Summary

Suppression of Pro-fibrotic Signaling Potentiates Factor-mediated Reprogramming of Mouse Embryonic Fibroblasts into Induced Cardiomyocytes

Published: June 03, 2018
doi:

Summary

Here we present a robust method to reprogram primary embryonic fibroblasts into functional cardiomyocytes through overexpression of GATA4, Hand2, Mef2c, Tbx5, miR-1, and miR-133 (GHMT2m) alongside inhibition of TGF-β signaling. Our protocol generates beating cardiomyocytes as early as 7 days post-transduction with up to 60% efficiency.

Abstract

Trans-differentiation of one somatic cell type into another has enormous potential to model and treat human diseases. Previous studies have shown that mouse embryonic, dermal, and cardiac fibroblasts can be reprogrammed into functional induced-cardiomyocyte-like cells (iCMs) through overexpression of cardiogenic transcription factors including GATA4, Hand2, Mef2c, and Tbx5 both in vitro and in vivo. However, these previous studies have shown relatively low efficiency. In order to restore heart function following injury, mechanisms governing cardiac reprogramming must be elucidated to increase efficiency and maturation of iCMs.

We previously demonstrated that inhibition of pro-fibrotic signaling dramatically increases reprogramming efficiency. Here, we detail methods to achieve a reprogramming efficiency of up to 60%. Furthermore, we describe several methods including flow cytometry, immunofluorescent imaging, and calcium imaging to quantify reprogramming efficiency and maturation of reprogrammed fibroblasts. Using the protocol detailed here, mechanistic studies can be undertaken to determine positive and negative regulators of cardiac reprogramming. These studies may identify signaling pathways that can be targeted to promote reprogramming efficiency and maturation, which could lead to novel cell therapies to treat human heart disease.

Introduction

Ischemic heart disease is a leading cause of death in the United States1. Approximately 800,000 Americans experience a first or recurrent myocardial infarction (MI) per year1. Following MI, the death of cardiomyocytes (CMs) and cardiac fibrosis, deposited by activated cardiac fibroblasts, impair heart function2,3. Progression of heart failure following MI is largely irreversible due to the poor regenerative capacity of adult CMs4,5. While current clinical therapies slow disease progression and decrease risk of future cardiac events6,7,8,9, no therapies reverse disease progression due to the inability to regenerate CMs post-infarction10. Novel cell therapies are emerging to treat patients following MI. Disappointingly, clinical trials delivering stem cells to the heart following MI thus far have shown inconclusive regenerative potential11,12,13,14,15,16,17,18.

The generation of human-derived induced pluripotent stem cells (hiPSCs) from fibroblasts by overexpression of four transcription factors, first demonstrated by Takahashi & Yamanaka, opened the door to new breakthroughs in cell therapy19. These cells can differentiate into all three germ layers19, and several highly efficient methods for generating large numbers of CMs have been previously shown20,21. HiPSC-derived CMs (hiPS-CMs) offers a powerful platform to study cardiomyogenesis and may have important implications for repairing the heart following injury. However, hiPS-CMs currently face translational hurdles due to concerns of teratoma formation22, and their immature nature may be pro-arrhythmogenic23. Reprogramming fibroblasts into hiPSCs sparked interest in directly reprogramming fibroblasts into other cell types. Ieda et al. demonstrated that overexpression of GATA4, Mef2c, and Tbx5 (GMT) in fibroblasts results in direct reprogramming to cardiac lineage, albeit at low efficiency24. Reprogramming efficiency was improved with the addition of Hand2 (GHMT)25. Since these early studies, many publications have demonstrated that altering the reprogramming factor cocktail with additional transcription factors26,27,28,29, chromatin modifiers30,31, microRNAs32,33, or small molecules34 leads to improved reprogramming efficiency and/or maturation of induced cardiomyocyte-like cells (iCMs).

Here we provide a detailed protocol to generate iCMs from mouse embryonic fibroblasts (MEFs) with high efficiency. We previously showed that the GHMT cocktail is significantly improved with the addition of miR-1 and miR-133 (GHMT2m) and is further improved when pro-fibrotic signaling pathways including transforming growth factor β (TGF-β) signaling or Rho-associated protein kinase (ROCK) signaling pathways are inhibited35. Using this protocol, we show that approximately 60% of cells express cardiac Troponin T (cTnT), approximately 50% express α-actinin, and a high number of beating cells can be observed as early as Day 11 following transduction of reprogramming factors and treatment with the TGF-β type I receptor inhibitor A-83-01. Furthermore, these iCMs express gap junction proteins including connexin 43 and exhibit spontaneous contraction and calcium transients. This marked improvement in reprogramming efficiency compared to earlier studies demonstrates the potential to regenerate CMs from endogenous cell populations that remain in the heart post-infarction.

Protocol

All experiments requiring animals were approved by the Institutional Animal Care and Use Committee at the UC Denver Anschutz Medical Campus. 1. Isolation of MEFs Purchase C57BL/6 pregnant mice at E13. Ship overnight. Euthanize the mother according to approved IACUC protocols (ex: ~1.3 L/min CO2 until animal appears dead followed by cervical dislocation) Spray the mother with 70% ethanol and open abdominal cavity. Remove the uterine horn containing emb…

Representative Results

Using the reprogramming strategy outlined above and in Figure 1B, we generated iCMs with approximately 70% of cells expressing cardiac Troponin T and approximately 55% of cells expressing cardiac α-actinin, quantified by flow cytometry at Day 9 following transduction of GHMT2m (Figure 2A and B). Additionally, the majority of cells express cardiac Troponin T, Troponin I, and cardiac α-actinin as well as …

Discussion

The present study outlines a high-efficiency strategy to directly reprogram fibroblasts into functional iCMs via delivery of GHMT2m reprogramming factors combined with suppression of pro-fibrotic signaling pathways. Using flow cytometry, immunofluorescent imaging, calcium imaging, and beating cell counts, we show the majority of cells in this protocol undergo successful reprogramming and adopt CM lineage fate. We have previously shown that the addition of anti-fibrotic compounds including the TGF-β type I receptor i…

Divulgaciones

The authors have nothing to disclose.

Acknowledgements

This research was supported by funds from the Boettcher Foundation's Webb-Waring Biomedical Research Program, American Heart Association Scientist Development Grant (13SDG17400031), University of Colorado Department of Medicine Outstanding Early Career Scholar Program, University of Colorado Division of Cardiology Barlow Nyle endowment, and NIH R01HL133230 (to K.S). A.S.R was supported by NIH/NCATS Colorado CTSA Grant Number TL1TR001081 and a pre-doctoral fellowship from the University of Colorado Consortium for Fibrosis Research & Translation (CFReT). This research was also supported by the Cancer Center Support Grant (P30CA046934), the Skin Diseases Research Cores Grant (P30AR057212), and the Flow Cytometry Core at the University of Colorado Anschutz Medical Campus.

Materials

C57BL/6 Mice Charles River's Laboratory 027 For MEF isolation
Platinum E (PE) Cells Cell Biolabs, INC RV-101 For retrovirus production
DMEM High Glucose Gibco SH30022.FS Component of iCM, PE, and Growth media
Medium 199 Life Technologies 11150-059 Component of iCM media
Fetal Bovine Serum Gemini 100106 Component of iCM, PE, and Growth media
Donor Horse Serum Gemini 100508 500 Component of iCM media
MEM Essential Amino Acids, 50X Life Technologies 11130051 Component of iCM media
Sodium Pyruvate Solution, 100X Life Technologies 11360070 Component of iCM media and for calcium imaging
MEM Non-Essential Amino Acids, 100X Life Technologies 11140050 Component of iCM media
MEM Vitamin Solution, 100X Life Technologies 11120-052 Component of iCM media
Insulin-Transferrin-Selenium Gibco 41400045 Component of iCM media
B27 Gibco 17504-044 Component of iCM media
Penicilin-Streptomycin Gibco 15140-122 Component of iCM, PE, and Growth media
GlutaMAX (L-Glutamine Supplement) Gibco 35050-061 Component of iCM, PE, and Growth media
Blasticidin-HCl Life Technologies A11139-03 Component of PE media
Puromycin dihydrochloride Life Technologies A11138-03 Component of PE media
0.25% Trypsin/EDTA Gibco 25200-056 For detaching cells from culture dishes
A-83-01 R&D Systems – Tocris 2939/10 Treat cells to inhibit TGF-β signaling – promotes high efficiecy reprogramming. Use at 0.5 µM
DMSO Thermo Scientific 85190 For dilution and storage of A-83-01 and component of Freeze Medium
SureCoat Cellutron SC-9035 For coating dishes to plate MEFs
FuGENE 6 Transfection Reagent Promega E2692 Transfection Reagent
Opti-MEM Reduced Serum Media Gibco 11058-021 Transfection Reagent
pBabe-X Myc-GATA4 Plasmid containing reprogramming factor
pBabe-X Myc-Hand2 Plasmid containing reprogramming factor
pBabe-X Myc-Mef2c Plasmid containing reprogramming factor
pBabe-X Myc-Tbx5 Plasmid containing reprogramming factor
pBabe-X miR-1 Plasmid containing reprogramming factor
pBabe-X miR-133 Plasmid containing reprogramming factor
pBabe-X GFP Plasmid containing reprogramming factor
Polybrene (Hexadimethrine bromide) Sigma H9268-5G For viral induction. Use at a concentration of 6 µg/mL
Vacuum Filter + bottles (0.22 µm pores) Nalgene  569-0020  For filtering media
Syringes Bd Vacutainer Labware  309654 For viral filtration
0.45 µm Filters Celltreat 229749 For viral filtration
70 µm cell strainers Falcon  352350 For MEF isolation and Flow Cytometry
Cytofix/Cytoperm Solution BD 554722 For fixation and permeabilization of cells for flow cytometry
perm/wash buffer  BD 554723 For washing cells for flow cytometry
DPBS 1X Gibco 14190-250 For washing cells
Bovine Serum Albumin VWR 0332-100g For flow cytometry and calcium imaging
Goat Serum Sigma  G9023 For blocking cells for Flow Cytometry
Donkey Serum Sigma D9663-10mg For blocking cells for Flow Cytometry
Mouse Troponin T Thermo Scientific ms-295-p 1:400 IF, 1:200 Flow Cytometry
Mouse α-actinin Sigma A7811L 1:400 IF, 1:200 Flow Cytometry
Rabbit Connexin 43 Sigma  C6219 1:400 IF
Rabbit Troponin I PhosphoSolutions 2010-TNI 1:400 IF
Hoechst Life Technologies 62249 1:10000 IF
Alexa 488, rabbit Life Technologies A-11034 1:800 IF
Alexa 555, mouse Life Technologies A-21422 1:800 IF
Alexa 647, mouse Life Technologies A-31571 1:200 Flow Cytometry
27-color ZE5 Flow Cytometer  Bio-RAD For FACS
Paraformaldehyde sigma P6148-500mg For fixing cells for IF
Triton X-100 Promega H5142 For permeabilization of cells for IF
EVOS™ FL Color Imaging System Thermo Scientific AMEFC4300 For IF
NaCl RPI S23020-5000 For calcium imaging
KCl VWR 395 For calcium imaging
CaCl2 Fisher C614-500 For calcium imaging
MgCl2 VWR 97061-352 For calcium imaging
glucose sigma G7528-250g For calcium imaging
HEPES sigma H4034-500g For calcium imaging
Fura-2 AM Life Technologies F1221 For calcium imaging
Fluronic F-127 Sigma P2443-250g For calcium imaging
Nifedipine Sigma N7634-1G For disruption calcium transients in iCMs – use at 10 µM
Isoproterenol sigma I6504-1g For increasing number of calcium transients in iCMs – use at 1-2 µM
Marianas Spinning Disk Confocal microscope 3i For calcium imaging
ethanol Decon Laboratories 2801
bleach Clorox
50 mL conical tubes GREINER BIO-ONE 227261
15 mL conical tubes GREINER BIO-ONE 188271
15 cm cell culture dishes Falcon 353025
10 cm cell culture dishes Falcon 353003
60 mm cell culture dishes GREINER BIO-ONE 628160
6 well cell culture plates GREINER BIO-ONE 657160 
12 well cell culture plates GREINER BIO-ONE 665180 
24 well cell culture plates GREINER BIO-ONE 662160 

Referencias

  1. Benjamin, E. J., et al. Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association. Circulation. 136 (20), (2017).
  2. Laflamme, M. A., Murry, C. E. Regenerating the heart. Nat. Biotechnol. 23 (7), 845-856 (2005).
  3. Mercola, M., Ruiz-Lozano, P., Schneider, M. D. Cardiac muscle regeneration: lessons from development. Genes & Development. 25 (4), 299-309 (2011).
  4. Bergmann, O., et al. Evidence for cardiomyocyte renewal in humans. Science. 324 (5923), 98-102 (2009).
  5. Senyo, S. E., et al. Mammalian heart renewal by pre-existing cardiomyocytes. Nature. 493 (7432), 433-436 (2013).
  6. Nabel, E. G., Braunwald, E. A tale of coronary artery disease and myocardial infarction. N Engl J Med. 366 (1), 54-63 (2012).
  7. Packer, M., et al. Effect of carvedilol on the morbidity of patients with severe chronic heart failure: results of the carvedilol prospective randomized cumulative survival (COPERNICUS) study. Circulation. 106 (17), 2194-2199 (2002).
  8. The CAPRICORN Investigators. Effect of carvedilol on outcome after myocardial infarction in patients with left-ventricular dysfunction: the CAPRICORN randomized trial. The Lancet. 357 (9266), 1385-1390 (2001).
  9. Marangou, J., Paul, V. Current attitudes on cardiac devices in heart failure: a review. Clin Ther. 37 (10), 2206-2214 (2015).
  10. Xin, M., Olson, E. N., Bassel-Duby, R. Mending broken hearts: cardiac development as a basis for adult heart regeneration and repair. Nat Rev Mol Cell Bio. 14 (8), 529-541 (2013).
  11. Wollert, K. C., et al. Intracoronary autologous bone-marrow cell transfer after myocardial infarction: the BOOST randomized controlled clinical trial. Lancet. 364 (9429), 141-148 (2004).
  12. Schachinger, V., et al. Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. N. Engl. J. Med. 355 (12), 1210-1221 (2006).
  13. Huikuri, H. V., et al. Effects of intracoronary injection of mononuclear bone marrow cells on left ventricular function, arrhythmia risk profile, and restenosis after thrombolytic therapy of acute myocardial infarction. Eur. Heart J. 29 (22), 2723-2732 (2008).
  14. Janssens, S., et al. Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet. 367 (9505), 113-121 (2006).
  15. Lunde, K., et al. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. N. Engl. J. Med. 355 (12), 1199-1209 (2006).
  16. Hirsch, A., et al. Intracoronary infusion of autologous mononuclear bone marrow cells or peripheral mononuclear blood cells after primary percutaneous coronary intervention: rationale and design of the HEBE trial – a prospective, multicenter, randomized trial. Am. Heart J. 152 (3), 434-441 (2006).
  17. Hirsch, A., et al. Intracoronary infusion of mononuclear cells from bone marrow or peripheral blood compared with standard therapy in patients after acute myocardial infarction treated by primary percutaneous coronary intervention: results of the randomized controlled HEBE trial. Eur. Heart J. 32 (14), 1736-1747 (2011).
  18. Karantalis, V., et al. Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting: The Prospective Randomized Study of Mesenchymal Stem Cell Therapy in Patients Undergoing Cardiac Surgery (PROMETHEUS) trial. Circ Res. 114 (8), 1302-1310 (2014).
  19. Takahashi, K., Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 126 (4), 663-676 (2006).
  20. Lian, X., et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci. 109 (27), E1848-E1857 (2012).
  21. Loh, K. M., et al. Mapping the Pairwise Choices Leading from Pluripotency to Human Bone, Heart, and Other Mesoderm Cell Types. Cell. 166 (2), 451-467 (2016).
  22. Yoshida, Y., Yamanaka, S. iPS cells: A source of cardiac regeneration. J. Mol. Cell. Cardiol. 50 (2), 327-332 (2011).
  23. Shiba, Y., et al. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nature. 538, 388-391 (2016).
  24. Ieda, M., et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell. 142 (3), 357-386 (2010).
  25. Song, K., et al. Heart repair by reprogramming non-myocytes with cardiac transcription factors. Nature. 485, 599-604 (2012).
  26. Hirai, H., Katoku-Kikyo, N., Keirstead, S. A., Kikyo, N. Accelerated direct reprogramming of fibroblasts into cardiomyocyte-like cells with the MyoD transactivation domain. Cardiovasc Res. 100 (1), 105-113 (2013).
  27. Qian, L., Berry, E. C., Fu, J., Ieda, M., Srivastava, D. Reprogramming of mouse fibroblasts into cardiomyocyte-like cells in vitro. Nat Protoc. 8 (6), 1204-1215 (2013).
  28. Addis, R. C., et al. Optimization of direct fibroblast reprogramming to cardiomyocytes using calcium activity as a functional measure of success. J Mol Cell Cardiol. 60, 97-106 (2013).
  29. Ifkovitz, J. L., Addis, R. C., Epstein, J. A., Gearhart, J. D. Inhibition of TGFβ signaling increases direct conversion of fibroblasts to induced cardiomyocytes. PLoS One. 9 (2), e89678 (2014).
  30. Zhou, Y., et al. Bmi1 Is a Key Epigenetic Barrier to Direct Cardiac Reprogramming. Cell Stem Cell. 18 (3), 382-395 (2016).
  31. Christoforou, N., et al. Transcription factors MYOCD, SRF, Mesp1 and SMARCD3 enhance the cardio-inducing effect of GATA4, TBX5, and MEF2C during direct cellular reprogramming. PLoS One. 8 (5), e63577 (2013).
  32. Jayawardena, T. M., et al. MicroRNA induced cardiac reprogramming in vivo: evidence for mature cardiac myocytes and improved cardiac function. Circ Res. 116 (3), 418-424 (2015).
  33. Jayawardena, T. M., et al. MicroRNA-mediated in vitro and in vivo direct reprogramming of cardiac fibroblasts to cardiomyocytes. Circ Res. 110 (11), 1465-1473 (2012).
  34. Cao, N., et al. Conversion of human fibroblasts into functional cardiomyocytes by small molecules. Science. 352 (6290), 1216-1220 (2016).
  35. Zhao, Y., et al. High-efficiency reprogramming of fibroblasts into cardiomyocytes requires suppression of pro-fibrotic signalling. Nature Communications. 6, 1-15 (2015).
  36. Morita, S., Kojima, T., Kitamura, T. Plat-E: an efficient and stable system for transient packaging of retroviruses. Gene Therapy. 7, 1063-1066 (2000).
  37. Zhou, H., Dickson, M. E., Kim, M. S., Bassel-Duby, R., Olson, E. N. Akt1/protein kinase B enhances reprogramming of fibroblasts to functional cardiomyocytes. Proc Natl Acad Sci. 112 (38), 11864-11869 (2015).
  38. Zhou, H., et al. ZFN281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression. Genes & Dev. 31, 1770-1783 (2017).
  39. Mohamed, T. M., et al. Chemical Enhancement of In Vitro and In Vivo Direct Cardiac Reprogramming. Circulation. 135, 978-995 (2017).
  40. Qian, L., et al. In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature. 485, 593-598 (2012).
  41. Mathison, M., et al. In situ reprogramming to transdifferentiate fibroblasts into cardiomyocytes using adenoviral vectors: Implications for clinical myocardial regeneration. J Thorac Cardiovasc Surg. , 329-339 (2017).
check_url/es/57687?article_type=t

Play Video

Citar este artículo
Riching, A. S., Zhao, Y., Cao, Y., Londono, P., Xu, H., Song, K. Suppression of Pro-fibrotic Signaling Potentiates Factor-mediated Reprogramming of Mouse Embryonic Fibroblasts into Induced Cardiomyocytes. J. Vis. Exp. (136), e57687, doi:10.3791/57687 (2018).

View Video