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.

Epicardial FSTL1 reconstitution regenerates the adult mammalian heart

This article has been updated

Abstract

The elucidation of factors that activate the regeneration of the adult mammalian heart is of major scientific and therapeutic importance. Here we found that epicardial cells contain a potent cardiogenic activity identified as follistatin-like 1 (Fstl1). Epicardial Fstl1 declines following myocardial infarction and is replaced by myocardial expression. Myocardial Fstl1 does not promote regeneration, either basally or upon transgenic overexpression. Application of the human Fstl1 protein (FSTL1) via an epicardial patch stimulates cell cycle entry and division of pre-existing cardiomyocytes, improving cardiac function and survival in mouse and swine models of myocardial infarction. The data suggest that the loss of epicardial FSTL1 is a maladaptive response to injury, and that its restoration would be an effective way to reverse myocardial death and remodelling following myocardial infarction in humans.

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: Epicardial secretome has cardiogenic activity, and improves cardiac function after MI via embryonic epicardium-like patches.
Figure 2: Fstl1 is an epicardial cardiogenic factor with dynamic expression after ischaemic injury.
Figure 3: FSTL1 recapitulates the in vivo restorative effect of epicardial conditioned media in the engineered epicardial patch, and promotes cardiomyocyte proliferation.
Figure 4: FSTL1 proliferative activity on early cardiomyocytes depends on the cells' selective post-transcriptional FSTL1 modifications.
Figure 5: Epicardial FSTL1 delivery activates cardiac regeneration in preclinical model of ischaemic heart injury.

Similar content being viewed by others

Change history

  • 23 September 2015

    Reference 11 was updated.

References

  1. van Wijk, B., Gunst, Q. D., Moorman, A. F. & van den Hoff, M. J. Cardiac regeneration from activated epicardium. PLoS ONE 7, e44692 (2012)

    Article  ADS  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  3. Lavine, K. J. & Ornitz, D. M. Rebuilding the coronary vasculature: hedgehog as a new candidate for pharmacologic revascularization. Trends Cardiovasc. Med. 17, 77–83 (2007)

    Article  CAS  Google Scholar 

  4. Brade, T. et al. Retinoic acid stimulates myocardial expansion by induction of hepatic erythropoietin which activates epicardial Igf2. Development 138, 139–148 (2011)

    Article  CAS  Google Scholar 

  5. Mellgren, A. M. et al. Platelet-derived growth factor receptor beta signaling is required for efficient epicardial cell migration and development of two distinct coronary vascular smooth muscle cell populations. Circ. Res. 103, 1393–1401 (2008)

    Article  CAS  Google Scholar 

  6. Smart, N. et al. Myocardial regeneration: expanding the repertoire of thymosin beta4 in the ischemic heart. Ann. NY Acad. Sci. 1269, 92–101 (2012)

    Article  ADS  CAS  Google Scholar 

  7. Kikuchi, K. et al. tcf21+ epicardial cells adopt non-myocardial fates during zebrafish heart development and regeneration. Development 138, 2895–2902 (2011)

    Article  CAS  Google Scholar 

  8. Zhou, B. et al. Adult mouse epicardium modulates myocardial injury by secreting paracrine factors. J. Clin. Invest. 121, 1894–1904 (2011)

    Article  CAS  Google Scholar 

  9. Serpooshan, V. et al. The effect of bioengineered acellular collagen patch on cardiac remodeling and ventricular function post myocardial infarction. Biomaterials 34, 9048–9055 (2013)

    Article  CAS  Google Scholar 

  10. Tanaka, M. et al. DIP2 disco-interacting protein 2 homolog A (Drosophila) is a candidate receptor for follistatin-related protein/follistatin-like 1–analysis of their binding with TGF-β superfamily proteins. FEBS J. 277, 4278–4289 (2010)

    Article  CAS  Google Scholar 

  11. Widera, C. et al. Identification of Follistatin-Like 1 by expression cloning as an activator of the growth differentiation factor 15 gene and a prognostic biomarker in acute coronary syndrome. Clin. Chem. 58, 1233–1241 (2012)

    Article  CAS  Google Scholar 

  12. Adams, D., Larman, B. & Oxburgh, L. Developmental expression of mouse Follistatin-like 1 (Fstl1): Dynamic regulation during organogenesis of the kidney and lung. Gene Expr. Patterns 7, 491–500 (2007)

    Article  CAS  Google Scholar 

  13. Shimano, M. et al. Cardiac myocyte follistatin-like 1 functions to attenuate hypertrophy following pressure overload. Proc. Natl Acad. Sci. USA 108, E899–E906 (2011)

    Article  CAS  Google Scholar 

  14. Oshima, Y. et al. Follistatin-Like 1 Is an Akt-regulated cardioprotective factor that is secreted by the heart. Circulation 117, 3099–3108 (2008)

    Article  CAS  Google Scholar 

  15. Ogura, Y. et al. Therapeutic impact of follistatin-like 1 on myocardial ischemic injury in preclinical models. Circulation 126, 1728–1738 (2012)

    Article  Google Scholar 

  16. Bergmann, O. et al. Identification of cardiomyocyte nuclei and assessment of ploidy for the analysis of cell turnover. Exp. Cell Res. 317, 188–194 (2011)

    Article  CAS  Google Scholar 

  17. Sohal, D. S. et al. Temporally regulated and tissue-specific gene manipulations in the adult and embryonic heart using a tamoxifen-inducible Cre protein. Circ. Res. 89, 20–25 (2001)

    Article  CAS  Google Scholar 

  18. Oh, H. et al. Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc. Natl Acad. Sci. USA 100, 12313–12318 (2003)

    Article  ADS  CAS  Google Scholar 

  19. Lepilina, A. et al. A dynamic epicardial injury response supports progenitor cell activity during zebrafish heart regeneration. Cell 127, 607–619 (2006)

    Article  CAS  Google Scholar 

  20. Mercola, M., Ruiz-Lozano, P. & Schneider, M. D. Cardiac muscle regeneration: lessons from development. Genes Dev. 25, 299–309 (2011)

    Article  CAS  Google Scholar 

  21. Bersell, K., Arab, S., Haring, B. & Kuhn, B. Neuregulin1/ErbB4 signaling induces cardiomyocyte proliferation and repair of heart injury. Cell 138, 257–270 (2009)

    Article  CAS  Google Scholar 

  22. Chen, H. S., Kim, C. & Mercola, M. Electrophysiological challenges of cell-based myocardial repair. Circulation 120, 2496–2508 (2010)

    Article  Google Scholar 

  23. Senyo, S. E. et al. Mammalian heart renewal by pre-existing cardiomyocytes. Nature 493, 433–436 (2013)

    Article  ADS  CAS  Google Scholar 

  24. Zhang, Y. et al. Dedifferentiation and proliferation of mammalian cardiomyocytes. PLoS ONE 5, e12559 (2010)

    Article  ADS  Google Scholar 

  25. Jopling, C. et al. Zebrafish heart regeneration occurs by cardiomyocyte dedifferentiation and proliferation. Nature 464, 606–609 (2010)

    Article  ADS  CAS  Google Scholar 

  26. Brown, R. A., Wiseman, M., Chuo, C. B., Cheema, U. & Nazhat, S. N. Ultrarapid engineering of biomimetic materials and tissues: fabrication of nano- and microstructures by plastic compression. Adv. Funct. Mater. 15, 1762–1770 (2005)

    Article  CAS  Google Scholar 

  27. Eid, H. et al. Role of epicardial mesothelial cells in the modification of phenotype and function of adult rat ventricular myocytes in primary coculture. Circ. Res. 71, 40–50 (1992)

    Article  CAS  Google Scholar 

  28. Kita-Matsuo, H. et al. Lentiviral vectors and protocols for creation of stable hESC lines for fluorescent tracking and drug resistance selection of cardiomyocytes. PLoS ONE 4, e5046 (2009)

    Article  ADS  Google Scholar 

  29. Fajardo, G. et al. Deletion of the β2-adrenergic receptor prevents the development of cardiomyopathy in mice. J. Mol. Cell Cardiol. 63, 155–164 (2013)

    Article  CAS  Google Scholar 

  30. Sakaue-Sawano, A. et al. Visualizing spatiotemporal dynamics of multicellular cell-cycle progression. Cell 132, 487–498 (2008)

    Article  CAS  Google Scholar 

  31. Bushway, P. J. & Mercola, M. High-throughput screening for modulators of stem cell differentiation. Methods Enzymol. 414, 300–316 (2006)

    Article  CAS  Google Scholar 

  32. Cerignoli, F. et al. High throughput drug risk assessment in human cardiomyocytes by kinetic image cytometry. J. Pharm. Toxicol. Methods 66, 246–256 (2012)

    Article  CAS  Google Scholar 

  33. Serpooshan, V. et al. Reduced hydraulic permeability of three-dimensional collagen scaffolds attenuates gel contraction and promotes the growth and differentiation of mesenchymal stem cells. Acta Biomater. 6, 3978–3987 (2010)

    Article  CAS  Google Scholar 

  34. Serpooshan, V., Muja, N., Marelli, B. & Nazhat, S. N. Fibroblast contractility and growth in plastic compressed collagen gel scaffolds with microstructures correlated with hydraulic permeability. J. Biomed. Mater. Res. A 96, 609–620 (2011)

    Article  Google Scholar 

  35. Abou Neel, E. A., Cheema, U., Knowles, J. C., Brown, R. A. & Nazhat, S. N. Use of multiple unconfined compression for control of collagen gel scaffold density and mechanical properties. Soft Matter 2, 986–992 (2006)

    Article  ADS  CAS  Google Scholar 

  36. Engler, A. J. et al. Embryonic cardiomyocytes beat best on a matrix with heart-like elasticity: scar-like rigidity inhibits beating. J. Cell Sci. 121, 3794–3802 (2008)

    Article  CAS  Google Scholar 

  37. Venugopal, J. R. et al. Biomaterial strategies for alleviation of myocardial infarction. J. Royal Soc. Interface 9, 1–19 (2012)

    Article  CAS  Google Scholar 

  38. Clement, S. et al. Expression and function of alpha-smooth muscle actin during embryonic-stem-cell-derived cardiomyocyte differentiation. J. Cell Sci. 120, 229–238 (2007)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank L. M. Brill for spectrometry, F. Cerignolli for providing valuable cells, S. Metzler and P. Kim for help with imaging, and P. Shah for assistance in mouse and pig experiments. This work was supported by NIH grants to P.R.-L. (HL065484 and R01 HL086879); M.Me. (HL113601, HL108176, P01 HL098053); P30 AR061303 and P30 CA030199 for shared services and by the California Institute for Regenerative Medicine (CIRM, RC1-00132) to M.Me. K.We. and C.H. were SBMRI CIRM postdoctoral fellows (TG2-0116). V.S. was an Oak Foundation postdoctoral fellow. Support was also provided by NIH/NHLBI 5UM1 HL113456 to P.C.Y; HL116591 to K.Wa., M.J.B. was supported by the NIH (K08 AI079268) and the Stanford BioX Interdisciplinary Initiatives Program and NSF NSEC(PHY-0830228). B.Z. was supported by the National Basic Research Program of China (2013CB945302 and 2012CB945102) and the National Natural Science Foundation of China (91339104, 31271552, and 31222038). A Seed Grant to P.R.-L. from the Stanford Cardiovascular Institute supported the swine study.

Author information

Authors and Affiliations

Authors

Contributions

K.We. and W.C. performed experiments on EMCs and mESCs. K.We. generated mCMsESC, and performed cardiomyogenic and proliferation assays on mCMsESC, proliferation assays on NRVC. K.We. and W.Z. performed mass spectrometry experiments. M.D.-C. performed immunostaining of Fstl1. K.We. and A.S. performed calcium transient experiment. V.S. generated cardiac patch. V.S., A.W., and M.J.B. performed biomechanical analysis of cardiac patch biomaterial. M.Z. and V.S. performed mouse MI experiments and echocardiography. Y.M. and P.C.Y. performed MRI analysis. K.We. and W.C. performed immunostaining of α-actinin, pH3 and aurora B. K.We. performed GFP, TUNEL and f4/80 staining. K.We. and P.B. performed analysis of high throughput imaging results. M.Mh. analysed the release of Fstl1 from the patch in vitro. V.S. and G.F. performed assays of adult mouse cardiomyocytes. K.Wa., K.N. and S.M. performed experiments with Fstl1-TG mice. B.Z. performed experiments with adult mouse epicardium-conditioned media. X.T., Q.L. and B.Z. performed Wt1CreERT2 lineage tracing studies and experiments with adult mouse epicardium conditioned medium. K.Wa. and S.M. provided data on systemic delivery of FSTL1. M.N. and M.D.S. provided data on myocyte progenitors. M.J.B. supervised AFM studies. D.B. supervised and coordinated in vivo mouse physiology experiments. V.S., Y.M., and P.C.Y. conducted the preclinical swine study. C.H. performed FSTL1 overexpression and western blot experiments. K.We., V.S., M.Me. and P.R.-L. designed experiments and prepared the manuscript. M.J.B.v.d.H. provided materials.

