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.

Modeling the mitochondrial cardiomyopathy of Barth syndrome with induced pluripotent stem cell and heart-on-chip technologies

Subjects

Abstract

Study of monogenic mitochondrial cardiomyopathies may yield insights into mitochondrial roles in cardiac development and disease. Here, we combined patient-derived and genetically engineered induced pluripotent stem cells (iPSCs) with tissue engineering to elucidate the pathophysiology underlying the cardiomyopathy of Barth syndrome (BTHS), a mitochondrial disorder caused by mutation of the gene encoding tafazzin (TAZ). Using BTHS iPSC-derived cardiomyocytes (iPSC-CMs), we defined metabolic, structural and functional abnormalities associated with TAZ mutation. BTHS iPSC-CMs assembled sparse and irregular sarcomeres, and engineered BTHS 'heart-on-chip' tissues contracted weakly. Gene replacement and genome editing demonstrated that TAZ mutation is necessary and sufficient for these phenotypes. Sarcomere assembly and myocardial contraction abnormalities occurred in the context of normal whole-cell ATP levels. Excess levels of reactive oxygen species mechanistically linked TAZ mutation to impaired cardiomyocyte function. Our study provides new insights into the pathogenesis of Barth syndrome, suggests new treatment strategies and advances iPSC-based in vitro modeling of cardiomyopathy.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Mitochondrial abnormalities in BTHS iPSC-CMs.
Figure 2: TAZ deficiency is necessary to cause the iPSC-CM metabolic phenotype.
Figure 3: Construction and characterization of TAZ mutant and isogenic control iPSCs by Cas9-mediated genome editing.
Figure 4: Sarcomere organization is impaired in BTH-H mutant iPSC-CMs.
Figure 5: BTHS myocardial tissue constructs exhibit depressed contractile stress generation.
Figure 6: Effect of small molecules on BTHS iPCS-CM ATP levels and mitochondrial function.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

References

  1. Bione, S. et al. A novel X-linked gene, G4.5. is responsible for Barth syndrome. Nat. Genet. 12, 385–389 (1996).

    Article  CAS  Google Scholar 

  2. Houtkooper, R.H. et al. The enigmatic role of tafazzin in cardiolipin metabolism. Biochim. Biophys. Acta 1788, 2003–2014 (2009).

    Article  CAS  Google Scholar 

  3. Chicco, A.J. & Sparagna, G.C. Role of cardiolipin alterations in mitochondrial dysfunction and disease. Am. J. Physiol. Cell Physiol. 292, C33–C44 (2007).

    Article  CAS  Google Scholar 

  4. Karikó, K., Buckstein, M., Ni, H. & Weissman, D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23, 165–175 (2005).

    Article  Google Scholar 

  5. Grosberg, A., Alford, P.W., McCain, M.L. & Parker, K.K. Ensembles of engineered cardiac tissues for physiological and pharmacological study: heart on a chip. Lab Chip 11, 4165–4173 (2011).

    Article  CAS  Google Scholar 

  6. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    Article  CAS  Google Scholar 

  7. Warren, L. et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 7, 618–630 (2010).

    Article  CAS  Google Scholar 

  8. Warren, L., Ni, Y., Wang, J. & Guo, X. Feeder-free derivation of human induced pluripotent stem cells with messenger RNA. Sci. Rep. 2, 657 (2012).

    Article  Google Scholar 

  9. Shiba, Y. et al. Human ES-cell-derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts. Nature 489, 322–325 (2012).

    Article  CAS  Google Scholar 

  10. Uosaki, H. et al. Efficient and scalable purification of cardiomyocytes from human embryonic and induced pluripotent stem cells by VCAM1 surface expression. PLoS ONE 6, e23657 (2011).

    Article  CAS  Google Scholar 

  11. Elliott, D.A. et al. NKX2–5(eGFP/w) hESCs for isolation of human cardiac progenitors and cardiomyocytes. Nat. Methods 8, 1037–1040 (2011).

    Article  CAS  Google Scholar 

  12. Schlame, M. et al. Phospholipid abnormalities in children with Barth syndrome. J. Am. Coll. Cardiol. 42, 1994–1999 (2003).

    Article  CAS  Google Scholar 

  13. Kulik, W. et al. Bloodspot assay using HPLC-tandem mass spectrometry for detection of Barth syndrome. Clin. Chem. 54, 371–378 (2008).

