Generic placeholder image

Current Molecular Medicine

Editor-in-Chief

ISSN (Print): 1566-5240
ISSN (Online): 1875-5666

Research Article

Overexpression of Pygo2 Increases Differentiation of Human Umbilical Cord Mesenchymal Stem Cells into Cardiomyocyte-like Cells

Author(s): Lei Yang, Shuoji Zhu, Yongqing Li, Jian Zhuang, Jimei Chen, Huanlei Huang, Yu Chen, Yulin Wen, Yao Wen, Huiming Guo, Xiongwei Fan, Wuzhou Yuan, Zhigang Jiang*, Yuequn Wang*, Xiushan Wu* and Ping Zhu*

Volume 20, Issue 4, 2020

Page: [318 - 324] Pages: 7

DOI: 10.2174/1566524019666191017150416

Price: $65

Abstract

Background: Our previous studies have shown that Pygo (Pygopus) in Drosophila plays a critical role in adult heart function that is likely conserved in mammals. However, its role in the differentiation of human umbilical cord mesenchymal stem cells (hUC-MSCs) into cardiomyocytes remains unknown.

Objective: To investigate the role of pygo2 in the differentiation of hUC-MSCs into cardiomyocytes.

Methods: Third passage hUC-MSCs were divided into two groups: a p+ group infected with the GV492-pygo2 virus and a p− group infected with the GV492 virus. After infection and 3 or 21 days of incubation, Quantitative real-time PCR (qRT-PCR) was performed to detect pluripotency markers, including OCT-4 and SOX2. Nkx2.5, Gata-4 and cTnT were detected by immunofluorescence at 7, 14 and 21 days post-infection, respectively. Expression of cardiac-related genes—including Nkx2.5, Gata-4, TNNT2, MEF2c, ISL-1, FOXH1, KDR, αMHC and α-Actin—were analyzed by qRT-PCR following transfection with the virus at one, two and three weeks.

Results: After three days of incubation, there were no significant changes in the expression of the pluripotency stem cell markers OCT-4 and SOX2 in the p+ group hUC-MSCs relative to controls (OCT-4: 1.03 ± 0.096 VS 1, P > 0.05, SOX2: 1.071 ± 0.189 VS 1, P > 0.05); however, after 21 days, significant decreases were observed (OCT-4: 0.164 ± 0.098 VS 1, P < 0.01, SOX2: 0.209 ± 0.109 VS 1, P < 0.001). Seven days following incubation, expression of mesoderm specialisation markers, such as Nkx2.5, Gata-4, MEF2c and KDR, were increased; at 14 days following incubation, expression of cardiac genes, such as Nkx2.5, Gata-4, TNNT2, MEF2c, ISL-1, FOXH1, KDR, αMHC and α-Actin, were significantly upregulated in the p+ group relative to the p− group (P < 0.05). Taken together, these findings suggest that overexpression of pygo2 results in more hUCMSCs gradually differentiating into cardiomyocyte-like cells.

Conclusion: We are the first to show that overexpression of pygo2 significantly enhances the expression of cardiac-genic genes, including Nkx2.5 and Gata-4, and promotes the differentiation of hUC-MSCs into cardiomyocyte-like cells.

Keywords: Human umbilical cord mesenchymal stem cells, pygo2, overexpression, differentiation, cardiomyocytes, Nkx2.5, SOX2.

« Previous
[1]
Reed GW, Rossi JE, Cannon CP. Acute myocardial infarction. Lancet 2017; 389(10065): 197-210.
[http://dx.doi.org/10.1016/S0140-6736(16)30677-8] [PMID: 27502078]
[2]
Mehta LS, Beckie TM, DeVon HA, et al. Acute myocardial infarction in women: A scientific statement from the american heart association. Circulation 2016; 133(9): 916-47.
[http://dx.doi.org/10.1161/CIR.0000000000000351] [PMID: 26811316]
[3]
Antman EM, Anbe DT, Armstrong PW, et al. ACC/AHA guidelines for the management of patients with ST-elevation myocardial infarction--executive summary. A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing Committee to revise the 1999 guidelines for the management of patients with acute myocardial infarction). J Am Coll Cardiol 2004; 44(3): 671-719.
[http://dx.doi.org/10.1016/j.jacc.2004.07.002] [PMID: 15358045]
[4]
Puymirat E, Simon T, Danchin N. Response by Puymirat et al. to Letter regarding article, “acute myocardial infarction changes in patient characteristics, management, and 6-month outcomes over a period of 20 years in the FAST-MI program (french registry of acute ST elevation or non-ST-elevation myocardial infarction) 1995 to 2015. Circulation 2018; 137(21): 2307-8.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.118.033969] [PMID: 29784688]
[5]
Ozaki Y, Katagiri Y, Onuma Y, et al. CVIT expert consensus document on primary percutaneous coronary intervention (PCI) for acute myocardial infarction (AMI) in 2018. Cardiovasc Interv Ther 2018; 33(2): 178-203.
[http://dx.doi.org/10.1007/s12928-018-0516-y] [PMID: 29594964]
[6]
Alpert JS, Thygesen KA, White HD, Jaffe AS. Diagnostic and therapeutic implications of type 2 myocardial infarction: review and commentary. Am J Med 2014; 127(2): 105-8.
[http://dx.doi.org/10.1016/j.amjmed.2013.09.031] [PMID: 24462011]
[7]
Liu S, Zhou J, Zhang X, et al. Strategies to optimize adult stem cell therapy for tissue regeneration. Int J Mol Sci 2016; 17(6): 982.
[http://dx.doi.org/10.3390/ijms17060982] [PMID: 27338364]
[8]
Bang OY, Kim EH, Cha JM, Moon GJ. Adult Stem Cell Therapy for Stroke: Challenges and Progress. J Stroke 2016; 18(3): 256-66.
[http://dx.doi.org/10.5853/jos.2016.01263] [PMID: 27733032]
[9]
Kwon SG, Kwon YW, Lee TW, Park GT, Kim JH. Recent advances in stem cell therapeutics and tissue engineering strategies. Biomater Res 2018; 22: 36.
[http://dx.doi.org/10.1186/s40824-018-0148-4] [PMID: 30598836]
[10]
Choi KA, Hong S. Induced neural stem cells as a means of treatment in Huntington’s disease. Expert Opin Biol Ther 2017; 17(11): 1333-43.
[http://dx.doi.org/10.1080/14712598.2017.1365133] [PMID: 28792249]
[11]
Tartarini D, Mele E. Adult stem cell therapies for wound healing: Biomaterials and computational models. Front Bioeng Biotechnol 2016; 3: 206.
[http://dx.doi.org/10.3389/fbioe.2015.00206] [PMID: 26793702]
[12]
Barron CC, Lalu MM, Stewart DJ, et al. Assessment of safety and efficacy of mesenchymal stromal cell therapy in preclinical models of acute myocardial infarction: a systematic review protocol. Syst Rev 2017; 6(1): 226.
[http://dx.doi.org/10.1186/s13643-017-0601-9] [PMID: 29116020]
[13]
Luxán G, D’Amato G, MacGrogan D, de la Pompa JL. Endocardial notch signaling in cardiac development and disease. Circ Res 2016; 118(1): e1-e18.
[http://dx.doi.org/10.1161/CIRCRESAHA.115.305350] [PMID: 26635389]
[14]
Garside VC, Chang AC, Karsan A, Hoodless PA. Co-ordinating Notch, BMP, and TGF-β signaling during heart valve development. Cell Mol Life Sci 2013; 70(16): 2899-917.
[http://dx.doi.org/10.1007/s00018-012-1197-9] [PMID: 23161060]
[15]
Wagner M, Siddiqui MA. Signal transduction in early heart development (I): cardiogenic induction and heart tube formation. Exp Biol Med (Maywood) 2007; 232(7): 852-65.
[PMID: 17609501]
[16]
Wagner M, Siddiqui MA. Signal transduction in early heart development (II): ventricular chamber specification, trabeculation, and heart valve formation. Exp Biol Med (Maywood) 2007; 232(7): 866-80.
[PMID: 17609502]
[17]
Alfieri CM, Cheek J, Chakraborty S, Yutzey KE. Wnt signaling in heart valve development and osteogenic gene induction. Dev Biol 2010; 338(2): 127-35.
[http://dx.doi.org/10.1016/j.ydbio.2009.11.030] [PMID: 19961844]
[18]
Cambier L, Plate M, Sucov HM, Pashmforoush M. Nkx2-5 regulates cardiac growth through modulation of Wnt signaling by R-spondin3. Development 2014; 141(15): 2959-71.
[http://dx.doi.org/10.1242/dev.103416] [PMID: 25053429]
[19]
Pahnke A, Conant G, Huyer LD, Zhao Y, Feric N, Radisic M. The role of Wnt regulation in heart development, cardiac repair and disease: A tissue engineering perspective. Biochem Biophys Res Commun 2016; 473(3): 698-703.
[http://dx.doi.org/10.1016/j.bbrc.2015.11.060] [PMID: 26626076]
[20]
Buikema JW, Mady AS, Mittal NV, et al. Wnt/β-catenin signaling directs the regional expansion of first and second heart field-derived ventricular cardiomyocytes. Development 2013; 140(20): 4165-76.
[http://dx.doi.org/10.1242/dev.099325] [PMID: 24026118]
[21]
Ruan Z, Zhu L, Yin Y, Chen G. Overexpressing NKx2.5 increases the differentiation of human umbilical cord drived mesenchymal stem cells into cardiomyocyte-like cells. Biomed Pharmacother 2016; 78: 110-5.
[http://dx.doi.org/10.1016/j.biopha.2016.01.020] [PMID: 26898431]
[22]
Tang M, Yuan W, Bodmer R, Wu X, Ocorr K. The role of pygopus in the differentiation of intracardiac valves in Drosophila. Genesis 2014; 52(1): 19-28.
[http://dx.doi.org/10.1002/dvg.22724] [PMID: 24265259]
[23]
Belenkaya TY, Han C, Standley HJ, et al. pygopus Encodes a nuclear protein essential for wingless/Wnt signaling. Development 2002; 129(17): 4089-101.
[PMID: 12163411]
[24]
Akazawa H, Komuro I. Cardiac transcription factor Csx/Nkx2-5: Its role in cardiac development and diseases. Pharmacol Ther 2005; 107(2): 252-68.
[http://dx.doi.org/10.1016/j.pharmthera.2005.03.005] [PMID: 15925411]
[25]
George V, Colombo S, Targoff KL. An early requirement for nkx2.5 ensures the first and second heart field ventricular identity and cardiac function into adulthood. Dev Biol 2015; 400(1): 10-22.
[http://dx.doi.org/10.1016/j.ydbio.2014.12.019] [PMID: 25536398]
[26]
Skerjanc IS, Petropoulos H, Ridgeway AG, Wilton S. Myocyte enhancer factor 2C and Nkx2-5 up-regulate each other’s expression and initiate cardiomyogenesis in P19 cells. J Biol Chem 1998; 273(52): 34904-10.
[http://dx.doi.org/10.1074/jbc.273.52.34904] [PMID: 9857019]
[27]
Tong YF. Mutations of NKX2.5 and GATA4 genes in the development of congenital heart disease. Gene 2016; 588(1): 86-94.
[http://dx.doi.org/10.1016/j.gene.2016.04.061] [PMID: 27154817]
[28]
Hoffmans R, Basler K. BCL9-2 binds Arm/beta-catenin in a Tyr142-independent manner and requires Pygopus for its function in Wg/Wnt signaling. Mech Dev 2007; 124(1): 59-67.
[http://dx.doi.org/10.1016/j.mod.2006.09.006] [PMID: 17113272]
[29]
Tang M, Yuan W, Fan X, et al. Pygopus maintains heart function in aging Drosophila independently of canonical Wnt signaling. Circ Cardiovasc Genet 2013; 6(5): 472-80.
[http://dx.doi.org/10.1161/CIRCGENETICS.113.000253] [PMID: 24046329]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy