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 「Orai1–STIM1 Regulates Increased Ca²⁺ Mobilization, Leading to Contractile Duchenne Muscular Dystrophy Phenotypes in Patient-Derived Induced Pluripotent Stem Cells」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

Orai1–STIM1 Regulates Increased Ca²⁺ Mobilization, Leading to Contractile Duchenne Muscular Dystrophy Phenotypes in Patient-Derived Induced Pluripotent Stem Cells

Uchimura, Tomoya Sakurai, Hidetoshi 京都大学 DOI:10.3390/biomedicines9111589

2021

概要

Ca²⁺ overload is one of the factors leading to Duchenne muscular dystrophy (DMD) pathogenesis. However, the molecular targets of dystrophin deficiency-dependent Ca²⁺ overload and the correlation between Ca²⁺ overload and contractile DMD phenotypes in in vitro human models remain largely elusive. In this study, we utilized DMD patient-derived induced pluripotent stem cells (iPSCs) to differentiate myotubes using doxycycline-inducible MyoD overexpression, and searched for a target molecule that mediates dystrophin deficiency-dependent Ca²⁺ overload using commercially available chemicals and siRNAs. We found that several store-operated Ca²⁺ channel (SOC) inhibitors effectively prevented Ca²⁺+ overload and identified that STIM1–Orai1 is a molecular target of SOCs. These findings were further confirmed by demonstrating that STIM1–Orai1 inhibitors, CM4620, AnCoA4, and GSK797A, prevented Ca²⁺+ overload in dystrophic myotubes. Finally, we evaluated CM4620, AnCoA4, and GSK7975A activities using a previously reported model recapitulating a muscle fatigue-like decline in contractile performance in DMD. All three chemicals ameliorated the decline in contractile performance, indicating that modulating STIM1–Orai1-mediated Ca²⁺+ overload is effective in rescuing contractile phenotypes. In conclusion, SOCs are major contributors to dystrophin deficiency-dependent Ca²⁺+ overload through STIM1–Orai1 as molecular mediators. Modulating STIM1–Orai1 activity was effective in ameliorating the decline in contractile performance in DMD.

参考文献

1. Fairclough, R.J.; Wood, M.J.; Davies, K.E. Therapy for Duchenne muscular dystrophy: Renewed optimism from genetic approaches. Nat. Rev. Genet. 2013, 14, 373–378. [CrossRef]

2. Hoffman, E.P.; Brown, R.H., Jr.; Kunkel, L.M. Dystrophin: The protein product of the Duchenne muscular dystrophy locus. Cell

1987, 51, 919–928. [CrossRef]

3. Manzur, A.Y.; Kuntzer, T.; Pike, M.; Swan, A. Glucocorticoid corticosteroids for Duchenne muscular dystrophy. Cochrane Database Syst. Rev. 2008, CD003725. [CrossRef]

4. Matthews, E.; Brassington, R.; Kuntzer, T.; Jichi, F.; Manzur, A.Y. Corticosteroids for the treatment of Duchenne muscular dystrophy. Cochrane Database Syst Rev. 2016, CD003725. [CrossRef]

5. Duan, D. Micro-Dystrophin Gene Therapy Goes Systemic in Duchenne Muscular Dystrophy Patients. Hum. Gene Ther. 2018, 29, 733–736. [CrossRef] [PubMed]

6. Wagner, K.R.; Abdel-Hamid, H.Z.; Mah, J.K.; Campbell, C.; Guglieri, M.; Muntoni, F.; Takeshima, Y.; McDonald, C.M.; Kostera- Pruszczyk, A.; Karachunski, P.; et al. Randomized phase 2 trial and open-label extension of domagrozumab in Duchenne muscular dystrophy. Neuromuscul. Disord. 2020, 30, 492–502. [CrossRef]

7. Dzierlega, K.; Yokota, T. Optimization of antisense-mediated exon skipping for Duchenne muscular dystrophy. Gene Ther. 2020,

27, 407–416. [CrossRef]

8. Allen, D.G. Skeletal muscle function: Role of ionic changes in fatigue, damage and disease. Clin. Exp. Pharmacol. Physiol. 2004, 31, 485–493. [CrossRef] [PubMed]

9. Okinaka, S.; Kumagai, H.; Ebashi, S.; Sugita, H.; Momoi, H.; Toyokura, Y.; Fujie, Y. Serum creatine phosphokinase. Activity in progressive muscular dystrophy and neuromuscular diseases. Arch. Neurol. 1961, 4, 520–525. [CrossRef]

10. Badalamente, M.A.; Stracher, A. Delay of muscle degeneration and necrosis in mdx mice by calpain inhibition. Muscle Nerve 2000,

23, 106–111. [CrossRef]

11. Evans, N.P.; Misyak, S.A.; Robertson, J.L.; Bassaganya-Riera, J.; Grange, R.W. Immune-mediated mechanisms potentially regulate the disease time-course of duchenne muscular dystrophy and provide targets for therapeutic intervention. PM&R 2009, 1, 755–768.

12. Menazza, S.; Blaauw, B.; Tiepolo, T.; Toniolo, L.; Braghetta, P.; Spolaore, B.; Reggiani, C.; Di Lisa, F.; Bonaldo, P.; Canton, M. Oxidative stress by monoamine oxidases is causally involved in myofiber damage in muscular dystrophy. Hum. Mol. Genet. 2010, 19, 4207–4215. [CrossRef]

13. Millay, D.P.; Goonasekera, S.A.; Sargent, M.A.; Maillet, M.; Aronow, B.J.; Molkentin, J.D. Calcium influx is sufficient to induce muscular dystrophy through a TRPC-dependent mechanism. Proc. Natl. Acad. Sci. USA 2009, 106, 19023–19028. [CrossRef] [PubMed]

14. Dubinin, M.V.; Talanov, E.Y.; Tenkov, K.S.; Starinets, V.S.; Mikheeva, I.B.; Sharapov, M.G.; Belosludtsev, K.N. Duchenne muscular dystrophy is associated with the inhibition of calcium uniport in mitochondria and an increased sensitivity of the organelles to the calcium-induced permeability transition. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165674. [CrossRef]

15. Sudevan, S.; Takiura, M.; Kubota, Y.; Higashitani, N.; Cooke, M.; Ellwood, R.A.; Etheridge, T.; Szewczyk, N.J.; Higashitani, A. Mitochondrial dysfunction causes Ca2+ overload and ECM degradation-mediated muscle damage in C. elegans. FASEB J. 2019, 33, 9540–9550. [CrossRef] [PubMed]

16. Hughes, M.C.; Ramos, S.V.; Turnbull, P.C.; Rebalka, I.A.; Cao, A.; Monaco, C.M.; Varah, N.E.; Edgett, B.A.; Huber, J.S.; Tadi, P.; et al. Early myopathy in Duchenne muscular dystrophy is associated with elevated mitochondrial H2O2 emission during impaired oxidative phosphorylation. J. Cachexia Sarcopenia Muscle 2019, 10, 643–661. [CrossRef]

17. Grskovic, M.; Javaherian, A.; Strulovici, B.; Daley, G.Q. Induced pluripotent stem cells—Opportunities for disease modelling and drug discovery. Nat. Rev. Drug Discov. 2011, 10, 915–929. [CrossRef]

18. Mareedu, S.; Million, E.D.; Duan, D.; Babu, G.J. Abnormal Calcium Handling in Duchenne Muscular Dystrophy: Mechanisms and Potential Therapies. Front. Physiol. 2021, 12, 647010. [CrossRef] [PubMed]

19. Shoji, E.; Sakurai, H.; Nishino, T.; Nakahata, T.; Heike, T.; Awaya, T.; Fujii, N.; Manabe, Y.; Matsuo, M.; Sehara-Fujisawa, A. Early pathogenesis of Duchenne muscular dystrophy modelled in patient-derived human induced pluripotent stem cells. Sci. Rep. 2015, 5, 12831. [CrossRef] [PubMed]

20. Avila-Medina, J.; Mayoral-Gonzalez, I.; Dominguez-Rodriguez, A.; Gallardo-Castillo, I.; Ribas, J.; Ordoñez, A.; Rosado, J.A.; Smani, T. The Complex Role of Store Operated Calcium Entry Pathways and Related Proteins in the Function of Cardiac, Skeletal and Vascular Smooth Muscle Cells. Front. Physiol. 2018, 9, 257. [CrossRef]