    Article  CAS  Google Scholar 

  14. Rana, P., Anson, B., Engle, S. & Will, Y. Characterization of human-induced pluripotent stem cell-derived cardiomyocytes: bioenergetics and utilization in safety screening. Toxicol. Sci. 130, 117–131 (2012).

    Article  CAS  Google Scholar 

  15. Karikó, K. et al. Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Mol. Ther. 16, 1833–1840 (2008).

    Article  Google Scholar 

  16. Zangi, L. et al. Modified mRNA directs the fate of heart progenitor cells and induces vascular regeneration after myocardial infarction. Nat. Biotechnol. 31, 898–907 (2013).

    Article  CAS  Google Scholar 

  17. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  Google Scholar 

  18. Hom, J.R. et al. The permeability transition pore controls cardiac mitochondrial maturation and myocyte differentiation. Dev. Cell 21, 469–478 (2011).

    Article  CAS  Google Scholar 

  19. Gerdes, A.M. & Capasso, J.M. Structural remodeling and mechanical dysfunction of cardiac myocytes in heart failure. J. Mol. Cell. Cardiol. 27, 849–856 (1995).

    Article  CAS  Google Scholar 

  20. Agarwal, A., Goss, J.A., Cho, A., McCain, M.L. & Parker, K.K. Microfluidic heart on a chip for higher throughput pharmacological studies. Lab Chip 13, 3599–3608 (2013).

    Article  CAS  Google Scholar 

  21. Feinberg, A.W. et al. Muscular thin films for building actuators and powering devices. Science 317, 1366–1370 (2007).

    Article  CAS  Google Scholar 

  22. Domian, I.J. et al. Generation of functional ventricular heart muscle from mouse ventricular progenitor cells. Science 326, 426–429 (2009).

    Article  CAS  Google Scholar 

  23. Parker, K.K., Tan, J., Chen, C.S. & Tung, L. Myofibrillar architecture in engineered cardiac myocytes. Circ. Res. 103, 340–342 (2008).

    Article  CAS  Google Scholar 

  24. Alford, P.W., Feinberg, A.W., Sheehy, S.P. & Parker, K.K. Biohybrid thin films for measuring contractility in engineered cardiovascular muscle. Biomaterials 31, 3613–3621 (2010).

    Article  CAS  Google Scholar 

  25. Malhotra, A. et al. Role of calcium-independent phospholipase A2 in the pathogenesis of Barth syndrome. Proc. Natl. Acad. Sci. USA 106, 2337–2341 (2009).

    Article  CAS  Google Scholar 

  26. Valianpour, F. et al. Linoleic acid supplementation of Barth syndrome fibroblasts restores cardiolipin levels: implications for treatment. J. Lipid Res. 44, 560–566 (2003).

    Article  CAS  Google Scholar 

  27. Rigaud, C. et al. Natural history of Barth syndrome: a national cohort study of 22 patients. Orphanet J. Rare Dis. 8, 70 (2013).

    Article  Google Scholar 

  28. Steinberg, S.F. Oxidative stress and sarcomeric proteins. Circ. Res. 112, 393–405 (2013).

    Article  CAS  Google Scholar 

  29. Sun, N. et al. Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. Sci. Transl. Med. 4, 130ra47 (2012).

    Article  Google Scholar 

  30. Lan, F. et al. Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell 12, 101–113 (2013).

    Article  CAS  Google Scholar 

  31. Park, I.H. et al. Disease-specific induced pluripotent stem cells. Cell 134, 877–886 (2008).

    Article  CAS  Google Scholar 

  32. Chan, E.M. et al. Live cell imaging distinguishes bona fide human iPS cells from partially reprogrammed cells. Nat. Biotechnol. 27, 1033–1037 (2009).

    Article  CAS  Google Scholar 

  33. Ichida, F. et al. Novel gene mutations in patients with left ventricular noncompaction or Barth syndrome. Circulation 103, 1256–1263 (2001).

    Article  CAS  Google Scholar 

  34. Whitman, G.J. et al. Diagnosis and therapeutic evaluation of a pediatric case of cardiomyopathy using phosphorus-31 nuclear magnetic resonance spectroscopy. J. Am. Coll. Cardiol. 5, 745–749 (1985).

    Article  CAS  Google Scholar 

  35. Chen, Y. & Dorn, G.W.n. PINK1-phosphorylated mitofusin 2 is a Parkin receptor for culling damaged mitochondria. Science 340, 471–475 (2013).

