期刊论文详细信息
BMC Cell Biology
Three-dimensional poly-(ε-caprolactone) nanofibrous scaffolds directly promote the cardiomyocyte differentiation of murine-induced pluripotent stem cells through Wnt/β-catenin signaling
Qiang-Sun Zheng3  Li Wei2  Jiang-Hui Xie1  Song Yan3  Ting Wei3  Wen-Ju Li3  Xiong-Tao Liu3  Xiao-Li Li3  Lu Ding3  Di Zeng3  Yan Chen1 
[1] Department of Emergency, Chinese PLA No.401 Hospital, 22 Minjiang Road, Qingdao 266071, China;Department of Cardiology, Chinese PLA No.401 Hospital, 22 Minjiang Road, Qingdao 266071, China;Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi’an 710038, China
关键词: Cardiomyocyte differentiation;    Induced pluripotent stem cell;    Nanofibrous scaffold;    Poly-(ε-caprolactone);   
Others  :  1224001
DOI  :  10.1186/s12860-015-0067-3
 received in 2015-03-20, accepted in 2015-08-21,  发布年份 2015
PDF
【 摘 要 】

Background

Environmental factors are important for stem cell lineage specification, and increasing evidence indicates that the nanoscale geometry/topography of the extracellular matrix (ECM) directs stem cell fate. Recently, many three-dimensional (3D) biomimetic nanofibrous scaffolds resembling many characteristics of the native ECM have been used in stem cell-based myocardial tissue engineering. However, the biophysical role and underlying mechanism of 3D nanofibrous scaffolds in cardiomyocyte differentiation of induced pluripotent stem cells (iPSCs) remain unclear.

Results

Here, we fabricated a 3D poly-(ε-caprolactone) (PCL) nanofibrous scaffold using the electrospinning method and verified its nanotopography and porous structure by scanning electron microscopy. We seeded murine iPSCs (miPSCs) directly on the 3D PCL nanofibrous scaffold and initiated non-directed, spontaneous differentiation using the monolayer method. After the 3D PCL nanofibrous scaffold was gelatin coated, it was suitable for monolayer miPSC cultivation and cardiomyocyte differentiation. At day 15 of differentiation, miPSCs differentiated into functional cardiomyocytes on the 3D PCL nanofibrous scaffold as evidenced by positive immunostaining of cardiac-specific proteins including cardiac troponin T (cTnT) and myosin light chain 2a (MLC2a). In addition, flow cytometric analysis of cTnT-positive cells and cardiac-specific gene and protein expression of cTnT and sarcomeric alpha actinin (α-actinin) demonstrated that the cardiomyocyte differentiation of miPSCs was more efficient on the 3D PCL nanofibrous scaffold than on normal tissue culture plates (TCPs). Furthermore, early inhibition of Wnt/β-catenin signaling by the selective antagonist Dickkopf-1 significantly reduced the activity of Wnt/β-catenin signaling and decreased the cardiomyocyte differentiation of miPSCs cultured on the 3D PCL nanofibrous scaffold, while the early activation of Wnt/β-catenin signaling by CHIR99021 further increased the cardiomyocyte differentiation of miPSCs.

Conclusion

These results indicated that the electrospun 3D PCL nanofibrous scaffolds directly promoted the cardiomyocyte differentiation of miPSCs, which was mediated by the activation of the Wnt/β-catenin signaling during the early period of differentiation. These findings highlighted the biophysical role of 3D nanofibrous scaffolds during the cardiomyocyte differentiation of miPSCs and revealed its underlying mechanism involving Wnt/β-catenin signaling, which will be helpful in guiding future stem cell- and scaffold-based myocardium bioengineering.

【 授权许可】

   
2015 Chen et al.

【 预 览 】
附件列表
Files Size Format View
20150907013242692.pdf 3394KB PDF download
Fig. 5. 72KB Image download
Fig. 4. 128KB Image download
Fig. 3. 104KB Image download
Fig. 2. 98KB Image download
Fig. 1. 105KB Image download
【 图 表 】

Fig. 1.

Fig. 2.

Fig. 3.

Fig. 4.

Fig. 5.

【 参考文献 】
  • [1]Neubauer S. The failing heart--an engine out of fuel. N Engl J Med. 2007; 356(11):1140-51.
  • [2]Zimmermann WH, Melnychenko I, Wasmeier G, Didie M, Naito H, Nixdorff U et al.. Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat hearts. Nat Med. 2006; 12(4):452-8.
  • [3]Martins AM, Vunjak-Novakovic G, Reis RL. The current status of iPS cells in cardiac research and their potential for tissue engineering and regenerative medicine. Stem Cell Rev. 2014; 10(2):177-90.
  • [4]Radisic M, Christman KL. Materials science and tissue engineering: repairing the heart. Mayo Clin Proc. 2013; 88(8):884-98.
  • [5]Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006; 126(4):663-76.
  • [6]Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S et al.. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007; 318(5858):1917-20.
  • [7]Takahashi K. Cellular reprogramming. Cold Spring Harb Perspect Biol 2014;6(2). doi:10.1101/cshperspect.a018606.
  • [8]Yoshida Y, Yamanaka S. iPS cells: a source of cardiac regeneration. J Mol Cell Cardiol. 2011; 50(2):327-32.
  • [9]Kshitiz, Park J, Kim P, Helen W, Engler AJ, Levchenko A et al.. Control of stem cell fate and function by engineering physical microenvironments. Integr Biol (Camb). 2012; 4(9):1008-18.
  • [10]Liu W, Thomopoulos S, Xia Y. Electrospun nanofibers for regenerative medicine. Adv Healthc Mater. 2012; 1(1):10-25.
  • [11]Oh B, Lee CH. Nanofiber for cardiovascular tissue engineering. Expert Opin Drug Deliv. 2013; 10(11):1565-82.
  • [12]Kharaziha M, Nikkhah M, Shin SR, Annabi N, Masoumi N, Gaharwar AK et al.. PGS: Gelatin nanofibrous scaffolds with tunable mechanical and structural properties for engineering cardiac tissues. Biomaterials. 2013; 34(27):6355-66.
  • [13]Kensah G, Roa LA, Dahlmann J, Zweigerdt R, Schwanke K, Hegermann J et al.. Murine and human pluripotent stem cell-derived cardiac bodies form contractile myocardial tissue in vitro. Eur Heart J. 2013; 34(15):1134-46.
  • [14]Mummery CL, Zhang J, Ng ES, Elliott DA, Elefanty AG, Kamp TJ. Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview. Circ Res. 2012; 111(3):344-58.
  • [15]Griffith LG, Swartz MA. Capturing complex 3D tissue physiology in vitro. Nat Rev Mol Cell Biol. 2006; 7(3):211-24.
  • [16]Huebsch N, Arany PR, Mao AS, Shvartsman D, Ali OA, Bencherif SA et al.. Harnessing traction-mediated manipulation of the cell/matrix interface to control stem-cell fate. Nat Mater. 2010; 9(6):518-26.
  • [17]Wu SM. Mesp1 at the heart of mesoderm lineage specification. Cell Stem Cell. 2008; 3(1):1-2.
  • [18]Bondue A, Tannler S, Chiapparo G, Chabab S, Ramialison M, Paulissen C et al.. Defining the earliest step of cardiovascular progenitor specification during embryonic stem cell differentiation. J Cell Biol. 2011; 192(5):751-65.
  • [19]Eisenberg LM, Eisenberg CA. Wnt signal transduction and the formation of the myocardium. Dev Biol. 2006; 293(2):305-15.
  • [20]Naito AT, Shiojima I, Akazawa H, Hidaka K, Morisaki T, Kikuchi A et al.. Developmental stage-specific biphasic roles of Wnt/beta-catenin signaling in cardiomyogenesis and hematopoiesis. Proc Natl Acad Sci U S A. 2006; 103(52):19812-7.
  • [21]Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM et al.. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci U S A. 2012; 109(27):E1848-57.
  • [22]Kawamura M, Miyagawa S, Miki K, Saito A, Fukushima S, Higuchi T et al.. Feasibility, safety, and therapeutic efficacy of human induced pluripotent stem cell-derived cardiomyocyte sheets in a porcine ischemic cardiomyopathy model. Circulation. 2012; 126(11 Suppl 1):S29-37.
