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.

  • Letter
  • Published:

A HCN4+ cardiomyogenic progenitor derived from the first heart field and human pluripotent stem cells

Abstract

Most of the mammalian heart is formed from mesodermal progenitors in the first and second heart fields (FHF and SHF), whereby the FHF gives rise to the left ventricle and parts of the atria and the SHF to the right ventricle, outflow tract and parts of the atria1,2,3. Whereas SHF progenitors have been characterized in detail, using specific molecular markers4,5,6,7,8, comprehensive studies on the FHF have been hampered by the lack of exclusive markers. Here, we present Hcn4 (hyperpolarization-activated cyclic nucleotide-gated channel 4) as an FHF marker. Lineage-traced Hcn4+/FHF cells delineate FHF-derived structures in the heart and primarily contribute to cardiomyogenic cell lineages, thereby identifying an early cardiomyogenic progenitor pool. As a surface marker, HCN4 also allowed the isolation of cardiomyogenic Hcn4+/FHF progenitors from human embryonic stem cells. We conclude that a primary purpose of the FHF is to generate cardiac muscle and support the contractile activity of the primitive heart tube, whereas SHF-derived progenitors contribute to heart cell lineage diversification.

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: Hcn4 specifically marks the FHF at the earliest stages of mouse cardiogenesis.
Figure 2: Hcn4+/FHF lineage tracing in the embryonic heart.
Figure 3: Hcn4+/FHF lineage contribution to the heart.
Figure 4: Clonal analysis of single Hcn4+/FHF cells (derived) from mouse embryos.
Figure 5: Isolation and differentiation of HCN4+ cells from differentiated hESCs.

Similar content being viewed by others

References

  1. Vincent, S. D. & Buckingham, M. E. How to make a heart: the origin and regulation of cardiac progenitor cells. Curr. Top. Dev. Biol. 90, 1–41 (2010).

    Article  Google Scholar 

  2. Laugwitz, K-L., Moretti, A., Caron, L., Nakano, A. & Chien, K. R. Islet1 cardiovascular progenitors: a single source for heart lineages? Development 135, 193–205 (2008).

    Article  CAS  Google Scholar 

  3. Martin-Puig, S., Wang, Z. & Chien, K. R. Lives of a heart cell: tracing the origins of cardiac progenitors. Cell Stem Cell 2, 320–331 (2008).

    Article  CAS  Google Scholar 

  4. Cai, C-L. et al. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev. Cell 5, 877–889 (2003).

    Article  CAS  Google Scholar 

  5. Moretti, A. et al. Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification. Cell 127, 1151–1165 (2006).

    Article  CAS  Google Scholar 

  6. Laugwitz, K-L. et al. Postnatal isl1+ cardioblasts enter fully differentiated cardiomyocyte lineages. Nature 433, 647–653 (2005).

    Article  CAS  Google Scholar 

  7. Bu, L. et al. Human ISL1 heart progenitors generate diverse multipotent cardiovascular cell lineages. Nature 460, 113–117 (2009).

    Article  CAS  Google Scholar 

  8. Sun, Y. et al. Islet 1 is expressed in distinct cardiovascular lineages, including pacemaker and coronary vascular cells. Dev. Biol. 304, 286–296 (2007).

    Article  CAS  Google Scholar 

  9. Zhou, B. et al. Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature 454, 109–113 (2008).

    Article  CAS  Google Scholar 

  10. Cai, C-L. et al. A myocardial lineage derives from Tbx18 epicardial cells. Nature 454, 104–108 (2008).

    Article  CAS  Google Scholar 

  11. Katz, T. C. et al. Distinct compartments of the proepicardial organ give rise to coronary vascular endothelial cells. Dev. Cell 22, 639–650 (2012).

    Article  CAS  Google Scholar 

  12. Jiang, X., Rowitch, D. H., Soriano, P., McMahon, A. P. & Sucov, H. M. Fate of the mammalian cardiac neural crest. Development 127, 1607–1616 (2000).

