Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Sigma non-opioid receptor 1 is a potential therapeutic target for long QT syndrome

Abstract

Some missense gain-of-function mutations in the CACNA1C gene, encoding calcium channel CaV1.2, cause a life-threatening form of long QT syndrome (LQTS) named Timothy syndrome with currently no clinically effective therapeutics. Here we report that pharmacological targeting of sigma non-opioid intracellular receptor 1 (SIGMAR1) can restore electrophysiological function in induced pluripotent stem cell (iPSC)-derived cardiomyocytes generated from patients with Timothy syndrome and two common forms of LQTS, type 1 (LQTS1) and type 2 (LQTS2), caused by missense trafficking mutations in potassium channels. Electrophysiological recordings demonstrate that a Food and Drug Administration (FDA)-approved cough suppressant, dextromethorphan, can be used as an agonist of SIGMAR1 to shorten the prolonged action potential in cardiomyocytes from patients with Timothy syndrome and human cellular models of LQTS1 and LQTS2. When tested in vivo, dextromethorphan also normalized the prolonged QT intervals in a mouse model of Timothy syndrome. Overall, our study demonstrates that SIGMAR1 is a potential therapeutic target for Timothy syndrome and possibly other inherited arrhythmias such as LQTS1 and LQTS2.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The effect of SIGMAR1 agonists on cardiac action potential phenotypes in cardiomyocytes derived from iPSCs of patients with TS.
Fig. 2: The effect of dextromethorphan on cardiac Ca2+ handling and ion channels in a human iPSC model of TS.
Fig. 3: The effect of dextromethorphan on phenotypes in LQTS1.
Fig. 4: The effect of dextromethorphan on phenotypes in LQTS2.
Fig. 5: The effect of dextromethorphan on the mouse model of TS.

Similar content being viewed by others

Data availability

Source data are provided with this paper.

References

  1. Berridge, M. J., Bootman, M. D. & Roderick, H. L. Calcium signalling: dynamics, homeostasis and remodelling. Nat. Rev. Mol. Cell Biol. 4, 517–529 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Wagner, S., Maier, L. S. & Bers, D. M. Role of sodium and calcium dysregulation in tachyarrhythmias in sudden cardiac death. Circ. Res. 116, 1956–1970 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Splawski, I. et al. CaV1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism. Cell 119, 19–31 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Mahida, S. et al. Genetics of congenital and drug-induced long QT syndromes: current evidence and future research perspectives. J. Interv. Card. Electrophysiol. 37, 9–19 (2013).

    Article  PubMed  Google Scholar 

  5. Yazawa, M. et al. Using induced pluripotent stem cells to investigate cardiac phenotypes in Timothy syndrome. Nature 471, 230–234 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Song, L., Park, S. E., Isseroff, Y., Morikawa, K. & Yazawa, M. Inhibition of CDK5 alleviates the cardiac phenotypes in Timothy syndrome. Stem Cell Rep. 9, 50–57 (2017).

    Article  CAS  Google Scholar 

  7. Massard, C. et al. A first in man, phase I dose-escalation study of PHA-793887, an inhibitor of multiple cyclin-dependent kinases (CDK2, 1 and 4) reveals unexpected hepatotoxicity in patients with solid tumors. Cell Cycle 10, 963–970 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Malumbres, M. Cyclin-dependent kinases. Genome Biol. 15, 122 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Maurice, T. & Goguadze, N. Sigma-1 (σ1) receptor in memory and neurodegenerative diseases. Handb. Exp. Pharmacol. 244, 81–108 (2017).

    Article  CAS  PubMed  Google Scholar 

  10. Kim, F. J. & Maher, C. M. Sigma1 pharmacology in the context of cancer. Handb. Exp. Pharmacol. 244, 237–308 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Ruscher, K. & Wieloch, T. The involvement of the sigma-1 receptor in neurodegeneration and neurorestoration. J. Pharmacol. Sci. 127, 30–35 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Morales-Lazaro, S. L., Gonzalez-Ramirez, R. & Rosenbaum, T. Molecular interplay between the sigma-1 receptor, steroids, and ion channels. Front. Pharmacol. 10, 419 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Tsai, S. Y., Pokrass, M. J., Klauer, N. R., Nohara, H. & Su, T. P. Sigma-1 receptor regulates tau phosphorylation and axon extension by shaping p35 turnover via myristic acid. Proc. Natl Acad. Sci. USA 112, 6742–6747 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Schmidt, H. R. et al. Crystal structure of the human σ1 receptor. Nature 532, 527–530 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Su, T. P. et al. Sigma compounds derived from phencyclidine: identification of PRE-084, a new, selective sigma ligand. J. Pharmacol. Exp. Ther. 259, 543–550 (1991).