Corresponding author

Correspondence to Pilar Ruiz-Lozano.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Characterization of mCMsESC cells used in this study.

a, Schematic time-line of cell preparation and treatment. bd, Immunostaining of α-actinin of mCMsESC, showing that the majority of the cells are α-actinin+(b), and the α-actinin lacks striation structures (c). d, Immunostaining of α-smooth muscle actin (αSMA) of mCMsESC, showing the majority of the cells are αSMA+, unlike mature cardiomyocytes with no SMA expression38. e, f, Automatic detection of EdU incorporation in mCMsESC. Captured image of mCMsESC treated with 10 μg ml−1 EdU for 24 h, stained with EdU, α-actinin and DAPI using InCell 1000 (General Electric) (e). Overlay of masks of EdU, α-actinin and DAPI channels with automatic detection software (f). g, EdU incorporation profile of mCMsESC over time. mCMsESC are treated with 10 μg ml−1 EdU for 24 h at time 0 h, 24 h, 48 h, and 144 h. The percentage of EdU+/α-actinin+ cardiomyocytes of all α-actinin+ cardiomyocytes is calculated for each time period. Note the decrease of EdU incorporation rate over time. h, i, Fluo 4 calcium images of mCMsESC, with baseline background image (h) and peak image (i). j, Comparison of representative calcium transients of mCMsESC (red) and neonatal rat ventricular cardiomyocytes (NRVC, blue). Note the reduced amplitude, slower rate of up and down strokes, and elongated duration of the calcium transient in mCMsESC compared to NRVC, suggesting immature calcium handling in mCMsESC. In all experiments, FSTL1 was added one day after plating of the mCMsESC (time 0–24 in this figure).

Extended Data Figure 2 Myocardial overexpression of Fstl1 (Fstl1-TG) mice after permanent LAD ligation.

ad, Fstl1 protein expression kinetics after myocardial infarction. Fstl1-TG mice (C57/Bl6 background) and littermate wild-type (WT) mice underwent LAD ligation. Heart tissue and serum were collected at baseline, day 1, day 3, day 7 and day 28 after surgery. Fstl1 protein levels in ischaemic area (IA) and remote area (RM) of heart were analysed by western blotting (a). Fstl1 expression expressed relative to tubulin levels is reported (b). Fstl1 serum levels were analysed by western blotting (c). Also shown in Ponceau-S staining to indicate equal loading of serum. Quantification of serum Fstl1 level is shown in (d). n > 3 in all groups. *P < 0.05 compared to WT baseline, #P < 0.05 compared to Fstl1-TG baseline. ANOVA was used for statistical significance (P < 0.05). ej, Morphometric and functional response of Fstl1-TG mice to permanent LAD ligation at long-term. Representative Masson’s trichrome staining of WT (e) and Fstl1-TG (f) 4 weeks after MI. Quantification of content in fibrotic tissue at week 4 after MI (g). Echocardiographic measurement of left ventricular internal dimension in systole (LVIDs) (h), and left ventricular internal diameter in diastole (LIVDd) (i) at weeks 2 and 4 after MI. Echocardiographic determination of fractional shortening (FS%) in the indicated genotypes at 2 and 4 weeks after MI (j). kn, Double immunofluorescent staining of α-actinin (cardiomyocytes) and pH3 (mitosis) (k) and α-actinin (cardiomyocytes) and von Willebrand factor (vascular endothelial cells) (m) in the Fstl1-TG and WT mice, quantified in (i, n). n = 5, *P < 0.05 indicates significantly different from WT.

Extended Data Figure 3 Patch with FSTL1 attenuated fibrosis after MI.

af, FSTL1 retention in the patch in vitro and in vivo. a, b, Enzyme-linked immunosorbent assay used to measure the amount of FSTL1 retained within collagen scaffolds exposed to PBS in vitro for different time intervals (0–21 days) (a). The Table lists the initial and final FSTL1 concentration, as well as the release values within the first 24 h (b). cf, FSTL1 retention in the patch in vivo. Representative images of Fstl1 immunostaining in the indicated animal treatment groups, week 4 after surgery. Note that, while Fstl1 is expressed in the uninjured epicardium (arrow in the inset in c), its expression became undetectable within the infarct area after MI (d). Similarly, no FSTL1 was detected in the MI plus patch animals (e), while it still persists (red staining) in the patch area of the MI plus patch with FSTL1 group (f). g, Representative Masson’s Trichrome staining on serial cross sections of hearts under 4 conditions (sham, MI only, MI with patch and MI plus patch with FSTL1) 4 weeks after MI. Note the severe fibrosis in MI only condition, and reduced fibrosis in MI plus patch condition, and further reduction in MI plus patch with FSTL1 condition, quantified in Fig. 3d. hj, Representative MRI images from the mouse MI only, MI plus patch and MI plus patch with FSTL1 treatment groups showing the 3D-FSPGR (fast spoiled gradient-echo) images and the delayed enhancement images using gadolinium contrasting agents, confirming a reduction in infarct area (demarcated in green) and preserved contractility (Supplementary Videos 3, 4, 5). k, Trichrome staining of infarct and border zone of the indicated treatments demonstrates the integration of the patch with the host tissue and massive patch cellularization by the native cardiac cells. Observe the abundant muscle (red) inside the patch and in the border zone of the patch with FSTL1 treated animals (three right panels, green arrowheads).

Extended Data Figure 4 Analysis of patch with FSTL1 function in the mouse model of ischaemia/reperfusion (I/R) with delayed patch grafting.

ac, Heart function evaluation for sham, I/R, and I/R treated with patch with FSTL1, at end- diastolic and systolic, pre-grafting (a, 1 week post-injury), 2 weeks post patch implantation (b), and 4 weeks post grafting (c). Values were normalized by dividing to pre-surgery baseline values for each individual animal. d, Absolute values of fractional shortening (FS, %) at different times pre and post I/R as evaluated by echocardiography of mice from ac. Abbreviations same as in Fig. 3. *P < 0.05 compared to sham and black circle P < 0.05 compared to I/R. e, Co-immunofluorescence staining of DNA duplication marker phospho-Histone3 Ser10 (pH3, green) and α-actinin (red) in the border zone of patch with FSTL1 treated heart 4 weeks after MI. f, Quantification of incidence of pH3+, α-actinin+ double positive cells in the 3 experimental groups. Data collected from 3 hearts in each group with 3 different cross sections counted for total pH3+, α-actinin+ cells in each heart. *P < 0.05 indicates statistically different from all other groups.

Extended Data Figure 5 Representative images and quantification of cardiomyocyte proliferation in vivo after patch with FSTL1 treatment.

ah, Immunostaining of the cardiomyocyte marker α-actinin (red) in the infarct area (bd) and co-immunofluorescence staining of DNA duplication marker phospho-Histone3 Ser10 (pH3, green) and α-actinin (red) in the border zone (fh), in the 4 treatment groups analysed 4 weeks post-MI, compared to sham-operated animals (a, e). Insets in (ad) show lower magnification images with broken lines demarcating the border between the patch and host tissues. Arrowheads in g, h, indicate α-actinin+ cardiomyocytes with pH3+ nuclei. ik, Representative images of pH3+ cardiomyocytes in a patch with FSTL1 treated heart. Masson’s Trichrome staining of a heart after MI 4 weeks treated with patch with FSTL1 (i). The adjacent slide was stained for α-actinin in j, corresponding to the black box area with infarction and the patch in i. The spotted line in j indicates the boundary between the heart and the patch. The adjacent slide was stained for α-actinin and pH3, and all α-actinin+, pH3+ double positive cardiomyocytes found were shown in k (white arrowhead), with each image corresponding to the area in numbered white boxes in j. ln, Quantification of cardiomyocyte proliferation measured in 3 cross sections covering the infarct, patch, and separated by 250 μm, between 1–2 mm from the apex in each heart (Fig. 3j). Data collected from 5–7 hearts in each group with the 3 cross-sections counted exhaustively for incidence of α-actinin+ cells positive for pH3 (l), midbody-localized aurora B kinase between α-actinin+ cells (m), and double-positive cells for pH3 and the nuclear cardiomyocyte maker PCM1 (n), and normalized to myocardium area quantified by trichrome staining of immediate adjacent section. *P < 0.05 statistically different from sham. **P < 0.05, statistically different from all other groups. o, Quantification of hypertrophy in all experimental groups, measured by counting cardiomyocytes in areas of intraventricular wall with perpendicular cross-sections of cardiomyocytes in all hearts analysed for cardiomyocyte proliferation. No significance were found between samples. pr, Quantification of incidence of α-actinin+ cells positive for pH3 (p), midbody-localized aurora B kinase between α-actinin+ cells (q), and double-positive cells for pH3 and the nuclear cardiomyocyte maker PCM1 (r) measured in l, m, to total number of cardiomyocytes, calculated using hypertrophic analysis results in o. *P < 0.05, statistically different from sham, **P < 0.05,: statistically different from all other groups. s, t, Quantification of incidence of α-actinin+ cells positive for pH3 (s) and midbody-localized aurora B kinase between α-actinin+ cells (t), separated by their localization in the border zone or infarcted area. Note the majority of proliferation quantified by both methods are located in the border zone, *P < 0.05, statistically different from all other groups.

Extended Data Figure 6 Effect of implantation of patch with FSTL1 on apoptosis and inflammation.

a, Representative TTC staining of day 2 post MI/patch treatment of all four groups (sham, MI, MI plus patch, MI plus patch with FSTL1). b, Quantification of area at risk comparing all 4 groups. Data collected from 4 hearts in each group, with 4 cross-sections, approximately 2 mm thick each, encompassing each heart. *P < 0.05, statistically different from the sham. c, d, Representative image of TUNEL assays (TUNEL, green, α-actinin, red) comparing hearts 2 days after MI with patch alone and patch with FSTL1. e, Quantification of TUNEL+, α-actinin+ in infarcted area, as percentage of total number of cardiomyocyte. No difference is observed between MI plus patch and MI plus patch with FSTL1 conditions. Data collected from 3 hearts in each group with 3 different cross-sections (same as in Fig. 3j) Ten 0.09 mm2 images were taken from infarcted area of each section and counted for TUNEL+, α-actinin+ and total α-actinin+ cells. f–j, TUNEL staining for cell death and α-actinin staining for cardiomyocytes were performed on hearts treated with patch-only and patch with FSTL1 at day 4 and day 8 after MI (fi). Minimal TUNEL+, α-actinin+ cells are detected while there are signification amount of TUNEL+, α-actinin- cells. Quantification of all TUNEL+ nuclei showed no significant differences between patch and patch with FSTL1 treated hearts at both time points (j). ko, Immunostaining of F4/80 for macrophages and α-actinin for cardiomyocyte were performed on the same hearts as in panels ad (kn). Quantification of F4/80+ cells showed no significant differences between patch and patch with FSTL1 treated hearts at both time points (o).

Extended Data Figure 7 FSTL1 does not induce proliferation in adult and neonatal cardiomyocytes, or cardiac progenitor cells.

af, Adult cardiomyocytes derived from mouse primary isolation. a, Visualization of GFP+ cardiomyocytes isolated from Myh6mERcremER:Rosa26Z/EG mice treated with 4-OH-tamoxifen (OH-Tam) in 3D-collagen patches. bd) Gene expression changes in adult cardiomyocyte treated with FSTL1, including proliferation (b), cardiac-specific (c), and hypertrophy (d) markers. Note no changes in expression of cardiac specific genes, no increase in cell cycle markers (consistent with undetectable Ki67 immunostaining), and decreased hypertrophy markers (n = 3). Cardiomyocytes were embedded within 3D patch were treated with FSTL1 (10 ng ml−1) for duration of 7 days with media change every 2 days. e, f, FUCCI assay in 3D-cultured adult cardiomyocytes, conducted 1 week after the 3D culture. e, Treatment with FSTL1 was performed for 7 days with media change every 2 days. f, Adult cardiomyocytes 3D-cultured control in absence of FSTL1. Note no detectable sign of cardiomyocytes in S/G2/M phases (GFP+) in either condition. Purple arrows point to purple-colored nuclei resulting from co-localization of Hoechst (blue) and G1 phase FUCCI (red) labelling. gj, Primary neonatal rat ventricular cardiomyocytes (NRVC). g, h, Freshly isolated NRVCs stimulated with FSTL1 for 48 h with 10 μg ml−1 EdU, and stained for α-actinin (red) and EdU (green). Percentages of EdU+/α-actinin+ cardiomyocytes of all α-actinin+ cardiomyocytes are quantified (h). i, j, NRVCs stimulated with FSTL1 for 48 h, and stained for α-actinin (red) and pH3 (green). Percentages of pH3+/α-actinin+ cardiomyocytes of all α-actinin+ cardiomyocytes are quantified (j). No increase of proliferation is found upon FSTL1 treatment. (n = 4) *P < 0.05, statistically different from control,. km, Sca1+ progenitor cells18 were starvation-synchronized for 48 h and stimulated with FSTL1 or control growth medium for 72 h in presence of EdU. Clone 3 was obtained by clonal growth from the Lin-Sca1+SP fraction. Sca1 pool was obtained from lin-Sca1+ without clonal growth. k, EdU and DAPI staining of Sca1+ cells after 72 h treatment. l, Percentage of EdU+ Sca1+ cells after 72 h treatment. FSTL1 concentration: 0, 1, 10, 100 ng ml−1. Abbreviation s: SS, serum starvation; CGM, control growth medium. m, Number of Sca1+ cells after 72 h FSTL1 treatment (n = 5). No significant change is found upon FSTL1 treatment.

Extended Data Table 1 Raw echocardiography values (average ± s.e.m.) obtained at days 0 (baseline), 14, and 28 post treatment in a mouse model of permanent LAD ligation
Extended Data Table 2 Raw echocardiography values (average ± s.e.m.) in a long term (months 2 and 3) post treatment, in a mouse model of permanent LAD ligation
Extended Data Table 3 Raw echocardiography values (average ± s.e.m) of delayed grafting in a mouse model of ischaemia/reperfusion

Supplementary information

Supplementary Figure

This file contains uncropped western blot scans with size marker indications. (PDF 361 kb)

Supplementary Table

This table contains a list of primers used in Q-RT-PCR. (XLSX 10 kb)

Autonomous contraction of mCMsESC

Bright field real time video of confluent mCMsESC culture, showing autonomous contraction of the cardiomyocytes. (MP4 6425 kb)

Autonomous calcium transient of mCMsESC

Fluo4 NW calcium transient of mCMsESC culture, imaged by KIC, showing autonomous calcium transient of the cardiomyocytes. (MP4 21504 kb)

MI-only mouse at week 4 after surgery

MRI video taken from representative study on MI-only mouse at week 4 after surgery (MOV 5538 kb)

MI+Patch mouse at week 4 after surgery

MRI video taken from representative study on MI+Patch mouse at week 4 after surgery (MOV 7116 kb)

MI+Patch+FSTL1 mouse at week 4 after surgery

MRI video taken from representative study on MI+Patch+FSTL1 mouse at week 4 after surgery. (MOV 3738 kb)

3-D rendering of Aurora B (red) and α-actinin (green) immunostaining captured by Apotome (Fig. 3m) were processed with ImageJ Z-function to rotate the image Z-Stack. Note the Aurora B+ (red) cleavage furrow between α-actinin+ (green) cardiomyocytes.

3-D rendering of Aurora B (red) and α-actinin (green) immunostaining captured by Apotome (Fig. 3m) were processed with ImageJ Z-function to rotate the image Z-Stack. Note the Aurora B+ (red) cleavage furrow between α-actinin+ (green) cardiomyocytes. (AVI 13644 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wei, K., Serpooshan, V., Hurtado, C. et al. Epicardial FSTL1 reconstitution regenerates the adult mammalian heart. Nature 525, 479–485 (2015). https://doi.org/10.1038/nature15372

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature15372

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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