    CAS  PubMed  Google Scholar 

  13. 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 

  14. Hornung, T. S. & Calder, L. Congenitally corrected transposition of the great arteries. Heart 96, 1154–1161 (2010).

    Article  Google Scholar 

  15. Winter, M. M. et al. Latest insights in therapeutic options for systemic right ventricular failure: a comparison with left ventricular failure. Heart 95, 960–963 (2009).

    Article  CAS  Google Scholar 

  16. Herrmann, S., Layh, B. & Ludwig, A. Novel insights into the distribution of cardiac HCN channels: an expression study in the mouse heart. J. Mol. Cell. Cardiol. 51, 997–1006 (2011).

    Article  CAS  Google Scholar 

  17. Vicente-Steijn, R. et al. Funny current channel HCN4 delineates the developing cardiac conduction system in chicken heart. Heart Rhythm 8, 1254–1263 (2011).

    Article  Google Scholar 

  18. Garcia-Frigola, C., Shi, Y. & Evans, S. M. Expression of the hyperpolarization-activated cyclic nucleotide-gated cation channel HCN4 during mouse heart development. Gene Exp. 3, 777–783 (2003).

    Article  CAS  Google Scholar 

  19. Stanley, E. G. et al. Efficient Cre-mediated deletion in cardiac progenitor cells conferred by a 3’UTR-ires-Cre allele of the homeobox gene Nkx2-5. Int. J. Dev. Biol. 46, 431–439 (2002).

    CAS  PubMed  Google Scholar 

  20. Bruneau, B. G. et al. Chamber-specific cardiac expression of Tbx5 and heart defects in Holt-Oram syndrome. Dev. Biol. 211, 100–108 (1999).

    Article  CAS  Google Scholar 

  21. Hoesl, E. et al. Tamoxifen-inducible gene deletion in the cardiac conduction system. J. Mol. Cell. Cardiol. 45, 62–69 (2008).

    Article  CAS  Google Scholar 

  22. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  Google Scholar 

  23. Verzi, M. P., McCulley, D. J., De Val, S., Dodou, E. & Black, B. L. The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field. Dev. Biol. 287, 134–145 (2005).

    Article  CAS  Google Scholar 

  24. Qyang, Y. et al. The renewal and differentiation of Isl1+ cardiovascularprogenitors are controlled by a Wnt/beta-catenin pathway. Cell Stem Cell 1, 165–179 (2007).

    Article  CAS  Google Scholar 

  25. Soriano, P. Generalized lacZ expression with the ROSA26 Cre reporter strain. Nat. Genet. 21, 70–71 (1999).

    Article  CAS  Google Scholar 

  26. Bressan, M., Liu, G. & Mikawa, T. Early mesodermal cues assign avian cardiac pacemaker fate potential in a tertiary heart field. Science 428, 821–827 (2013).

    Google Scholar 

  27. Mommersteeg, M. T. M. et al. The sinus venosus progenitors separate and diversify from the first and second heart fields early in development. Cardiovasc. Res. 87, 92–101 (2010).

    Article  CAS  Google Scholar 

  28. Miquerol, L. et al. Biphasic development of the mammalian ventricular conduction system. Circ. Res. 107, 153–161 (2010).

    Article  CAS  Google Scholar 

  29. Fijnvandraat, A. C. et al. Cardiomyocytes derived from embryonic stem cells resemble cardiomyocytes of the embryonic heart tube. Cardiovasc. Res. 58, 399–409 (2003).

    Article  CAS  Google Scholar 

  30. Milgrom-Hoffman, M. et al. The heart endocardium is derived from vascular endothelial progenitors. Development 138, 4777–4787 (2011).

    Article  CAS  Google Scholar 

  31. Lui, K. O. et al. Driving vascular endothelial cell fate of human multipotent Isl1+ heart progenitors with VEGF modified mRNA. Cell Res.http://dx.doi.org/10.1038/cr.2013.112 (2013).

  32. Prall, O. W. J. et al. An Nkx2-5/Bmp2/Smad1 negative feedback loop controls heart progenitor specification and proliferation. Cell 128, 947–959 (2007).

    Article  CAS  Google Scholar 

  33. Ma, Q., Zhou, B. & Pu, W. T. Reassessment of Isl1 and Nkx2-5 cardiac fate maps using a Gata4-based reporter of Cre activity. Dev. Biol. 323, 98–104 (2008).

    Article  CAS  Google Scholar 

  34. Xie, L. et al. Tbx5-hedgehog molecular networks are essential in the second heart field for atrial septation. Dev. Cell 23, 280–291 (2012).

    Article  CAS  Google Scholar 

  35. Watanabe, Y. & Buckingham, M. The formation of the embryonic mouse heart: heart fields and myocardial cell lineages. Ann. NY Acad. Sci. 1188, 15–24 (2010).

    Article  Google Scholar 

  36. Aanhaanen, W. T. J. et al. The Tbx2 primary myocardium of the atrioventricular canal forms the atrioventricular node and the base of the left ventricle. Circ. Res. 104, 1267–1274 (2009).

    Article  CAS  Google Scholar 

  37. Buckingham, M. E. & Meilhac, S. M. Tracing cells for tracking cell lineage and clonal behavior. Dev. Cell 21, 394–409 (2011).

    Article  CAS  Google Scholar 

  38. Brent, A. E., Schweitzer, R. & Tabin, C. J. A somitic compartment of tendon progenitors. Cell 113, 235–248 (2003).

    Article  CAS  Google Scholar 

  39. Dietrich, S., Schubert, F. R. & Lumsden, A control of dorsoventral pattern in the chick paraxial mesoderm. Development 124, 3895–3908 (1997).

    CAS  Google Scholar 

  40. Fujimori, S. et al. Wnt/β-catenin signaling in the dental mesenchymeregulates incisor development by regulating Bmp4. Dev. Biol. 348, 97–106 (2010).

    Article  CAS  Google Scholar 

  41. El-Badawi, A. & Schenk, E. A. Histochemical methods for separate, consecutive and simultaneous demonstration of acetylcholinesterase and norepinephrine in cryostat sections. J. Histochem. Cytochem. 15, 580–588 (1967).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank C. Cowan for his support, L. Prickett-Rice, K. Folz-Donahue and M. Weglarz of the Harvard Stem Cell Institute Flow Cytometry Core Facility for assistance with FACS analysis, C. Du of the Tufts Electrophysiology Core for assistance with electrophysiology recordings, L. Bu for technical advice, K. Buac, E. Hansson, C. Riedel and L. Bu for critical reading of the manuscript and discussions, and C. Hartmann for advice on double-fluorescence in situ hybridizations. D.S. has received a D.F.G. (German Research Foundation) postdoctoral fellowship. K.B. was supported by a NHLBI T32HL007208 grant. This work is financially supported by the NIH U01 HL098 166 and NIH U01H100408 research grants.

Author information

Authors and Affiliations

Authors

Contributions

D.S. and K.R.C. designed the experiments and wrote the manuscript. D.S. carried out most of the experiments and analysed most of the data. M.K.A. helped to perform and analyse clonal analysis experiments. K.B. contributed PCRs with reverse transcription of in vivo clonal analysis. L.Z. performed some immunohistochemical staining, M.W.S. helped with some hESC differentiation assays and RNA isolations, J.C. performed acetylcholinesterase staining, M.S. helped with some mESC clonal analysis cultures and A.L. provided the Hcn4CreErt2 mouse line.

Corresponding author

Correspondence to Kenneth R. Chien.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Section in situ hybridisation analysis of crescent stage mouse embryos (E7.5).

(a) More anterior section level of the same embryo as shown in Fig. 1h ( 4ss). Expression domains of Hcn4, Tbx5 and Mlc2a (all shown in green) are predominantly overlapping, but not with that of Isl1(shown in red). (b) Corresponding section levels through an E7.5 embryo as shown below in (c+d). (c-d) Double in situ hybridisations. Blue staining: NBT/BCIP (BM Purple) and brown staining: INT/BCIP. (c) Hcn4 (blue) and Isl1 (brown) are expressed in predominantly non-overlapping expression domains. (d) Tbx5 (blue) and Isl1 (brown) are expressed in predominantly non-overlapping expression domains. Expression domains of Hcn4 and Tbx5 are mostly overlapping, when comparing their expression on adjacent sections. (e) Single fluorescence in situ hybridisations on adjacent sections of an E7.5 ( 4ss) embryo. (e-f) Expression of Mlc2a, Hcn4 and Tbx5 (all in green) are predominantly expressed within the presumptive FHF, in contrary to Isl1 expression primarily in the SHF (red). (e’-f’) Section levels corresponding to (e) and (f), respectively. Scale bar: 50μm.

Supplementary Figure 2 Section in situ hybridisation and IHC analysis of a crescent stage embryo (E7.5).

(a) Single in situ hybridisations (NBT/BCIP, BM Purple) on adjacent sections of an E7.5 ( 2–4ss) mouse embryo. Corresponding section levels are shown in panel on left side. Expression domains of Hcn4 and Tbx5 are mostly overlapping in presumptive FHF, but not with Isl1. However, in the posterior area of the FHF and SHF domains there is an overlapping expression domain between Tbx5 and Isl1 visible (orange arrows), but not with Hcn4 (black arrow), suggesting that Hcn4 is more exclusive to the FHF than Tbx5. (b) Co-staining showing single fluorescent in situ hybridisation of Tbx5(green) and ISL1 protein location (red) on adjacent sections of an E7.5 ( 2–4ss) mouse embryo. The two domains are largely non-overlapping; however, there is some co-localization visible in the most lateral sides of the splanchnic mesoderm (arrows). There are also some areas, where there is weak staining for ISL1 protein visible (asterix), overlapping with Tbx5 mRNA, but the ISL1 protein does not co-localize with the nucleus but seems to be present within the cytoplasma, indicating that it may not be functional in these areas. Scale bar: 50μm.

Supplementary Figure 3 Section in situ hybridisation analysis of an early heart tube stage embryo (E8.0).

(a) Single colour in situ hybridisations on adjacent sections. Corresponding section level as shown in drawing. Hcn4, Tbx5, Mlc2a and Nkx2.5 are expressed within the primitive heart tube, unlike Isl1, which is expressed in the pharyngeal mesoderm. At this developmental stage, Nkx2.5 is mostly non-overlapping with Isl1 expression. (b) Single fluorescent in situ hybridisations on adjacent sections. Corresponding section levels are shown in drawings above images. Expression domains of Hcn4, Tbx5 and Mlc2a (all green) are mostly overlapping, but predominantly non-overlapping with Isl1 expression (red). However, posterior to the primitive heart tube, there is an area of overlap between Tbx5 and Isl1 expression (arrow), but not with Hcn4 and Mlc2a (most likely no overlap with Isl1, as the section level is a bit more anterior compared to Isl1 (adjacent sections) and it seems more lateral compared to Isl1). Scale bar: 50μm.

Supplementary Figure 4 Hcn4+/FHF lineage tracing in the embryonic heart and Hcn4+/ FHF cell lineage contribution to atria.

(a) Whole-mount X-gal staining of Hcn4CreErt2/+;R26RlacZ hearts at E19.5. Tamoxifen induced Hcn4CreErt2/+ activation at E8.0 labels the Hcn4+/FHF derivatives, such as the left ventricle (LV) and parts of the atria. At this stage, also the sinus venosus (SV) and sinoatrial node (SAN) are stained positive. (b) Section through an E19.5 Hcn4CreErt2/+;R26ReYFP heart; Hcn4CreErt2/+ activation by tamoxifen at E7.0. Within the left ventricle, there is Hcn4+/FHF lineage contribution to the trabecular and compact layer of the left ventricle, and some minor contribution to the septum. (c-d) Whole-mount X-gal staining of a Hcn4CreErt2/+;R26RlacZ heart at E12.5 (c), and embryos at E9.5 (d), and E10.5 (e). Tamoxifen induced Hcn4CreErt2/+ activation at E7.0 labels the Hcn4+/FHF derivatives, such as the left ventricle (LV) and parts of the atria. No staining is visible within the pharyngeal mesoderm (arrows, d+e). In situ hybridisation for Isl1 of E9.5 embryo marks the pharyngeal mesoderm (arrow, f). Hcn4+/FHF cells also contribute to the AV canal (red asterix, d+e). (g) Section through an E19.5 atria of Hcn4CreErt2/+;R26ReYFP mice; Hcn4CreErt2/+ activation by tamoxifen at E7.0. Co-immunostaining for YFP as a marker for Hcn4+/FHF lineage traced cells and TNNT2 shows contribution to cardiomyocytes. Yellow box represents magnified area shown in merge image (h) FACS quantification of the cellular contribution of Hcn4+/FHF lineage traced cells to an Hcn4CreErt2/+;R26RtdTomato E19.5 atria (Tamoxifen E7.0). FACS analysis of tomato+ Hcn4+/FHF lineage traced cells co-stained for TNNT2, PECAM-1 (CD31) or smMHC (MYH11) showed a predominant contribution to cardiomyocytes, but no double positive cells for endothelial/endocardial-, and vascular smooth muscle markers, suggesting no contribution to these cell lineages within atria. Representative FACS dot-plots are shown. Secondary antibody controls are shown in Supplementary Fig. S6b. Scale bar: (a+c) 0.5mm, (b) 100μm, (d-f) 0.2mm, (g) 50μm.

Supplementary Figure 5 C lonal analysis of Hcn4+/FHF cells isolated from mouse ESCs and TgMef2c-AHF-eGFP/SHF cell lineage contribution in vivo.

(a) Embryonic stem cells (ECSs) were derived from Hcn4CreErt2/+;R26ReYFP mice. Mouse ESCs were differentiated using the embryoid body technique. Hcn4CreErt2/+ activity was induced by administration of 4-OH-Tamoxifen on day 4 and 6 of differentiation. eYFP positive Hcn4+/FHF cells were FACS sorted (representative FACS dot-plots are shown). (b) Single eYFP positive Hcn4+/FHF cells were sorted as 1 cell/well into 96-well plates. After 2-3 weeks single cell derived clones showed limited expansion and efficient differentiation into cardiomyogenic clones, visualized by direct fluorescence of eYFP and immunostaining for the cardiac muscle marker TNNT2 (cTnT). (c) RT-PCR analysis, showing expression of cardiac marker genes in single Hcn4+/FHF cell derived cardiomyogenic clones. (d) Single cell patch clamp analysis (n = 39). Most clamped cells showed a ventricular-like action potential (AP) (64%), while some showed a more atrial-like AP (16%), or nodal-like AP (20%). Representative action potentials are shown. (e) FACS quantification of the cellular contribution of TgMef2c-AHF-eGFP/SHF lineage traced cells to the right ventricle and outflowtract (OFT). Fixed single cells were co-stained for TNNT2, demonstrating contribution to cardiomyogenic cell lineages, particularly within the RV. Co-staining for smMHC (MYH11) showed a minor contribution to vascular smooth muscle cells in RV and contribution in the OFT. Co-staining for PECAM-1 (CD31) showed a minor contribution to endothelial cell lineage in RV and contribution in the OFT. Representative FACS dot-plots are shown. Scale bar: 50μm.

Supplementary Figure 6 Uncropped key electrophoresis data and secondary antibody controls of FACS analyses.

(a) Uncropped electrophoresis data of RT-PCR shown in Fig. 4c. (b) Background signal of secondary antibodies used for FACS sorting as shown in Fig. 3f and Supplementary Fig. S4h.

Supplementary Table 1 Forward and reverse PCR primer sequences utilised for generation of RNA probes used in whole-mount in situ hybridisation.
Supplementary Table 2 Forward and reverse PCR primer sequences used for clonal analysis from mouse embryos and mouse ESCs, and qPCR analysis of human HCN4+/FHF cells.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1469 kb)

Supplementary Table 1

Supplementary Information (XLSX 10 kb)

Supplementary Table 2

Supplementary Information (XLSX 11 kb)

Clonal analysis of single Hcn4+/FHF cells isolated from mouse embryos.

Representative example of a beating cardiomyogenic clone derived from a single Hcn4+/FHF cell from mouse embryos. (AVI 7234 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Später, D., Abramczuk, M., Buac, K. et al. A HCN4+ cardiomyogenic progenitor derived from the first heart field and human pluripotent stem cells. Nat Cell Biol 15, 1098–1106 (2013). https://doi.org/10.1038/ncb2824

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2824

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