21. Prakriya, M.; Lewis, R.S. Store-Operated Calcium Channels. Physiol. Rev. 2015, 95, 1383–1436. [CrossRef] [PubMed]

22. Michelucci, A.; Garcia-Castaneda, M.; Boncompagni, S.; Dirksen, R.T. Role of STIM1/ORAI1-mediated store-operated Ca(2+) entry in skeletal muscle physiology and disease. Cell Calcium 2018, 76, 101–115. [CrossRef]

23. Wei-LaPierre, L.; Carrell, E.M.; Boncompagni, S.; Protasi, F.; Dirksen, R.T. Orai1-dependent calcium entry promotes skeletal muscle growth and limits fatigue. Nat. Commun. 2013, 4, 2805. [CrossRef]

24. Boncompagni, S.; Michelucci, A.; Pietrangelo, L.; Dirksen, R.T.; Protasi, F. Exercise-dependent formation of new junctions that promote STIM1-Orai1 assembly in skeletal muscle. Sci. Rep. 2017, 7, 14286. [CrossRef]

25. Zhao, X.; Moloughney, J.G.; Zhang, S.; Komazaki, S.; Weisleder, N. Orai1 mediates exacerbated Ca2+ entry in dystrophic skeletal

muscle. PLoS ONE 2012, 7, e49862. [CrossRef]

26. Kiviluoto, S.; Decuypere, J.P.; De Smedt, H.; Missiaen, L.; Parys, J.B.; Bultynck, G. STIM1 as a key regulator for Ca2+ homeostasis in skeletal-muscle development and function. Skelet. Muscle 2011, 1, 16. [CrossRef] [PubMed]

27. Antigny, F.; Sabourin, J.; Saüc, S.; Bernheim, L.; Koenig, S.; Frieden, M. TRPC1 and TRPC4 channels functionally interact with STIM1L to promote myogenesis and maintain fast repetitive Ca2+ release in human myotubes. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 806–813. [CrossRef]

28. Goonasekera, S.A.; Davis, J.; Kwong, J.Q.; Accornero, F.; Wei-LaPierre, L.; Sargent, M.A.; Dirksen, R.T.; Molkentin, J.D. Enhanced Ca2+ influx from STIM1-Orai1 induces muscle pathology in mouse models of muscular dystrophy. Hum. Mol. Genet. 2014, 23, 3706–3715. [CrossRef]

29. Li, H.L.; Fujimoto, N.; Sasakawa, N.; Shirai, S.; Ohkame, T.; Sakuma, T.; Tanaka, M.; Amano, N.; Watanabe, A.; Sakurai, H.; et al. Precise correction of the dystrophin gene in duchenne muscular dystrophy patient induced pluripotent stem cells by TALEN and CRISPR-Cas9. Stem Cell Rep. 2015, 4, 143–154. [CrossRef]

30. Uchimura, T.; Otomo, J.; Sato, M.; Sakurai, H. A human iPS cell myogenic differentiation system permitting high-throughput drug screening. Stem Cell Res. 2017, 25, 98–106. [CrossRef]

31. Tanaka, A.; Woltjen, K.; Miyake, K.; Hotta, A.; Ikeya, M.; Yamamoto, T.; Nishino, T.; Shoji, E.; Sehara-Fujisawa, A.; Manabe, Y.; et al. Efficient and reproducible myogenic differentiation from human iPS cells: Prospects for modeling Miyoshi Myopathy in vitro. PLoS ONE 2013, 8, e61540. [CrossRef]

32. Oceguera-Yanez, F.; Kim, S.I.; Matsumoto, T.; Tan, G.W.; Xiang, L.; Hatani, T.; Kondo, T.; Ikeya, M.; Yoshida, Y.; Inoue, H.; et al. Engineering the AAVS1 locus for consistent and scalable transgene expression in human iPSCs and their differentiated derivatives. Methods 2016, 101, 43–55. [CrossRef] [PubMed]

33. Darbellay, B.; Arnaudeau, S.; Bader, C.R.; Konig, S.; Bernheim, L. STIM1L is a new actin-binding splice variant involved in fast repetitive Ca2+ release. J. Cell Biol. 2011, 194, 335–346. [CrossRef]

34. Michelucci, A.; Boncompagni, S.; Pietrangelo, L.; García-Castañeda, M.; Takano, T.; Malik, S.; Dirksen, R.T.; Protasi, F. Transverse tubule remodeling enhances Orai1-dependent Ca2+ entry in skeletal muscle. eLife 2019, 8, e47576. [CrossRef]

35. Michelucci, A.; Boncompagni, S.; Pietrangelo, L.; Takano, T.; Protasi, F.; Dirksen, R.T. Pre-assembled Ca2+ entry units and constitutively active Ca2+ entry in skeletal muscle of calsequestrin-1 knockout mice. J. Gen. Physiol. 2020, 152, e202012617. [CrossRef] [PubMed]

36. Uchimura, T.; Asano, T.; Nakata, T.; Hotta, A.; Sakurai, H. A muscle fatigue-like contractile decline was recapitulated using skeletal myotubes from Duchenne muscular dystrophy patient-derived iPSCs. Cell Rep. Med. 2021, 2, 100298. [CrossRef]

37. Kurebayashi, N.; Ogawa, Y. Depletion of Ca2+ in the sarcoplasmic reticulum stimulates Ca2+ entry into mouse skeletal muscle

fibres. J. Physiol. 2001, 533, 185–199. [CrossRef]

38. Gnanasambandam, R.; Ghatak, C.; Yasmann, A.; Nishizawa, K.; Sachs, F.; Ladokhin, A.S.; Sukharev, S.I.; Suchyna, T.M. GsMTx4: Mechanism of Inhibiting Mechanosensitive Ion Channels. Biophys. J. 2017, 112, 31–45. [CrossRef] [PubMed]

39. Xu, L.; Tripathy, A.; Pasek, D.A.; Meissner, G. Ruthenium red modifies the cardiac and skeletal muscle Ca2+ release channels

(ryanodine receptors) by multiple mechanisms. J. Biol. Chem. 1999, 274, 32680–32691. [CrossRef] [PubMed]

40. Bootman, M.D.; Collins, T.J.; Mackenzie, L.; Roderick, H.L.; Berridge, M.J.; Peppiatt, C.M. 2-aminoethoxydiphenyl borate (2-APB) is a reliable blocker of store-operated Ca2+ entry but an inconsistent inhibitor of InsP3-induced Ca2+ release. FASEB J. 2002, 16, 1145–1150. [CrossRef]

41. Hogan, P.G. The STIM1-ORAI1 microdomain. Cell Calcium 2015, 58, 357–367. [CrossRef]

42. Sadaghiani, A.M.; Lee, S.M.; Odegaard, J.I.; Leveson-Gower, D.B.; McPherson, O.M.; Novick, P.; Kim, M.R.; Koehler, A.N.; Negrin, R.; Dolmetsch, R.E.; et al. Identification of Orai1 channel inhibitors by using minimal functional domains to screen small molecule microarrays. Chem. Biol. 2014, 21, 1278–1292. [CrossRef]

43. Waldron, R.T.; Chen, Y.; Pham, H.; Go, A.; Su, H.Y.; Hu, C.; Wen, L.; Husain, S.Z.; Sugar, C.A.; Roos, J.; et al. The Orai Ca2+

channel inhibitor CM4620 targets both parenchymal and immune cells to reduce inflammation in experimental acute pancreatitis.

J. Physiol. 2019, 597, 3085–3105. [CrossRef] [PubMed]

44. Kuo, I.Y.; Ehrlich, B.E. Signaling in muscle contraction. Cold Spring Harb. Perspect. Biol. 2015, 7, a006023. [CrossRef] [PubMed]

45. Gailly, P. New aspects of calcium signaling in skeletal muscle cells: Implications in Duchenne muscular dystrophy. Biochim. Biophys. Acta 2002, 1600, 38–44. [CrossRef]

46. Tutdibi, O.; Brinkmeier, H.; Rudel, R.; Fohr, K.J. Increased calcium entry into dystrophin-deficient muscle fibres of MDX and ADR-MDX mice is reduced by ion channel blockers. J. Physiol. 1999, 515 Pt 3, 859–868. [CrossRef]

47. Bellinger, A.M.; Reiken, S.; Carlson, C.; Mongillo, M.; Liu, X.; Rothman, L.; Matecki, S.; Lacampagne, A.; Marks, A.R. Hy- pernitrosylated ryanodine receptor calcium release channels are leaky in dystrophic muscle. Nat. Med. 2009, 15, 325–330. [CrossRef]

48. Kushnir, A.; Todd, J.J.; Witherspoon, J.W.; Yuan, Q.; Reiken, S.; Lin, H.; Munce, R.H.; Wajsberg, B.; Melville, Z.; Clarke, O.B.; et al. Intracellular calcium leak as a therapeutic target for RYR1-related myopathies. Acta Neuropathol. 2020, 139, 1089–1104. [CrossRef]

49. Williams, G.S.; Boyman, L.; Chikando, A.C.; Khairallah, R.J.; Lederer, W.J. Mitochondrial calcium uptake. Proc. Natl. Acad. Sci. USA 2013, 110, 10479–10486. [CrossRef]

50. Dubinin, M.V.; Talanov, E.Y.; Tenkov, K.S.; Starinets, V.S.; Belosludtseva, N.V.; Belosludtsev, K.N. The Effect of Deflazacort Treatment on the Functioning of Skeletal Muscle Mitochondria in Duchenne Muscular Dystrophy. Int. J. Mol. Sci. 2020, 21, 8763. [CrossRef]

51. Morgan, A.J.; Jacob, R. Ionomycin enhances Ca2+ influx by stimulating store-regulated cation entry and not by a direct action at the plasma membrane. Biochem. J. 1994, 300 Pt 3, 665–672. [CrossRef] [PubMed]

52. Nikolic´, N.; Görgens, S.W.; Thoresen, G.H.; Aas, V.; Eckel, J.; Eckardt, K. Electrical pulse stimulation of cultured skeletal muscle cells as a model for in vitro exercise—Possibilities and limitations. Acta Physiol. 2017, 220, 310–331. [CrossRef] [PubMed]

53. Robin, G.; Berthier, C.; Allard, B. Sarcoplasmic reticulum Ca2+ permeation explored from the lumen side in mdx muscle fibers under voltage control. J. Gen. Physiol. 2012, 139, 209–218. [CrossRef]

54. Cho, C.H.; Woo, J.S.; Perez, C.F.; Lee, E.H. A focus on extracellular Ca2+ entry into skeletal muscle. Exp. Mol. Med. 2017, 49, e378. [CrossRef]

55. Harisseh, R.; Chatelier, A.; Magaud, C.; Deliot, N.; Constantin, B. Involvement of TRPV2 and SOCE in calcium influx disorder in DMD primary human myotubes with a specific contribution of alpha1-syntrophin and PLC/PKC in SOCE regulation. Am. J. Physiol.-Cell Physiol. 2013, 304, C881–C894. [CrossRef]

56. Zhang, I.; Hu, H. Store-Operated Calcium Channels in Physiological and Pathological States of the Nervous System. Front. Cell. Neurosci. 2020, 14, 600758. [CrossRef] [PubMed]

57. Vriens, J.; Appendino, G.; Nilius, B. Pharmacology of vanilloid transient receptor potential cation channels. Mol. Pharmacol. 2009, 75, 1262–1279. [CrossRef]

58. Griffiths, E.J. Use of ruthenium red as an inhibitor of mitochondrial Ca2+ uptake in single rat cardiomyocytes. FEBS Lett. 2000,

486, 257–260. [CrossRef]

59. Saüc, S.; Bulla, M.; Nunes, P.; Orci, L.; Marchetti, A.; Antigny, F.; Bernheim, L.; Cosson, P.; Frieden, M.; Demaurex, N. STIM1L traps and gates Orai1 channels without remodeling the cortical ER. J. Cell Sci. 2015, 128, 1568–1579. [CrossRef]

60. Horinouchi, T.; Higashi, T.; Higa, T.; Terada, K.; Mai, Y.; Aoyagi, H.; Hatate, C.; Nepal, P.; Horiguchi, M.; Harada, T.; et al. Different binding property of STIM1 and its novel splice variant STIM1L to Orai1, TRPC3, and TRPC6 channels. Biochem. Biophys. Res. Commun. 2012, 428, 252–258. [CrossRef]

参考文献をもっと見る