Skip to main content

Advertisement

Log in

Cyclic Strain Promotes H19 Expression and Vascular Tube Formation in iPSC-Derived Endothelial Cells

  • Original Article
  • Published:
Cellular and Molecular Bioengineering Aims and scope Submit manuscript

Abstract

Introduction

Induced pluripotent stem cell (iPSC)-derived endothelial cells (ECs) have the potential for therapeutic application in several cardiovascular diseases. Mechanical strain is known to regulate EC behavior and stem cell differentiation and may play a role in directing EC differentiation of iPSCs. H19, a long non-coding RNA (lncRNA), is known to affect ECs in several mechanically relevant pathologies and may play a role in this process as well. Therefore, we investigated expression changes of H19 resulting from mechanical stimulation during EC differentiation, as well as functional effects on EC tube formation.

Methods

iPSCs were subjected to 5% cyclic mechanical strain during EC differentiation. RT-PCR and flow cytometry were used to assess changes in mesoderm differentiation and gene expression in the final ECs as a result of strain. Functional outcomes of mechanically differentiated ECs were assessed with a tube formation assay and changes in H19. H19 was also overexpressed in human umbilical vein endothelial cells (HUVECs) to assess its role in non-H19-expressing ECs.

Results

Mechanical strain promoted mesoderm differentiation, marked by increased expression of brachyury 24 h after initiation of differentiation. Strain also increased expression of H19, CD31, VE-cadherin, and VEGFR2 in differentiated ECs. Strain-differentiated ECs formed tube networks with higher junction and endpoint density than statically-differentiated ECs. Overexpression of H19 in HUVECs resulted in similar patterns of tube formation.

Conclusions

H19 expression is increased by mechanical strain and promotes tube branching in iPSC-derived ECs.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Figure 1
Figure 2
Figure 3
Figure 4

Similar content being viewed by others

References

  1. Ahsan, T., and R. M. Nerem. Fluid shear stress promotes an endothelial-like phenotype during the early differentiation of embryonic stem cells. Tissue Eng. Part A 16:3547–3553, 2010.

    Article  Google Scholar 

  2. Cai, J., E. Pardali, G. Sánchez-Duffhues, and P. ten Dijke. BMP signaling in vascular diseases. FEBS Lett. 586:1993–2002, 2012.

    Article  Google Scholar 

  3. Dejana, E. The role of wnt signaling in physiological and pathological angiogenesis. Circ. Res. 107:943–952, 2010.

    Article  Google Scholar 

  4. Dyer, L. A., X. Pi, and C. Patterson. The role of BMPs in endothelial cell function and dysfunction. Trends Endocrinol. Metab. 25:472–480, 2014.

    Article  Google Scholar 

  5. Engler, A. J., S. Sen, H. L. Sweeney, and D. E. Discher. Matrix elasticity directs stem cell lineage specification. Cell 126:677–689, 2006.

    Article  Google Scholar 

  6. Gabory, A., H. Jammes, and L. Dandolo. The H19 locus: role of an imprinted non-coding RNA in growth and development. BioEssays 32:473–480, 2010.

    Article  Google Scholar 

  7. Gridley, T. Notch signaling in vascular development and physiology. Development 134:2709–2718, 2007.

    Article  Google Scholar 

  8. Hadji, F., et al. Altered DNA methylation of long noncoding RNA H19 in calcific aortic valve disease promotes mineralization by silencing NOTCH1. Circulation 134:1848–1862, 2016.

    Article  Google Scholar 

  9. Han, D. K., Z. Z. Khaing, R. A. Pollock, C. C. Haudenschild, and G. Liau. H19, a marker of developmental transition, is reexpressed in human atherosclerotic plaques and is regulated by the insulin family of growth factors in cultured rabbit smooth muscle cells. J. Clin. Invest. 97:1276–1285, 1996.

    Article  Google Scholar 

  10. Hofmann, P., et al. Long non-coding RNA H19 regulates endothelial cell aging via inhibition of STAT3 signalling. Cardiovasc. Res. 115:230–242, 2019.

    Article  Google Scholar 

  11. James, D., et al. Expansion and maintenance of human embryonic stem cell – derived endothelial cells by TGFb inhibition is Id1 dependent. Nat. Biotechnol. 28:1–7, 2010.

    Article  Google Scholar 

  12. Jia, P., et al. Long non-coding RNA H19 regulates glioma angiogenesis and the biological behavior of glioma-associated endothelial cells by inhibiting microRNA-29a. Cancer Lett. 381:359–369, 2016.

    Article  Google Scholar 

  13. Kumar, A., J. K. Placone, and A. J. Engler. Understanding the extracellular forces that determine cell fate and maintenance. Development 144:4261–4270, 2017.

    Article  Google Scholar 

  14. Li, B., et al. LncRNA-H19 modulates Wnt/β-catenin signaling by targeting Dkk4 in hindlimb unloaded rat. Orthop. Surg. 2017. https://doi.org/10.1111/os.12321.

    Article  Google Scholar 

  15. Lian, X., et al. Efficient differentiation of human pluripotent stem cells to endothelial progenitors via small-molecule activation of WNT signaling. Stem Cell Rep. 3:804–816, 2014.

    Article  Google Scholar 

  16. Liang, W.-C., et al. H19 activates Wnt signaling and promotes osteoblast differentiation by functioning as a competing endogenous RNA. Sci. Rep. 6:20121, 2016.

    Article  Google Scholar 

  17. Liao, J., et al. lncRNA H19 mediates BMP9-induced osteogenic differentiation of mesenchymal stem cells (MSCs) through Notch signaling. Oncotarget 8:53581–53601, 2017.

    Article  Google Scholar 

  18. Lin, Y., C.-H. Gil, and M. C. Yoder. Differentiation, evaluation, and application of human induced pluripotent stem cell-derived endothelial cells. Arterioscler. Thromb. Vasc. Biol. 37:2014–2025, 2017.

    Article  Google Scholar 

  19. Liu, L., et al. The H19 long noncoding RNA is a novel negative regulator of cardiomyocyte hypertrophy. Cardiovasc. Res. 111:56–65, 2016.

    Article  Google Scholar 

  20. Lu, D., and G. S. Kassab. Role of shear stress and stretch in vascular mechanobiology. J. R. Soc. Interface 8:1379–1385, 2011.

    Article  Google Scholar 

  21. MacGrogan, D., J. Münch, and J. L. de la Pompa. Notch and interacting signalling pathways in cardiac development, disease, and regeneration. Nat. Rev. Cardiol. 15:685–704, 2018.

    Article  Google Scholar 

  22. Pahnke, A., et al. The role of Wnt regulation in heart development, cardiac repair and disease: a tissue engineering perspective. Biochem. Biophys. Res. Commun. 473:698–703, 2016.

    Article  Google Scholar 

  23. Patsch, C., et al. Generation of vascular endothelial and smooth muscle cells from human pluripotent stem cells. Nat. Cell Biol. 17:994–1003, 2015.

    Article  Google Scholar 

  24. Rajendran, P., et al. The vascular endothelium and human diseases. Int. J. Biol. Sci. 9:1057–1069, 2013.

    Article  Google Scholar 

  25. Resnick, N., et al. Fluid shear stress and the vascular endothelium: for better and for worse. Prog. Biophys. Mol. Biol. 81:177–199, 2003.

    Article  Google Scholar 

  26. Saha, S., L. Ji, J. J. de Pablo, and S. P. Palecek. Inhibition of human embryonic stem cell differentiation by mechanical strain. J. Cell. Physiol. 206:126–137, 2006.

    Article  Google Scholar 

  27. Sharifpanah, F., S. Behr, M. Wartenberg, and H. Sauer. Mechanical strain stimulates vasculogenesis and expression of angiogenesis guidance molecules of embryonic stem cells through elevation of intracellular calcium, reactive oxygen species and nitric oxide generation. Biochim. Biophys. Acta. 1863:3096–3105, 2016.

    Article  Google Scholar 

  28. Shimizu, N., et al. Cyclic strain induces mouse embryonic stem cell differentiation into vascular smooth muscle cells by activating PDGF receptor β. J. Appl. Physiol. 104:766–772, 2008.

    Article  Google Scholar 

  29. Thorvaldsen, J. L., A. M. Fedoriw, S. Nguyen, and M. S. Bartolomei. Developmental profile of H19 differentially methylated domain (DMD) deletion alleles reveals multiple roles of the DMD in regulating allelic expression and DNA methylation at the imprinted H19/Igf2 locus. Mol. Cell. Biol. 26:1245–1258, 2006.

    Article  Google Scholar 

  30. Wu, J., et al. Long non-coding RNA H19 mediates mechanical tension-induced osteogenesis of bone marrow mesenchymal stem cells via FAK by sponging miR-138. Bone 108:62–70, 2018.

    Article  Google Scholar 

  31. Yamamoto, K., et al. Fluid shear stress induces differentiation of Flk-1-positive embryonic stem cells into vascular endothelial cells in vitro. Am. J. Physiol. Circ. Physiol. 288:H1915–H1924, 2005.

    Article  Google Scholar 

  32. Zhu, A., L. Chu, Q. Ma, and Y. Li. Long non-coding RNA H19 down-regulates miR-181a to facilitate endothelial angiogenic function. Artif. Cells Nanomed. Biotechnol. 47:2698–2705, 2019.

    Article  Google Scholar 

  33. Zudaire, E., L. Gambardella, C. Kurcz, and S. Vermeren. A computational tool for quantitative analysis of vascular networks. PLoS ONE 6:e27385, 2011.

    Article  Google Scholar 

Download references

Funding

This work was supported by the National Heart, Lung, and Blood Institute (HL135790 and HL007411) and the Fondation Leducq.

Conflict of interest

No conflict of interest, financial or otherwise, are declared by Dr. Vander Roest or Dr. Merryman.

Ethical Approval

This article does not contain any studies with human participants or animals performed by any of the authors

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to W. David Merryman.

Additional information

Associate Editor Michael R. King oversaw the review of this article.

Publisher's Note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vander Roest, M.J., Merryman, W.D. Cyclic Strain Promotes H19 Expression and Vascular Tube Formation in iPSC-Derived Endothelial Cells. Cel. Mol. Bioeng. 13, 369–377 (2020). https://doi.org/10.1007/s12195-020-00617-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12195-020-00617-0

Keywords

Navigation