DOI QR코드

DOI QR Code

β-arrestin2 Affects Cardiac Progenitor Cell Survival through Cell Mobility and Tube Formation in Severe Hypoxia

  • Seo, Seul-ki (Department of Internal Medicine, Chungbuk National University College of Medicine) ;
  • Kim, Nari (Department of Internal Medicine, Chungbuk National University College of Medicine) ;
  • Lee, Ju-Hee (Chungbuk Regional Cardiocerebrovascular Center, Chungbuk National University Hospital) ;
  • Kim, Sang Min (Chungbuk Regional Cardiocerebrovascular Center, Chungbuk National University Hospital) ;
  • Lee, Sang Yeub (Department of Internal Medicine, Chungbuk National University College of Medicine) ;
  • Bae, Jang-Whan (Department of Internal Medicine, Chungbuk National University College of Medicine) ;
  • Hwang, Kyung-Kuk (Department of Internal Medicine, Chungbuk National University College of Medicine) ;
  • Kim, Dong-Woon (Department of Internal Medicine, Chungbuk National University College of Medicine) ;
  • Koch, Walter J. (Center for Translational Medicine, Department of Pharmacology, Temple University Lewis Katz School of Medicine) ;
  • Cho, Myeong-Chan (Department of Internal Medicine, Chungbuk National University College of Medicine)
  • Received : 2017.06.06
  • Accepted : 2018.01.17
  • Published : 2018.04.06

Abstract

Background and Objectives: ${\beta}$-arrestin2 (${\beta}$-arr2) basically regulates multiple signaling pathways in mammalian cells by desensitization and internalization of G-protein coupled receptors (GPCRs). We investigated impacts of ${\beta}$-arr2 on survival, mobility, and tube formation of cardiac progenitor cells (CPCs) obtained from wild-type (WT) mouse (CPC-WT), and ${\beta}$-arr2 knock-out (KO) mouse (CPC-KO) cultured in presence or absence of serum and oxygen as non-canonical roles in GPCR system. Methods: CPCs were cultured in Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12-based media containing fetal bovine serum and growth factors. Survival of 2 types of CPCs in hypoxia and/or serum deprivation was measured by fluorescence-activated cell sorting. Wound healing ability, and tube formation ability on Matrigel of 2 kinds of CPCs were compared in normoxic and hypoxic cultures. Protein expression related to survival and mobility were measured with the Western blot for each culture conditions. Result: CPC-KO showed significantly worse mobility in the wound healing assay and in tube formation on Matrigel especially in hypoxic culture than did the CPC-WT. Also, CPC-KO showed significantly higher apoptosis fraction in both normoxic and hypoxic cultures than did the CPC-WT. Expression of proteins associated with cell survival and mobility, e.g., protein kinase B (Akt), ${\beta}$-catenin, and glycogen synthase kinase-$3{\beta}$ (GSK-$3{\beta}$) was significantly worse in CPC-KO. Conclusions: The CPC-KO had significantly worse cell mobility, tube formation ability, and survival than the CPC-WT, especially in the hypoxic cultures. Apparently, ${\beta}$-arr2 is important on CPC survival by means of mobility and tube formation in myocardial ischemia.

Keywords

References

  1. Youn JC, Han S, Ryu KH. Temporal trends of hospitalized patients with heart failure in Korea. Korean Circ J 2017;47:16-24. https://doi.org/10.4070/kcj.2016.0429
  2. Lee JH, Lim NK, Cho MC, Park HY. Epidemiology of heart failure in Korea: present and future. Korean Circ J 2016;46:658-64. https://doi.org/10.4070/kcj.2016.46.5.658
  3. Liang SX, Phillips WD. Migration of resident cardiac stem cells in myocardial infarction. Anat Rec (Hoboken) 2013;296:184-91. https://doi.org/10.1002/ar.22633
  4. Beltrami AP, Barlucchi L, Torella D, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 2003;114:763-76. https://doi.org/10.1016/S0092-8674(03)00687-1
  5. Kuang D, Zhao X, Xiao G, et al. Stem cell factor/c-kit signaling mediated cardiac stem cell migration via activation of p38 MAPK. Basic Res Cardiol 2008;103:265-73. https://doi.org/10.1007/s00395-007-0690-z
  6. Lefkowitz RJ, Rajagopal K, Whalen EJ. New roles for ${\beta}$-arrestins in cell signaling: not just for seventransmembrane receptors. Mol Cell 2006;24:643-52. https://doi.org/10.1016/j.molcel.2006.11.007
  7. DeFea KA. Arrestins in actin reorganization and cell migration. Prog Mol Biol Transl Sci 2013;118:205-22.
  8. Yang P, Li Z, Wang Y, Zhang L, Wu H, Li Z. Secreted pyruvate kinase M2 facilitates cell migration via PI3K/Akt and Wnt/${\beta}$-catenin pathway in colon cancer cells. Biochem Biophys Res Commun 2015;459:327-32. https://doi.org/10.1016/j.bbrc.2015.02.112
  9. Traynham CJ, Hullmann J, Koch WJ. "Canonical and non-canonical actions of GRK5 in the heart". J Mol Cell Cardiol 2016;92:196-202. https://doi.org/10.1016/j.yjmcc.2016.01.027
  10. Bohn LM, Lefkowitz RJ, Gainetdinov RR, Peppel K, Caron MG, Lin FT. Enhanced morphine analgesia in mice lacking beta-arrestin 2. Science 1999;286:2495-8. https://doi.org/10.1126/science.286.5449.2495
  11. Attramadal H, Arriza JL, Aoki C, et al. Beta-arrestin2, a novel member of the arrestin/beta-arrestin gene family. J Biol Chem 1992;267:17882-90.
  12. Fransioli J, Bailey B, Gude NA, et al. Evolution of the c-kit-positive cell response to pathological challenge in the myocardium. Stem Cells 2008;26:1315-24. https://doi.org/10.1634/stemcells.2007-0751
  13. Chun WJ, Nah DY, Bae JH, Chung JW, Lee H, Moon IS. Glucose-insulin-potassium solution protects ventricular myocytes of neonatal rat in an in vitro coverslip ischemia/reperfusion model. Korean Circ J 2015;45:234-41. https://doi.org/10.4070/kcj.2015.45.3.234
  14. Naaldijk Y, Johnson AA, Ishak S, Meisel HJ, Hohaus C, Stolzing A. Migrational changes of mesenchymal stem cells in response to cytokines, growth factors, hypoxia, and aging. Exp Cell Res 2015;338:97-104. https://doi.org/10.1016/j.yexcr.2015.08.019
  15. Park KJ, Kim YJ, Choi EJ, et al. Expression pattern of the thioredoxin system in human endothelial progenitor cells and endothelial cells under hypoxic injury. Korean Circ J 2010;40:651-8. https://doi.org/10.4070/kcj.2010.40.12.651
  16. Takeda N, Manabe I. Cellular interplay between cardiomyocytes and nonmyocytes in cardiac remodeling. Int J Inflamm 2011;2011:535241.
  17. Williams AR, Hatzistergos KE, Addicott B, et al. Enhanced effect of combining human cardiac stem cells and bone marrow mesenchymal stem cells to reduce infarct size and to restore cardiac function after myocardial infarction. Circulation 2013;127:213-23. https://doi.org/10.1161/CIRCULATIONAHA.112.131110
  18. Makkar RR, Smith RR, Cheng K, et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet 2012;379:895-904. https://doi.org/10.1016/S0140-6736(12)60195-0
  19. van Berlo JH, Kanisicak O, Maillet M, et al. c-kit+ cells minimally contribute cardiomyocytes to the heart. Nature 2014;509:337-41. https://doi.org/10.1038/nature13309
  20. Luttrell LM, Lefkowitz RJ. The role of beta-arrestins in the termination and transduction of G-proteincoupled receptor signals. J Cell Sci 2002;115:455-65.
  21. Buchanan FG, DuBois RN. Emerging roles of beta-arrestins. Cell Cycle 2006;5:2060-3. https://doi.org/10.4161/cc.5.18.3212
  22. Walker JK, Fong AM, Lawson BL, et al. ${\beta}$-arrestin-2 regulates the development of allergic asthma. J Clin Invest 2003;112:566-74. https://doi.org/10.1172/JCI200317265
  23. Ge L, Ly Y, Hollenberg M, DeFea K. A beta-arrestin-dependent scaffold is associated with prolonged MAPK activation in pseudopodia during protease-activated receptor-2-induced chemotaxis. J Biol Chem 2003;278:34418-26. https://doi.org/10.1074/jbc.M300573200
  24. McCubrey JA, Steelman LS, Bertrand FE, et al. Multifaceted roles of GSK-3 and Wnt/${\beta}$-catenin in hematopoiesis and leukemogenesis: opportunities for therapeutic intervention. Leukemia 2014;28:15-33. https://doi.org/10.1038/leu.2013.184
  25. Reya T, Clevers H. Wnt signalling in stem cells and cancer. Nature 2005;434:843-50. https://doi.org/10.1038/nature03319
  26. Mannoury la Cour C, Salles MJ, Pasteau V, Millan MJ. Signaling pathways leading to phosphorylation of Akt and GSK-$3{\beta}$ by activation of cloned human and rat cerebral $D_2$ and $D_3$ receptors. Mol Pharmacol 2011;79:91-105. https://doi.org/10.1124/mol.110.065409
  27. Krock BL, Skuli N, Simon MC. Hypoxia-induced angiogenesis: good and evil. Genes Cancer 2011;2:1117-33. https://doi.org/10.1177/1947601911423654
  28. Zou L, Yang R, Chai J, Pei G. Rapid xenograft tumor progression in beta-arrestin1 transgenic mice due to enhanced tumor angiogenesis. FASEB J 2008;22:355-64. https://doi.org/10.1096/fj.07-9046com
  29. Zhang T, Lee YW, Rui YF, Cheng TY, Jiang XH, Li G. Bone marrow-derived mesenchymal stem cells promote growth and angiogenesis of breast and prostate tumors. Stem Cell Res Ther 2013;4:70. https://doi.org/10.1186/scrt221
  30. Takeda K, Sowa Y, Nishino K, Itoh K, Fushiki S. Adipose-derived stem cells promote proliferation, migration, and tube formation of lymphatic endothelial cells in vitro by secreting lymphangiogenic factors. Ann Plast Surg 2015;74:728-36. https://doi.org/10.1097/SAP.0000000000000084

Cited by

  1. Generation of directly reprogrammed human endothelial cells derived from fibroblast using ultrasound vol.126, pp.None, 2018, https://doi.org/10.1016/j.yjmcc.2018.11.016
  2. The Role of β-Arrestins in Regulating Stem Cell Phenotypes in Normal and Tumorigenic Cells vol.21, pp.23, 2020, https://doi.org/10.3390/ijms21239310
  3. β-Arrestin2 deficiency attenuates oxidative stress in mouse hepatic fibrosis through modulation of NOX4 vol.42, pp.7, 2021, https://doi.org/10.1038/s41401-020-00545-9