    Article  CAS  Google Scholar 

  36. He, A., Kong, S.W., Ma, Q. & Pu, W.T. Co-occupancy by multiple cardiac transcription factors identifies transcriptional enhancers active in heart. Proc. Natl. Acad. Sci. USA 108, 5632–5637 (2011).

    Article  CAS  Google Scholar 

  37. Houtkooper, R.H. et al. Cardiolipin and monolysocardiolipin analysis in fibroblasts, lymphocytes, and tissues using high-performance liquid chromatography-mass spectrometry as a diagnostic test for Barth syndrome. Anal. Biochem. 15, 230–237 (2009).

    Article  Google Scholar 

  38. Bray, M.A., Sheehy, S.P. & Parker, K.K. Sarcomere alignment is regulated by myocyte shape. Cell Motil. Cytoskeleton 65, 641–651 (2008).

    Article  Google Scholar 

  39. Chen, C.S., Mrksich, M., Huang, S., Whitesides, G.M. & Ingber, D.E. Geometric control of cell life and death. Science 276, 1425–1428 (1997).

    Article  CAS  Google Scholar 

  40. Sato, Y. et al. Three-dimensional multi-scale line filter for segmentation and visualization of curvilinear structures in medical images. Med. Image Anal. 2, 143–168 (1998).

    Article  CAS  Google Scholar 

  41. Melkman, A.A. On-line construction of the convex hull of a simple polyline. Inf. Process. Lett. 25, 11–12 (1987).

    Article  Google Scholar 

  42. Wei, S. et al. T-tubule remodeling during transition from hypertrophy to heart failure. Circ. Res. 107, 520–531 (2010).

    Article  CAS  Google Scholar 

  43. Timoshenko, S. & Woinowsky-Krieger, S. in Engineering Societies Monographs 5 (McGraw-Hill, 1959).

  44. Stoney, G.G. The tension of metallic films deposited by electrolysis. Proc. R. Soc. Lond. 82, 172–175 (1909).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work supported by the Barth Syndrome Foundation, the Boston Children's Hospital Translational Investigator Service, the US National Institutes of Health (NIH) NHLBI Progenitor Cell Biology Consortium (NIH U01 HL100401 and U01 HL100408), NIH RC1 HL099618, NIH UH2 TR000522 and charitable donations from E. Marram, K. Carpenter and G.F. Smith.

Author information

Authors and Affiliations

Authors

Contributions

G.W. designed and performed experiments and analyzed data. M.L.M. designed and performed experiments on MTFs and sarcomere organization and analyzed data. L.Y. and G.M.C. provided expert assistance and reagents for genome editing. F.S.P. designed the sarcomere organization analysis method. H.Y. developed the MTF analysis method. A.A. assisted with MTF substrate fabrication and experiments. D.J. provided advice on mitochondrial assays. D.Z. imaged iPSC-CMs to assess their mitochondrial organization and potential. L.Z. and K.R.C. provided expert assistance with modRNA, and J.C., J.D. and D.-Z.W. helped construct modRNAs. K.L. contributed to genome editing. R.J.A.W., W.K. and F.M.V. analyzed phospholipids. M.A.L. and C.E.M. provided expert assistance in iPSC differentiation to cardiomyocytes. A.H. developed TAZ shRNA viruses and provided technical assistance. J.G. and A.E.R. obtained patient samples. Q.M. assisted in teratoma analysis. J.W. contributed control iPSC lines. R.I.K. provided expert input, patient samples and 31P nuclear magnetic resonance data. K.K.P. and W.T.P. supervised the study. W.T.P. wrote the manuscript, and it was revised by K.K.P., G.W. and M.L.M.

Corresponding authors

Correspondence to Kevin Kit Parker or William T Pu.

Ethics declarations

Competing interests

J.W. is an employee of Allele Biotechnology & Pharmaceuticals.

Supplementary information

Supplementary Text and Figures

Supplementary Tables 1–4 and Supplementary Figures 1–14 (PDF 8956 kb)

Supplementary Data Set

Supplementary Data Sets (XLSX 82 kb)

Supplementary Movies

Supplementary Movies 1–6 (PDF 12265 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, G., McCain, M., Yang, L. et al. Modeling the mitochondrial cardiomyopathy of Barth syndrome with induced pluripotent stem cell and heart-on-chip technologies. Nat Med 20, 616–623 (2014). https://doi.org/10.1038/nm.3545

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3545

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing