B. R., Matsuo, Y., Cashikar, A. G. & Kamitani, T. Role of Ser129

phosphorylation of α-synuclein in melanoma cells. J. Cell Sci. 126,

696–704 (2013).

39. Terada, M. et al. The effect of truncation on prion-like properties of

α-synuclein. J. Biol. Chem. 293, 13910–13920 (2018).

40. Saito, Y. et al. Accumulation of phosphorylated α-synuclein in aging

human brain. J. Neuropathol. Exp. Neurol. 62, 644–654 (2003).

41. Delic, V. et al. Sensitivity and specificity of phospho-Ser129 αsynuclein monoclonal antibodies. J. Comp. Neurol. 526,

1978–1990 (2018).

42. Nonaka, T., Watanabe, S. T., Iwatsubo, T. & Hasegawa, M. Seeded

aggregation and toxicity of α-synuclein and tau: cellular models of

neurodegenerative diseases. J. Biol. Chem. 285,

34885–34898 (2010).

43. Guan, H. et al. Mitochondrial ferritin protects SH-SY5Y cells against

H2O2-induced oxidative stress and modulates α-synuclein expression. Exp. Neurol. 291, 51–61 (2017).

44. Schultheis, P. J. et al. Characterization of the P5 subfamily of P-type

transport ATPases in mice. Biochem. Biophys. Res. Commun. 323,

731–738 (2004).

45. Podhajska, A. et al. Common pathogenic effects of missense

mutations in the P-type ATPase ATP13A2 (PARK9) associated with

early-onset parkinsonism. PLoS ONE 7, e39942 (2012).

46. Abe, K., Irie, K., Nakanishi, H., Suzuki, H. & Fujiyoshi, Y. Crystal

structures of the gastric proton pump. Nature 556, 214–218 (2018).

Nature Communications | (2023)14:2174

https://doi.org/10.1038/s41467-023-37815-z

47. Dubey, V. et al. K+ binding and proton redistribution in the E2P state

of the H+, K+-ATPase. Sci. Rep. 8, 12732 (2018).

48. Bowman, E. J., Siebers, A. & Altendorf, K. Bafilomycins: a class of

inhibitors of membrane ATPases from microorganisms, animal

cells, and plant cells. Proc. Natl. Acad. Sci. USA 85,

7972–7976 (1988).

49. Wang, R. et al. Molecular basis of V-ATPase inhibition by bafilomycin A1. Nat. Commun. 12, 1782 (2021).

50. Vrijsen, S. et al. ATP13A2-mediated endo-lysosomal polyamine

export counters mitochondrial oxidative stress. Proc. Natl. Acad.

Sci. USA 117, 31198–31207 (2020).

51. Cao, Q. et al. BK channels alleviate lysosomal storage diseases by

providing positive feedback regulation of lysosomal Ca2+ release.

Dev. Cell 33, 427–441 (2015).

52. Wang, W. et al. A voltage-dependent K+ channel in the lysosome is

required for refilling lysosomal Ca2+ stores. J. Cell Biol. 216,

1715–1730 (2017).

53. Cang, C., Aranda, K., Seo, Y.-J., Gasnier, B. & Ren, D. TMEM175 is an

organelle K+ channel regulating lysosomal function. Cell 162,

1101–1112 (2015).

54. Jinn, S. et al. TMEM175 deficiency impairs lysosomal and mitochondrial function and increases α-synuclein aggregation. Proc.

Natl. Acad. Sci. USA 114, 2389–2394 (2017).

55. Wie, J. et al. A growth-factor-activated lysosomal K+ channel regulates Parkinson’s pathology. Nature 591, 431–437 (2021).

56. Fujii, T. et al. Survival of detached cancer cells is regulated by

movement of intracellular Na+,K+-ATPase. iScience 24, 102412 (2021).

57. Fujii, T. et al. K+-Cl- cotransporter-3a up-regulates Na+,K+-ATPase in

lipid rafts of gastric luminal parietal cells. J. Biol. Chem. 283,

6869–6877 (2008).

Acknowledgements

This work was supported by Grants-in-Aid for Scientific Research

(KAKENHI) from Japan Society for the Promotion of Science (JSPS) (to

H.S. (22H02801), Ta.F. (20K07258), S.N. (21H03365), and T.S.

(22K06827)), by JSPS Core-to-Core Program, B. Asia-Africa Science

Platforms, Tamura Science & Technology Foundation, and Platform for

drug discovery, informatics, and structural life science.

Author contributions

Ta.F. and H.S. designed all experiments and wrote the paper. Ta.F., S.N.,

P.W., S.Z., A.Y., T.S., Y.T., and T.O. performed the experiments. Ts.F. and

H.T. gave conceptual advice. All authors reviewed the results and

approved the final version of 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/s41467-023-37815-z.

Correspondence and requests for materials should be addressed to

Takuto Fujii or Hideki Sakai.

Peer review information Nature Communications thanks Curtis Okamoto and the other, anonymous, reviewer(s) for their contribution to the

peer review of this work. Peer reviewer reports are available.

Reprints and permissions 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.

10

Article

https://doi.org/10.1038/s41467-023-37815-z

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

Nature Communications | (2023)14:2174

11

...

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