Skip to main content
Log in

A CD63+ve/c-kit+ve stem cell population isolated from the mouse heart

  • Published:
Molecular and Cellular Biochemistry Aims and scope Submit manuscript

Abstract

Cardiac cell regeneration from endogenous cardiac stem cells (CSCs) following MI is rather low. Therefore, identifying mechanisms to boost endogenous CSC activation and participation in cardiac repair appears to be the most promising strategy for MI patients. We previously engineered tissue inhibitor of metalloproteinases-1 (TIMP-1) overexpressing embryonic stem (ES-TIMP-1) cells and transplanted them into the infarcted murine heart. Collected data demonstrated that TIMP-1 enhanced transplanted ES cell engraftment, survival and differentiation into cardiac myocytes post-transplantation. Therefore, we postulated that there may be a new stem cell population present in the heart that is regulated by extracellular protein TIMP-1. Furthermore, we hypothesized that this cell population has a potential for cell proliferation and differentiation into cardiac cell types. Therefore, we isolated CSCs from 4 weeks old C57BL/6 mice and cultured them in vitro in presence of ESCM, ES-TIMP-1-CM or TIMP-1. Our immunostaining data demonstrated the existence of a novel CSC subpopulation, CD63+ve/c-kit+ve. When treated with TIMP-1, these cells showed significantly (p < 0.05) increased proliferation rates compared to control cells, enhanced TIMP-1 receptor (CD63), along with improved expression of phospho and total β-catenin proteins as demonstrated by Western blot analysis. Next, we demonstrate significantly (p < 0.05) improved cardiac myocyte, vascular smooth muscle cell, and endothelial cell differentiation. Furthermore, our RT-PCR data shows increase in cardiac gene (GATA-4, Mef2C, and Nkx-2.5) expression when compared to ESCM and control cells. Collectively, these data, for the first time, establish the existence of a new CD63+ve/c-kit+ve CSC subpopulation that has a significant potential for proliferation and differentiation into cardiac cell types once stimulated with TIMP-1.

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.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

References

  1. Abdelli LS, Merino H, Rocher CM et al (2012) Cell therapy in the heart. Can J Physiol Pharmacol 90:307–315

    Article  CAS  PubMed  Google Scholar 

  2. Anversa P, Leri A, Kajstura J (2006) Cardiac regeneration. J Am Coll Cardiol 47:1769–1776

    Article  PubMed  Google Scholar 

  3. Bergmann O, Bhardwaj RD, Bernard S et al (2009) Evidence for cardiomyocyte renewal in humans. Science 324:98–102

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  4. Dawn B, Stein AB, Urbanek K et al (2005) Cardiac stem cells delivered intravascularly traverse the vessel barrier, regenerate infarcted myocardium, and improve cardiac function. Proc Natl Acad Sci USA 102:3766–3771

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Singla DK, Singla RD, McDonald DE (2008) Factors released from embryonic stem cells inhibit apoptosis in H9c2 cells through PI3K/Akt but not ERK pathway. Am J Physiol Heart Circ Physiol 295:H907–H913

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. Koudstaal S, Jansen Of Lorkeers SJ, Gaetani R et al (2013) Concise review: heart regeneration and the role of cardiac stem cells. Stem Cells Transl Med 2:434–443

    Article  PubMed Central  PubMed  Google Scholar 

  7. Rajala K, Pekkanen-Mattila M, Alto-Setala K (2011) Cardiac differentiation of pluripotent stem cells. Stem Cells Int 2011:383709

    Article  PubMed Central  PubMed  Google Scholar 

  8. Glass C, Singla DK (2012) Overexpression of TIMP-1 in embryonic stem cells attenuates adverse cardiac remodeling following myocardial infarction. Cell Transpl 21:1931–1944

    Article  Google Scholar 

  9. Yan B, Abdelli LS, Singla DK (2011) Transplanted induced pluripotent stem cells improve cardiac function and induce neovascularization in the infarcted hearts of db/db mice. Mol Pharm 8:1602–1610

    Article  CAS  PubMed  Google Scholar 

  10. Merino H, Singla DK (2014) Notch-1 mediated cardiac protection following embryonic and induced pluripotent stem cell transplantation in doxorubicin-induced heart failure. PLoS One 9:e101024

    Article  PubMed Central  PubMed  Google Scholar 

  11. Long X, Singla DK (2013) Inactivation of Klf5 by zinc finger nuclease downregulates expression of pluripotent genes and attenuates colony formation in embryonic stem cells. Mol Cell Biochem 382:113–119

    Article  CAS  PubMed  Google Scholar 

  12. Leri A, Kajstura J, Anversa P (2005) Cardiac stem cells and mechanisms of myocardial regeneration. Physiol Rev 85:1373–1416

    Article  CAS  PubMed  Google Scholar 

  13. Kopitz C, Gerg M, Bandapalli OR et al (2007) Tissue inhibitor of metalloproteinases-1 promotes liver metastasis by induction of hepatocyte growth factor signaling. Cancer Res 67:8615–8623

    Article  CAS  PubMed  Google Scholar 

  14. Egea V, Zahler S, Rieth N et al (2012) Tissue inhibitor of metalloproteinase-1 (TIMP-1) regulates mesenchymal stem cells through let-7f microRNA and Wnt/beta-catenin signaling. Proc Natl Acad Sci USA 109:E309–E316

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  15. Jung KK, Liu XW, Chirco R et al (2006) Identification of CD63 as a tissue inhibitor of metalloproteinase-1 interacting cell surface protein. EMBO J 25:3934–3942

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  16. Singla DK, Singla RD, Lamm S et al (2011) TGF-beta2 treatment enhances cytoprotective factors released from embryonic stem cells and inhibits apoptosis in infarcted myocardium. Am J Physiol Heart Circ Physiol 300:H1442–H1450

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  17. Moore CS, Milner R, Nishiyama A et al (2011) Astrocytic tissue inhibitor of metalloproteinase-1 (TIMP-1) promotes oligodendrocyte differentiation and enhances CNS myelination. J Neurosci 31:6247–6254

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Collot-Teixeira S, Barbatis C, Bultelle F et al (2008) CD36 is significantly correlated with adipophilin in human carotid lesions and inversely correlated with plasma ApoAI. J Biomed Biotechnol 2008:813236

    Article  PubMed Central  PubMed  Google Scholar 

  19. Doyle EL, Ridger V, Ferraro F et al (2011) CD63 is an essential cofactor to leukocyte recruitment by endothelial P-selectin. Blood 118:4265–4273

    Article  CAS  PubMed  Google Scholar 

  20. Dodou E, Xu SM, Black BL (2003) mef2c is activated directly by myogenic basic helix-loop-helix proteins during skeletal muscle development in vivo. Mech Dev 120:1021–1032

    Article  CAS  PubMed  Google Scholar 

  21. Durocher D, Charron F, Warren R et al (1997) The cardiac transcription factors Nk2–5 and GATA-4 are mutual cofactors. EMBO J 16:5687–5696

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgments

The authors are thankful to Reetu Singla and Jing Wang for their technical assistance in the paper.

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical approval

All applicable international, national, and/or institutional guidelines for the care and use of animals were followed. All procedures performed in studies involving animals were in accordance with the University of Central Florida (UCF) Institutional Animal Care and Use Committee (IACUC) ethical standards. This article does not contain any studies with human participants performed by any of the authors.

Funding

This work was supported, in part, by Grants from the National Institute of Health [1R01HL090646-01, and 5R01HL094467-02 to DKS].

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dinender K. Singla.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Abdelli, L.S., Singla, D.K. A CD63+ve/c-kit+ve stem cell population isolated from the mouse heart. Mol Cell Biochem 406, 101–109 (2015). https://doi.org/10.1007/s11010-015-2428-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11010-015-2428-9

Keywords

Navigation