    CAS  PubMed  Google Scholar 

  16. Figgitt, D. P. & McClellan, K. J. Fluvoxamine. An updated review of its use in the management of adults with anxiety disorders. Drugs 60, 925–954 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Nguyen, L., Robson, M. J., Healy, J. R., Scandinaro, A. L. & Matsumoto, R. R. Involvement of sigma-1 receptors in the antidepressant-like effects of dextromethorphan. PLoS ONE 9, e89985 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  18. De Blasio, F. et al. Cough management: a practical approach. Cough 7, 7 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Taylor, C. P., Traynelis, S. F., Siffert, J., Pope, L. E. & Matsumoto, R. R. Pharmacology of dextromethorphan: relevance to dextromethorphan/quinidine (Nuedexta®) clinical use. Pharmacol. Ther. 164, 170–182 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Rosen, H. Dextromethorphan/quinidine sulfate for pseudobulbar affect. Drugs Today 44, 661–668 (2008).

    Article  CAS  Google Scholar 

  21. Chaki, S., Tanaka, M., Muramatsu, M. & Otomo, S. NE-100, a novel potent sigma ligand, preferentially binds to sigma 1 binding sites in guinea pig brain. Eur. J. Pharmacol. 251, R1–R2 (1994).

    Article  CAS  PubMed  Google Scholar 

  22. Okuyama, S. et al. NE-100, a novel sigma receptor ligand: in vivo tests. Life Sci. 53, PL285–PL290 (1993).

    Article  CAS  PubMed  Google Scholar 

  23. Tanaka, M., Shirasaki, T., Kaku, S., Muramatsu, M. & Otomo, S. Characteristics of binding of [3H]NE-100, a novel sigma-receptor ligand, to guinea-pig brain membranes. Naunyn Schmiedebergs Arch. Pharmacol. 351, 244–251 (1995).

    Article  CAS  PubMed  Google Scholar 

  24. Mitsuda, T. et al. Sigma-1Rs are upregulated via PERK/eIF2α/ATF4 pathway and execute protective function in ER stress. Biochem. Biophys. Res. Commun. 415, 519–525 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Bhuiyan, M. S., Tagashira, H. & Fukunaga, K. Sigma-1 receptor stimulation with fluvoxamine activates Akt–eNOS signaling in the thoracic aorta of ovariectomized rats with abdominal aortic banding. Eur. J. Pharmacol. 650, 621–628 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. Montastruc, G. et al. Drugs and dilated cardiomyopathies: a case/noncase study in the French PharmacoVigilance Database. Br. J. Clin. Pharmacol. 69, 287–294 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hashimoto, K. Sigma-1 receptors and selective serotonin reuptake inhibitors: clinical implications of their relationship. Cent. Nerv. Syst. Agents Med. Chem. 9, 197–204 (2009).

    Article  CAS  PubMed  Google Scholar 

  28. Chen, T. W. et al. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature 499, 295–300 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Mueller, B. H. 2nd et al. Sigma-1 receptor stimulation attenuates calcium influx through activated L-type voltage gated calcium channels in purified retinal ganglion cells. Exp. Eye Res. 107, 21–31 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Pan, B., Guo, Y., Kwok, W. M., Hogan, Q. & Wu, H. E. Sigma-1 receptor antagonism restores injury-induced decrease of voltage-gated Ca2+ current in sensory neurons. J. Pharmacol. Exp. Ther. 350, 290–300 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Tchedre, K. T. et al. Sigma-1 receptor regulation of voltage-gated calcium channels involves a direct interaction. Invest. Ophthalmol. Vis. Sci. 49, 4993–5002 (2008).

    Article  PubMed  Google Scholar 

  32. Crottes, D. et al. Sig1R protein regulates hERG channel expression through a post-translational mechanism in leukemic cells. J. Biol. Chem. 286, 27947–27958 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Balasuriya, D. et al. A direct interaction between the sigma-1 receptor and the hERG voltage-gated K+ channel revealed by atomic force microscopy and homogeneous time-resolved fluorescence (HTRF®). J. Biol. Chem. 289, 32353–32363 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wu, Z. Y., Yu, D. J., Soong, T. W., Dawe, G. S. & Bian, J. S. Progesterone impairs human ether-a-go-go-related gene (HERG) trafficking by disruption of intracellular cholesterol homeostasis. J. Biol. Chem. 286, 22186–22194 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liang, P. et al. Drug screening using a library of human induced pluripotent stem cell-derived cardiomyocytes reveals disease-specific patterns of cardiotoxicity. Circulation 127, 1677–1691 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Mehta, A. et al. Re-trafficking of hERG reverses long QT syndrome 2 phenotype in human iPS-derived cardiomyocytes. Cardiovasc. Res. 102, 497–506 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Pasca, S. P. et al. Using iPSC-derived neurons to uncover cellular phenotypes associated with Timothy syndrome. Nat. Med. 17, 1657–1662 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Salama, G. & London, B. Mouse models of long QT syndrome. J. Physiol. 578, 43–53 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Landry, N. M., Cohen, S. & Dixon, I. M. C. Periostin in cardiovascular disease and development: a tale of two distinct roles. Basic Res. Cardiol. 113, 1 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. McCalmon, S. A. et al. Modulation of angiotensin II-mediated cardiac remodeling by the MEF2A target gene Xirp2. Circ. Res. 106, 952–960 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Yazawa, M. et al. TRIC channels are essential for Ca2+ handling in intracellular stores. Nature 448, 78–82 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Brandt, R. R., Wright, R. S., Redfield, M. M. & Burnett, J. C. Jr. Atrial natriuretic peptide in heart failure. J. Am. Coll. Cardiol. 22, 86A–92A (1993).

    Article  CAS  PubMed  Google Scholar 

  43. Thiel, W. H. et al. Proarrhythmic defects in Timothy syndrome require calmodulin kinase II. Circulation 118, 2225–2234 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ottolia, M., Torres, N., Bridge, J. H., Philipson, K. D. & Goldhaber, J. I. Na/Ca exchange and contraction of the heart. J. Mol. Cell. Cardiol. 61, 28–33 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Deisemann, H. et al. Effects of common antitussive drugs on the hERG potassium channel current. J. Cardiovasc. Pharmacol. 52, 494–499 (2008).

    Article  CAS  PubMed  Google Scholar 

  46. Itzhaki, I. et al. Modelling the long QT syndrome with induced pluripotent stem cells. Nature 471, 225–229 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Uresin, Y., Ozek, M. & Sevgi, S. Protective effects of dextromethorphan and tizanidine on ouabain-induced arrhythmias. Methods Find. Exp. Clin. Pharmacol. 24, 421–423 (2002).

    Article  CAS  PubMed  Google Scholar 

  48. Boland, D. M., Rein, J., Lew, E. O. & Hearn, W. L. Fatal cold medication intoxication in an infant. J. Anal. Toxicol. 27, 523–526 (2003).

    Article  CAS  PubMed  Google Scholar 

  49. Kaplan, B., Buchanan, J. & Krantz, M. J. QTc prolongation due to dextromethorphan. Int. J. Cardiol. 148, 363–364 (2011).

    Article  PubMed  Google Scholar 

  50. Nitert, M. D., Nagorny, C. L., Wendt, A., Eliasson, L. & Mulder, H. CaV1.2 rather than CaV1.3 is coupled to glucose-stimulated insulin secretion in INS-1 832/13 cells. J. Mol. Endocrinol. 41, 1–11 (2008).

    Article  CAS  PubMed  Google Scholar 

  51. Stracina, T. & Novakova, M. Cardiac sigma receptors—an update. Physiol. Res. 67, S561–S576 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Lewis, R., Li, J., McCormick, P. J., C, L. H. H. & Jeevaratnam, K. Is the sigma-1 receptor a potential pharmacological target for cardiac pathologies? A systematic review. Int. J. Cardiol. Heart Vasc. 26, 100449 (2020).

    PubMed  Google Scholar 

  53. Reis, G. et al. Effect of early treatment with fluvoxamine on risk of emergency care and hospitalisation among patients with COVID-19: the TOGETHER randomised, platform clinical trial. Lancet Glob. Health 10, e42–e51 (2021).

  54. Gordon, D. E. et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature 583, 459–468 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Song, L. et al. Dual optical recordings for action potentials and calcium handling in induced pluripotent stem cell models of cardiac arrhythmias using genetically encoded fluorescent indicators. Stem Cells Transl. Med. 4, 468–475 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Skarnes, W. C., Pellegrino, E. & McDonough, J. A. Improving homology-directed repair efficiency in human stem cells. Methods 164-165, 18–28 (2019).

    Article  CAS  PubMed  Google Scholar 

  57. Navarrete-Perea, J., Yu, Q., Gygi, S. P. & Paulo, J. A. Streamlined tandem mass tag (SL-TMT) protocol: an efficient strategy for quantitative (phospho)proteome profiling using tandem mass tag-synchronous precursor selection-MS3. J. Proteome Res. 17, 2226–2236 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).

    Article  CAS  PubMed  Google Scholar 

  59. Pan, L. et al. Isolation and characterization of alternatively spliced variants of the mouse sigma1 receptor gene, Sigmar1. PLoS ONE 12, e0174694 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Nie, X. et al. An appropriate loading control for western blot analysis in animal models of myocardial ischemic infarction. Biochem. Biophys. Rep. 12, 108–113 (2017).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J.C. Wu (Stanford University) for sharing iPSC lines from patients with LQTS1 or LQTS2; G. Pitt (Weill Cornell Medicine), R. Katz, A. Rinderspacher, S. Deng, B. Corneo, R. Robinson, H. Colecraft, S. Marx, R. Kass, E. Passagué and J. Stein (Columbia University) and R. Kaye (SSI Strategy) for helpful discussions; and S. Asano (Pfizer) for helpful input about image analysis. This work was supported by NIH K99/R00HL111345, NIH R01HL138486, the Columbia University Core Usage Funding Program and a Helmsley Stem Cell Seed Grant (to M.Y.), NIH R01HL136758 (to J.P.M.), NIH F31HL142239 (to J.R.Q.), a JSPS postdoctoral fellowship (to K.M.) and the Columbia TRANSFORM TL1 training program (to R.B. and D.A.).

Author information

Authors and Affiliations

Authors

Contributions

L.S. and M.Y. conceived of and designed this project. L.S., R.B., K.M., J.R.Q. and M.Y. designed experiments. L.S., R.B., K.M., J.R.Q., A.D.K., D.A., K.Y., H.E.Y., A.C., R.Y., A.P., D.S., L.C.J., T.M.L., F.K., R.K.S., J.P.M. and M.Y. performed experiments and analyzed data: M.Y. conducted whole-cell patch clamping; R.K.S. conducted proteomics; F.K. conducted computational modeling. L.S., R.B., K.M., J.R.Q., J.S.W. and M.Y. interpreted data. M.Y. wrote the manuscript. L.S., R.B., K.M., J.R.Q., D.A., T.M.L. and J.P.M. proofread and edited the manuscript. All authors approved the manuscript.

Corresponding author

Correspondence to Masayuki Yazawa.

Ethics declarations

Competing interests

L.S., R.B., K.M., J.R.Q. and M.Y. (inventors) filed a patent (attorney docket no. 01001/005273-US1; status, filed, 4 October 2019) related to this manuscript. This patent is for using SIGMAR1 agonists to treat LQTS (types 1, 2 and 8) and related cardiac channelopathy. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 The effect of SIGMAR1 agonists on CDK5, contraction and calcium channel in Timothy syndrome iPSC-derived cardiomyocytes.

(a) Docking models of SIGMAR1-Fluvo, -Dxm and -PRE. (b) PRE-084 (5 μM, 2 hr) reduced CDK5 kinase activity in Timothy syndrome (TS) cardiomyocytes. PHA-793887 (PHA, 5 μM), a CDK5 inhibitor, was used as positive control for the assay (n = 8/group). (c-e) The effects of PRE (5 μM, 2 hr) on CDK5R1/p35 protein (c, d, n = 12 for baseline and n = 9 for 2 hr after treatment) and CDK5 protein (c, d, n = 5/group) and mRNA (e, n = 4/group). (f) Representative epi-fluorescent and phase-contrast images of TS cardiomyocytes without treatment, with PRE-084 (+PRE, 5 μM, 2 hr) or PRE-084 and NE-100 treatment (+PRE & NE-100, both 5 μM, 2 hr) from proximity ligation assay (PLA, SIGMAR1-CDK5, red, DAPI, blue). Scale bar, 10μm. (g) SIGMAR1-CDK5 PLA quantification in PRE-treated (n = 16), PRE&NE-100-treated (n = 38) and non-treated TS cardiomyocytes (n = 14). (h) Representative traces of relative motion analysis of TS cardiomyocyte contractions before (black) and after 2-hr PRE-084 treatment (blue). The representative traces were obtained from Supplementary Movies 1 and 2. (i-j) Relative changes of beating rate (i) and irregularity (j) (n = 11) of TS cardiomyocytes after PRE-084 treatment. (k) Representative traces of Ba2+ currents in TS cardiomyocytes without treatment or treated with PRE-084 (+PRE), or fluvoxamine (+Fluvo) (each, 5 μM, 2 hr) and in isogenic control cardiomyocytes (Ctrl). +Dxm representative trace is shown in Fig. 2f. (l) Voltage-dependent calcium channel inactivation was significantly enhanced by PRE (n = 10), Fluvo (n = 10) and Dxm (n = 16) treatment in TS cardiomyocytes compared to non-treated cells (n = 25). n.s., no significant differences between +PRE, + Fluvo, +Dxm and isogenic Ctrl groups (n = 13). (m) Representative traces of Ba2+ currents in TS cardiomyocytes before treatment and acutely treated with PRE (5 μM, ~5-10 mins). (n) Voltage-dependent calcium channel inactivation was not significantly changed by acute PRE treatment in TS cardiomyocytes (n = 10). All data are mean ± s.d. One-way ANOVA with Tukey’s multiple comparisons was used for b, g and One-way ANOVA with Sidak’s multiple comparisons was used for l. Paired two-tailed Student’s t-test was used for i, j, n, and unpaired two-tailed Student’s t-test was used for d, e. *P < 0.05, ** P < 0.01, *** P < 0.001, n.s., not significant. Cell samples from at least two independent differentiations were used.

Source data

Extended Data Fig. 2 The effect of SIGMAR1 agonists on action potential and SIGMAR1 and ATF4 expression profiling in Timothy syndrome iPSC-derived cardiomyocytes.

(a-c) Action potential parameters, APD50 (a), resting potential (b) and peak amplitude (c) in Timothy syndrome (TS) cardiomyocytes without treatment (n = 15) or treated for 2 hrs with 5 μM SIGMAR1 agonists, PRE-084 (+PRE, n = 13), fluvoxamine (+Fluvo, n = 10) or dextromethorphan (+Dxm, n = 11). APD90 is shown in Fig. 1d. (d-f) Action potential parameters, APD50 (d), resting potential (e) and peak amplitude (f) in isogenic control (Ctrl) cardiomyocytes without treatment (n = 10) and treated for 2 hrs with 5 μM SIGMAR1 agonists, Fluvo (n = 10) or Dxm (n = 11). (g) Quantification of human SIGMAR1 transcripts (normalized to GAPDH) in Timothy syndrome (n = 9 from two independent lines) and control cardiomyocytes (n = 12 from four independent lines). (h) Representative immunoblots of human SIGMAR1 and GAPDH protein using lysates from TS and control iPSC-derived cardiomyocytes. (i-j) Quantification of SIGMAR1 25 kDa protein band (i) and 35 kDa protein expression (j, normalized to GAPDH) in TS iPSC-derived cardiomyocytes compared to the isogenic Ctrl (n = 6/group). The molecular weight of SIGMAR1 is ~25 kDa while the 35 kDa band (#) has been reported previously and might be a dimer of a full-length SIGMAR1 with a SIGMAR1 splice variant59. (k) Representative immunoblots of human ATF4 using the same lysates from TS and isogenic Ctrl iPSC-derived cardiomyocytes shown in Extended Data Fig. 2h. (l-n) Quantification of ATF4 38 kDa protein band (l, non-modified), 50 kDa (m, phosphorylated) and 70 kDa protein expression (n, ubiquitinated, normalized to GAPDH) in TS iPSC-derived cardiomyocytes compared to the isogenic Ctrl (n = 6/group). (o-r) ATF4 overexpression (o) significantly increased SIGMAR1 transcription (p) and protein expression (q,r) in normal human cardiomyocytes transfected with ATF4 plasmid (+ ATF4, o&p, n = 7, r, n = 5). The cardiomyocytes were harvested 24 hr after the lipofection. The empty vector was used as a negative control (Mock, o&p, n = 5, r, n = 4). (q) Representative immunoblots of human SIGMAR1 and GAPDH using the lysate from the transfected cardiomyocytes. All data are mean ± s.d. One-way ANOVA with Tukey’s multiple comparisons was used for a-f and unpaired two-tailed Student’s t-test was used for g, i, j, l, m, n, o, p, r. * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001, n.s., not significant. Cell samples from at least two independent differentiations were used.

Source data

Extended Data Fig. 3 The effect of dextromethorphan on cardiac calcium and hERG channels in Timothy syndrome iPSC-derived cardiomyocytes.

(a-b) Representative traces of time-course calcium imaging in spontaneously contracting isogenic control (Ctrl, black) and Timothy syndrome (TS, red) cardiomyocytes. (c-d) Calcium transient frequency (c) and duration (d) analysis in TS cardiomyocytes during the 2-hr imaging (n = 32). (e) Current-voltage relationship of Ba2+ recordings in Dxm-treated (n = 12) and non-treated TS cardiomyocytes (n = 13). (f) Representative traces of Ba2+ currents in isogenic control cardiomyocytes without treatment (Ctrl) or treated with dextromethorphan (+Dxm). (g) Voltage-dependent calcium channel inactivation was not altered by Dxm (n = 10) in the isogenic control cardiomyocytes compared to non-treated cells (n = 13). (h) Representative epi-fluorescent and phase-contrast images of TS cardiomyocytes without and with Dxm (+Dxm) from proximity ligation assay (PLA, SIGMAR1-CaV1.2, red, DAPI, blue). Scale bar, 10μm. (i) SIGMAR1-CaV1.2 PLA quantification in Dxm-treated (n = 33) and non-treated TS cardiomyocytes (TS, n = 32). (j) IKr current steady-state amplitudes were significantly reduced in TS cardiomyocytes (n = 10) compared to isogenic control (n = 10) at -10, 0 and 10 mV steps from -40mV hold. (k) IKr tail currents were significantly reduced in TS cardiomyocytes (n = 10) compared to isogenic control (n = 10) at -10, 0, 10 and 20 mV steps. (l) Representative traces of IKr currents (E-4031-sensitive) in isogenic Ctrl cardiomyocytes (black) and TS cardiomyocytes treated with Dxm (purple), Dxm & NE-100 (green) or without treatment (red, TS). The IKr traces are shown in Fig. 2h. (m) There was no significant difference in IKr current (tail) between Dxm-treated (n = 9) and non-treated TS cardiomyocytes (n = 10) while Dxm & NE-100 (n = 10) significantly reduced tail currents compared to Dxm at 10, 20, 30, 40 and 50 mV steps and also to non-treated TS at 20 mV step from -40mV hold. All data are mean ± s.d. The treatment of Dxm or NE-100 for all experiments was 5 μM, 2hrs. One-way ANOVA with Tukey’s multiple comparisons was used for c, d between the time points to before and used for m at each voltage step. Unpaired two-tailed Student’s t-test were used for g, i between the groups and used for e, j, k at each voltage step. * P < 0.05, n.s., no significant. The samples were from at least two independent differentiations.

Source data

Extended Data Fig. 4 The effect of dextromethorphan on potassium channels in human cardiomyocytes derived from isogenic control iPSCs.

(a) Representative traces of IKr current (E-4031-sensitive) in isogenic control (Ctrl) cardiomyocytes treated with Dxm (5 μM, 2 hr) or without treatment. (b-c) IKr current amplitude (b) and tail IKr current (c) were not significantly changed by Dxm in the isogenic Ctrl cardiomyocytes (n = 10/group). (d) Schematic representation of confocal imaging for proximity ligation assay (PLA, SIGMAR1-hERG, red) in human iPSC-derived cardiomyocytes. (e) Representative confocal fluorescent images of TS cardiomyocytes used for PLA (SIGMAR1-hERG, red) and DAPI (blue) staining. Dxm treatment (+Dxm, 5 μM, 2 hr) was conducted to examine the effect of Dxm on PLA signals in TS cardiomyocytes. #3 images are used in Fig. 2j. Scale bar, 10μm. (f) Representative epi-fluorescent and phase-contrast images of isogenic Ctrl cardiomyocytes used for PLA (red, SIGMAR1-hERG) and DAPI (blue) staining. Dxm treatment (+Dxm, 5 μM, 2 hr) was conducted to examine the effect of Dxm on PLA signals in the cardiomyocytes. Scale bar, 10μm. (g) Quantification of PLA signal number of SIGMAR1-hERG in Dxm-treated (n = 21) and non-treated (n = 21) isogenic Ctrl cardiomyocytes. (h) Representative traces of IKs current (Chromanol 293B-sensitive) in isogenic control (Ctrl) cardiomyocytes treated with Dxm or without treatment. (i) IKs current amplitude analysis in the isogenic Ctrl cardiomyocytes with Dxm (5 μM, 2 hr, n = 9) or without treatment (n = 10). (j) Representative epi-fluorescent and phase-contrast images of isogenic Ctrl cardiomyocytes used for PLA (red, SIGMAR1-KV7.1) and DAPI (blue) staining. Dxm treatment (+Dxm, 5 μM, 2 hr) was conducted to examine the effect of Dxm on PLA signals in the cardiomyocytes. Scale bar, 10μm. (k) Quantification of PLA signal number of SIGMAR1-KV7.1 in Dxm-treated (n = 40) and non-treated (n = 44) isogenic Ctrl cardiomyocytes. All data are mean ± s.d. Unpaired two-tailed Student t-test was used for g,k between the groups, and for b,c,i at each voltage step. * P < 0.05. n.s., not significant. The cell samples were from at least two independent differentiations.

Source data

Extended Data Fig. 5 The effect of dextromethorphan on potassium channels in Timothy syndrome iPSC-derived cardiomyocytes.

(a) Representative confocal fluorescent images of Timothy syndrome cardiomyocytes without treatment (left, TS) and with dextromethorphan (+Dxm). Scale bar, 5μm. (b) Quantification of fraction of green (hERG) over red (WGA) fluorescence signals in TS cardiomyocytes without treatment (n = 38) and with Dxm (n = 31). (c) Quantification of KCNH2 transcript in TS cardiomyocytes with and without Dxm (n = 4/group). (d) Representative immunoblots of human hERG and GAPDH protein using lysates from non-treated and Dxm-treated TS cardiomyocytes. (e) Quantification of hERG protein expressions (normalized to GAPDH) in non-treated and Dxm-treated TS cardiomyocytes (n = 3/group). The trend towards a decreased hERG protein expression in the treated group might be driven by one sample (#) with a reduced loading amount compared with others. (f) IKs current steady-state amplitudes were significantly reduced in TS cardiomyocytes (n = 10) compared to isogenic control (n = 10) at 20 and 50 mV steps from -40mV hold. (g) Representative traces of IKs currents (Chromanol 293B-sensitive) in isogenic Ctrl cardiomyocytes (black) and TS cardiomyocytes treated with Dxm (purple), Dxm and NE-100 (green) or without treatment (red, TS). The IKs traces are shown in Fig. 2l. (h) Representative confocal fluorescent images of Timothy syndrome cardiomyocyte without treatment (left, TS) and treated with dextromethorphan (+Dxm). Scale bar, 5μm. (i) Quantification of fraction of green (KV7.1) over red (WGA) fluorescence signals in TS cardiomyocytes without treatment (n = 21) and treated with Dxm (n = 31). (j) Quantification of KCNQ1 transcripts (isoform 1 and 2) in TS cardiomyocytes without treatment and treated with Dxm (n = 4/group). (k) Representative immunoblots of human KV7.1 and GAPDH protein using lysates from non-treated and Dxm-treated TS cardiomyocytes. (l-m) Quantification of KV7.1 isoform 1 (l) and isoform 2 protein expressions (m, normalized to GAPDH) in non-treated and Dxm-treated TS cardiomyocytes (n = 3/group). All data are mean ± s.d. The treatment of Dxm or NE-100 for all experiments was 5 μM, 2hrs. Unpaired two-tailed Student’s t-test was used for b, c, e, i, j, l, m between the groups and used for f at each voltage step. * P < 0.05. n.s., not significant. The cell samples were from at least two independent differentiations.

Source data

Extended Data Fig. 6 Generation of isogenic controls from long QT syndrome type 1 and 2 iPSCs.

(a-b) Sequencing results confirmed the successful generation of isogenic control (Ctrl) iPSC lines from long QT syndrome type 1 (a) and type 2 (b). (c) Characterization of the isogenic Ctrl iPSC lines (top, phase contrast image; middle, teratoma formation assay). Scale bar, 200μm. The isogenic Ctrl iPSC demonstrated normal karyotype (bottom). (d) Representative images of TRA-1-60 (red)/TRA-1-81 (green)/Hoechst (nuclei, blue) stain in isogenic Ctrl iPSC lines. Scale bar, 20μm. (e) Representative traces of 0.5Hz-paced action potentials in LQTS1 patient-specific isogenic Ctrl iPSC-derived cardiomyocytes without treatment or treated with Dxm. (f-i) Analysis of action potential parameters, APD90 (f), APD50 (g), resting potential (h) and peak amplitude (i), in isogenic Ctrl cardiomyocytes without treatment (n = 11) or treated with Dxm (n = 10). (j) Representative traces of IKs currents (Chromanol-293B-sensitive) in long QT syndrome type 1 (LQTS1) patient-specific cardiomyocytes and the isogenic Ctrl cardiomyocytes. (k) IKs current steady-state amplitudes were significantly reduced in LQTS1 cardiomyocytes (n = 10) compared to the isogenic control (n = 11) at all voltage steps from -40mV hold. (l) Representative traces of 0.5Hz-paced action potentials in long QT syndrome type 2 (LQTS2) patient-specific isogenic Ctrl iPSC-derived cardiomyocytes without treatment or treated with Dxm. (m-p) analysis of action potential parameters, APD90 (m), APD50 (n), resting potential (o) and peak amplitude (p), in the isogenic Ctrl cardiomyocytes without treatment (n = 13) or treated with Dxm (n = 15). (q) Representative traces of IKr currents (E4031-sensitive) in LQTS2 patient-specific cardiomyocytes and the isogenic Ctrl cardiomyocytes. (r) IKr current steady-state amplitudes were significantly reduced in LQTS2 cardiomyocytes (n = 10) compared to the isogenic control (n = 12) at 10 and 20 mV steps from -40mV hold. (s) IKr tail currents were significantly reduced in LQTS2 cardiomyocytes compared to the isogenic control at 10, 20, 30, 40 and 50 mV steps from -40mV hold. All data are mean ± s.d. The treatment of Dxm for the experiments was 5 μM, 2hrs. Unpaired two-tailed Student’s t-test were used for f, g, h, i, m, n, o, p between the groups and for k, r, s at each voltage step. * P < 0.05, ** P < 0.01, n.s., not significant. The cardiomyocytes were from at least two independent differentiations.

Source data

Extended Data Fig. 7 The effect of dextromethorphan on the phenotypes in long QT syndrome type 1 and type 2.

(a-f) Analysis of APD50 (50% from peak, a, b), peak amplitude (c, d) and resting potential (e, f) in LQTS1 cardiomyocytes without treatment (n = 11) or treated with fluvoxamine (Fluvo, n = 10) or dextromethorphan (Dxm, n = 10) and in LQTS2 cardiomyocytes without treatment (n = 11) or treated with Fluvo (n = 10) or Dxm (n = 11). (g,h) IKr current (tail) was significantly increased by Dxm in LQTS1 cardiomyocytes at 40 mV step (g, LQTS1, n = 10; +Dxm, n = 9) and in LQTS2 cardiomyocytes at 10, 30 and 40 mV steps (h, LQTS2, n = 10; +Dxm, n = 9). (i,j) Representative traces of Ba2+ currents in LQTS1 (i) or LQTS2 cardiomyocytes(j) without treatment or treated with Dxm. (k,l) Voltage-dependent calcium channel inactivation was not significantly changed by Dxm in LQTS1(k) or LQTS2 cardiomyocytes (l) compared to non-treated cells (n = 10/group). (m,n) Current-voltage relationship of Ba2+ recordings in Dxm-treated (n = 10) and non-treated LQTS1 cardiomyocytes (n = 7)(m) and in Dxm-treated (n = 7) and non-treated LQTS2 cardiomyocytes (n = 8)(n). (o,p) Representative immunoblots of human SIGMAR1, ATF4, CDK5, CDK5R1/p35 and GAPDH protein using lysates from control (Ctrl) cardiomyocytes, LQTS1 cardiomyocytes (o) and LQTS2 cardiomyocytes (p). ^, p35 antibody used for Extended Data Fig. 1c was discontinued (Santa Cruz, sc-820). Therefore, another antibody (Cell Signaling Technology, C64B10) was used for this blotting series while its signal-to-noise ratio was not as high as sc-820. (q-s) Quantification of SIGMAR1 25 kDa protein band (q, r) and #35 kDa protein band (s, possibly a dimer of a full-length SIGMAR1 with a SIGMAR1 splice variant59) in LQTS1 or LQTS2 cardiomyocytes compared to Ctrl (n = 4/group). (t,u) Quantification of CDK5 (left) and CDK5R1/p35 protein expressions (right) in LQTS1 or LQTS2 cardiomyocytes compared to control cardiomyocytes (n = 4/group). (v,w) Quantification of ATF4 38 kDa protein band (left, non-modified), 50 kDa (center, phosphorylated) and 70 kDa protein expression (right, ubiquitinated) in LQTS1 or LQTS2 cardiomyocytes compared to control cardiomyocytes (n = 4/group). All data are mean ± s.d. The treatment of Dxm or Fluvo for the experiments was 5 μM, 2hrs. One-way ANOVA with Tukey’s multiple comparisons was used for a,b,c,d,e,f. Unpaired two-tailed Student’s t-test was used for k,l,q,r,s,t,u,v,w between the groups and for g,h,m,n at each voltage step. * P < 0.05, ** P < 0.01, n.s., not significant. The cardiomyocytes were from at least two independent differentiations.

Source data

Extended Data Fig. 8 The effect of dextromethorphan on Timothy syndrome mouse model.

(a) Plasma dextromethorphan (Dxm) concentrations in control mice at day 4 and day 11 after continuous dosing of Dxm in drinking water (n = 13, mean ± s.e.m.). Dxm was provided in water since day 1 at 8am. * 8 p.m. time point at day 4 had an outlier defined by statistics and the outlier was excluded from the final analysis. (b) Representative analysis procedures for ECG traces in Timothy syndrome (TS) mice without treatment or treated with Dxm at day 4 (also used in Fig. 5c,d). (c) QT intervals were significantly shortened by Dxm treatment in TS mice at day 11 (n = 8 for ctrl, n = 4 for TS, n = 7 for TS + Dxm, female, 12-week old). Three mouse heart samples from each group were used for the global proteomics analysis. All data of Extended Data Fig. 8 are mean ± s.d. except Extended Data Fig. 8a. One-way ANOVA with Tukey’s multiple comparisons were used for statistics. ** P < 0.01, n.s., not significant.

Source data

Extended Data Fig. 9 The effect of dextromethorphan on the protein expression in mouse hearts.

Altered protein expressions between control hearts (Ctrl), non-treated (TS) and Dxm-treated TS hearts (TS + Dxm) (n = 3/group). A comprehensive heat map and summary of proteomics results are shown in Fig. 5e. All data are mean ± s.d. One-way ANOVA with Tukey’s multiple comparisons was used. * P < 0.05, ** P < 0.01, n.s., not significant.

Source data

Extended Data Fig. 10 The effect of dextromethorphan on KV7.1 protein expression in mouse hearts.

(a) Representative immunoblots of KV7.1, β-tubulin and Gapdh protein using mouse heart lysates from TS (non-treated and Dxm-treated) mice and the control littermates (Ctrl) at day 11 (each, n = 3, using the same samples with the global proteomics shown in Fig. 5e). #, bands indicate putative degradation of KV7.1 in TS mouse hearts. (b-c) Quantification of β-tubulin (b) and Kcnq1/KV7.1 protein expression (c, normalized to Gapdh) in TS (non-treated and Dxm-treated) and Ctrl mouse hearts (n = 3/group). Altered β-tubulin expression has been reported previously in animal model of myocardial infarction and Gapdh was recommended as a more reliable loading control for Western blot analyses60. (d) Kcnq1/KV7.1 mRNA expression (normalized to Gapdh) in TS (non-treated and Dxm-treated) and Ctrl mouse hearts (n = 3/group). All data are mean ± s.d. One-way ANOVA with Tukey’s multiple comparisons was used. ** P < 0.01, n.s., not significant.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1

Reporting Summary

Supplementary Video 1

Representative time-lapse phase-contrast images of cardiomyocyte clusters derived from iPSCs of patients with TS before treatment with 5 μM PRE-084. Analyses are shown in Extended Data Fig. 1h–j. Red (bottom left), a marker used for this cluster area.

Supplementary Video 2

Representative time-lapse phase-contrast images of cardiomyocyte clusters derived from iPSCs of patients with TS after 2 h of treatment with 5 μM PRE-084. Analyses are shown in Extended Data Fig. 1h–j. Red (bottom left), a marker used for this cluster area.

Source data

Source Data Fig. 1

Source data for Fig 1.

Source Data Fig. 2

Source data for Fig. 2.

Source Data Fig. 3

Source data for Fig. 3.

Source Data Fig. 4

Source data for Fig. 4.

Source Data Fig. 5

Data including the global proteomic dataset (Fig. 5e).

Source Data Extended Data Fig. 1

Source data for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Source data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Source data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Source data for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Source data for Extended Data Fig. 9.

Source Data Extended Data Fig. 10

Source data for Extended Data Fig. 10.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Song, L., Bekdash, R., Morikawa, K. et al. Sigma non-opioid receptor 1 is a potential therapeutic target for long QT syndrome. Nat Cardiovasc Res 1, 142–156 (2022). https://doi.org/10.1038/s44161-021-00016-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s44161-021-00016-2

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research