695
Views
0
CrossRef citations to date
0
Altmetric
Research Paper

Translation of Tudor-SN, a novel terminal oligo-pyrimidine (TOP) mRNA, is regulated by the mTORC1 pathway in cardiomyocytes

, , , , ORCID Icon, , , , , & show all
Pages 900-913 | Received 08 May 2020, Accepted 21 Sep 2020, Published online: 15 Oct 2020

References

  • Yang J, Aittomaki S, Pesu M, et al. Identification of p100 as a coactivator for STAT6 that bridges STAT6 with RNA polymerase II. Embo J. 2002;21:4950–4958.
  • Yang J, Valineva T, Hong J, et al. Transcriptional co-activator protein p100 interacts with snRNP proteins and facilitates the assembly of the spliceosome. Nucleic Acids Res. 2007;35:4485–4494.
  • Fu X, Zhang C, Meng H, et al. Oncoprotein Tudor-SN is a key determinant providing survival advantage under DNA damaging stress. Cell Death Differ. 2018;25:1625–1637.
  • Su C, Gao X, Yang W, et al. Phosphorylation of Tudor-SN, a novel substrate of JNK, is involved in the efficient recruitment of Tudor-SN into stress granules. Biochim Biophys Acta Mol Cell Res. 2017;1864:562–571.
  • Su C, Zhang C, Tecle A, et al. Tudor staphylococcal nuclease (Tudor-SN), a novel regulator facilitating G1/S phase transition, acting as a co-activator of E2F-1 in cell cycle regulation. J Biol Chem. 2015;290:7208–7220.
  • Armengol S, Arretxe E, Rodriguez L, et al. NF-kappaB, Sp1 and NF-Y as transcriptional regulators of human SND1 gene. Biochimie. 2013;95:735–742.
  • Porrello ER, Mahmoud AI, Simpson E, et al. Transient regenerative potential of the neonatal mouse heart. Science. 2011;331:1078–1080.
  • Walsh S, Ponten A, Fleischmann BK, et al. Cardiomyocyte cell cycle control and growth estimation in vivo–an analysis based on cardiomyocyte nuclei. Cardiovasc Res. 2010;86:365–373.
  • Bergmann O, Bhardwaj RD, Bernard S, et al. Evidence for cardiomyocyte renewal in humans. Science. 2009;324:98–102.
  • Ali SR, Hippenmeyer S, Saadat LV, et al. Existing cardiomyocytes generate cardiomyocytes at a low rate after birth in mice. Proc Natl Acad Sci U S A. 2014;111:8850–8855.
  • Senyo SE, Steinhauser ML, Pizzimenti CL, et al. Mammalian heart renewal by pre-existing cardiomyocytes. Nature. 2013;493:433–436.
  • Ahuja P, Sdek P, MacLellan WR. Cardiac myocyte cell cycle control in development, disease, and regeneration. Physiol Rev. 2007;87:521–544.
  • Zhou Q, Li L, Zhao B, et al. The hippo pathway in heart development, regeneration, and diseases. Circ Res. 2015;116:1431–1447.
  • Mahmoud AI, Kocabas F, Muralidhar SA, et al. Meis1 regulates postnatal cardiomyocyte cell cycle arrest. Nature. 2013;497:249–253.
  • Mohamed TMA, Ang YS, Radzinsky E, et al. Regulation of cell cycle to stimulate adult cardiomyocyte proliferation and cardiac regeneration. Cell. 2018;173:104–116 e112.
  • Lin Z, von Gise A, Zhou P, et al. Cardiac-specific YAP activation improves cardiac function and survival in an experimental murine MI model. Circ Res. 2014;115:354–363.
  • Vagnozzi RJ, Molkentin JD, Houser SR. New myocyte formation in the adult heart: endogenous sources and therapeutic implications. Circ Res. 2018;123:159–176.
  • Ran FA, Hsu PD, Lin CY, et al. Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell. 2013;154:1380–1389.
  • Gao X, Fu X, Song J, et al. Poly(A)(+) mRNA-binding protein Tudor-SN regulates stress granules aggregation dynamics. Febs J. 2015;282:874–890.
  • Yamashita R, Suzuki Y, Takeuchi N, et al. Comprehensive detection of human terminal oligo-pyrimidine (TOP) genes and analysis of their characteristics. Nucleic Acids Res. 2008;36:3707–3715.
  • Suzuki A, Wakaguri H, Yamashita R, et al. DBTSS as an integrative platform for transcriptome, epigenome and genome sequence variation data. Nucleic Acids Res. 2015;43:D87–91.
  • Thoreen CC, Chantranupong L, Keys HR, et al. A unifying model for mTORC1-mediated regulation of mRNA translation. Nature. 2012;485:109–113.
  • Nandagopal N, Roux PP. Regulation of global and specific mRNA translation by the mTOR signaling pathway. Translation. 2015;3:e983402.
  • Wang R, Su C, Wang X, et al. Global gene expression analysis combined with a genomics approach for the identification of signal transduction networks involved in postnatal mouse myocardial proliferation and development. Int J Mol Med. 2018;41:311–321.
  • Zhang D, Contu R, Latronico MV, et al. MTORC1 regulates cardiac function and myocyte survival through 4E-BP1 inhibition in mice. J Clin Invest. 2010;120:2805–2816.
  • Hamilton TL, Stoneley M, Spriggs KA, et al. TOPs and their regulation. Biochem Soc Trans. 2006;34:12–16.
  • Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017;169:361–371.
  • Sciarretta S, Volpe M, Sadoshima J. Mammalian target of rapamycin signaling in cardiac physiology and disease. Circ Res. 2014;114:549–564.
  • Heallen TR, Kadow ZA, Wang J, et al. Determinants of cardiac growth and size. Cold Spring Harb Perspect Biol. 2020;12:a037150.
  • Sedmera D, Thompson RP. Myocyte proliferation in the developing heart. Dev Dyn. 2011;240:1322–1334.
  • Takeuchi T. Regulation of cardiomyocyte proliferation during development and regeneration. Dev Growth Differ. 2014;56:402–409.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.