  • [23]Baarsma HA, Konigshoff M, Gosens R. The WNT signaling pathway from ligand secretion to gene transcription: molecular mechanisms and pharmacological targets. Pharmacol Ther. 2013; 138(1):66-83.
  • [24]Kuppan P, Sethuraman S, Krishnan UM. PCL and PCL-gelatin nanofibers as esophageal tissue scaffolds: optimization, characterization and cell-matrix interactions. J Biomed Nanotechnol. 2013; 9(9):1540-55.
  • [25]Sill TJ, von Recum HA. Electrospinning: applications in drug delivery and tissue engineering. Biomaterials. 2008; 29(13):1989-2006.
  • [26]Hashemi SM, Soleimani M, Zargarian SS, Haddadi-Asl V, Ahmadbeigi N, Soudi S et al.. In vitro differentiation of human cord blood-derived unrestricted somatic stem cells into hepatocyte-like cells on poly(epsilon-caprolactone) nanofiber scaffolds. Cells Tissues Organs. 2009; 190(3):135-49.
  • [27]Lim SH, Mao HQ. Electrospun scaffolds for stem cell engineering. Adv Drug Deliv Rev. 2009; 61(12):1084-96.
  • [28]Fleischer S, Feiner R, Shapira A, Ji J, Sui X, Daniel WH et al.. Spring-like fibers for cardiac tissue engineering. Biomaterials. 2013; 34(34):8599-606.
  • [29]Oberwallner B, Brodarac A, Anic P, Saric T, Wassilew K, Neef K et al.. Human cardiac extracellular matrix supports myocardial lineage commitment of pluripotent stem cells. Eur J Cardiothorac Surg. 2015; 47(3):416-25.
  • [30]Ghasemi-Mobarakeh L, Prabhakaran MP, Morshed M, Nasr-Esfahani MH, Ramakrishna S. Electrospun poly(epsilon-caprolactone)/gelatin nanofibrous scaffolds for nerve tissue engineering. Biomaterials. 2008; 29(34):4532-9.
  • [31]Abbasi N, Soudi S, Hayati-Roodbari N, Dodel M, Soleimani M. The Effects of Plasma Treated Electrospun Nanofibrous Poly (epsilon-caprolactone) Scaffolds with Different Orientations on Mouse Embryonic Stem Cell Proliferation. Cell J. 2014; 16(3):245-54.
  • [32]Gupta MK, Walthall JM, Venkataraman R, Crowder SW, Jung DK, Yu SS et al.. Combinatorial polymer electrospun matrices promote physiologically-relevant cardiomyogenic stem cell differentiation. PLoS One. 2011; 6(12): Article ID e28935
  • [33]Tran TH, Wang X, Browne C, Zhang Y, Schinke M, Izumo S et al.. Wnt3a-induced mesoderm formation and cardiomyogenesis in human embryonic stem cells. Stem Cells. 2009; 27(8):1869-78.
  • [34]Jho EH, Zhang T, Domon C, Joo CK, Freund JN, Costantini F. Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol Cell Biol. 2002; 22(4):1172-83.
  • [35]Sun Y, Chen CS, Fu J. Forcing stem cells to behave: a biophysical perspective of the cellular microenvironment. Annu Rev Biophys. 2012; 41:519-42.
  • [36]Jansen JH, Eijken M, Jahr H, Chiba H, Verhaar JA, van Leeuwen JP et al.. Stretch-induced inhibition of Wnt/beta-catenin signaling in mineralizing osteoblasts. J Orthop Res. 2010; 28(3):390-6.
  • [37]Geiger B, Spatz JP, Bershadsky AD. Environmental sensing through focal adhesions. Nat Rev Mol Cell Biol. 2009; 10(1):21-33.
  • [38]Willems E, Spiering S, Davidovics H, Lanier M, Xia Z, Dawson M et al.. Small-molecule inhibitors of the Wnt pathway potently promote cardiomyocytes from human embryonic stem cell-derived mesoderm. Circ Res. 2011; 109(4):360-4.
  • [39]Wang H, Hao J, Hong CC. Cardiac induction of embryonic stem cells by a small molecule inhibitor of Wnt/beta-catenin signaling. ACS Chem Biol. 2011; 6(2):192-7.
  • [40]Schier AF. Nodal morphogens. Cold Spring Harb Perspect Biol. 2009; 1(5):a3459.
  • [41]Van Vliet P, Wu SM, Zaffran S, Puceat M. Early cardiac development: a view from stem cells to embryos. Cardiovasc Res. 2012; 96(3):352-62.
  • [42]Zeng D, Ou DB, Wei T, Ding L, Liu XT, Hu XL et al.. Collagen/beta(1) integrin interaction is required for embryoid body formation during cardiogenesis from murine induced pluripotent stem cells. BMC Cell Biol. 2013; 14:5.
  文献评价指标  
  下载次数:9次 浏览次数:21次