1,518
Views
14
CrossRef citations to date
0
Altmetric
Research Paper

PAX3-FOXO1 escapes miR-495 regulation during muscle differentiation

, , , , &
Pages 144-153 | Received 27 Sep 2018, Accepted 19 Dec 2018, Published online: 11 Jan 2019

References

  • Yuan H, Qin F, Movassagh M, et al. A chimeric RNA characteristic of rhabdomyosarcoma in normal myogenesis process. Cancer Discov. 2013;3:1394–1403.
  • Ridgeway AG, Skerjanc IS. Pax3 is essential for skeletal myogenesis and the expression of Six1 and Eya2. J Biol Chem. 2001;276:19033–19039.
  • Bennicelli JL, Advani S, Schafer BW, et al. PAX3 and PAX7 exhibit conserved cis-acting transcription repression domains and utilize a common gain of function mechanism in alveolar rhabdomyosarcoma. Oncogene. 1999;18:4348–4356.
  • Fredericks WJ, Galili N, Mukhopadhyay S, et al. The PAX3-FKHR fusion protein created by the t(2;13) translocation in alveolar rhabdomyosarcomas is a more potent transcriptional activator than PAX3. Mol Cell Biol. 1995;15:1522–1535.
  • Cao L, Yu Y, Bilke S, et al. Genome-wide identification of PAX3-FKHR binding sites in rhabdomyosarcoma reveals candidate target genes important for development and cancer. Cancer Res. 2010;70:6497–6508.
  • Graf Finckenstein F, Shahbazian V, Davicioni E, et al. PAX-FKHR function as pangenes by simultaneously inducing and inhibiting myogenesis. Oncogene. 2008;27:2004–2014.
  • Khan J, Bittner ML, Saal LH, et al. cDNA microarrays detect activation of a myogenic transcription program by the PAX3-FKHR fusion oncogene. Proc Natl Acad Sci U S A. 1999;96:13264–13269.
  • Kikuchi K, Tsuchiya K, Otabe O, et al. Effects of PAX3-FKHR on malignant phenotypes in alveolar rhabdomyosarcoma. Biochem Biophys Res Commun. 2008;365:568–574.
  • Crose LE, Galindo KA, Kephart JG, et al. Alveolar rhabdomyosarcoma-associated PAX3-FOXO1 promotes tumorigenesis via Hippo pathway suppression. J Clin Invest. 2014;124:285–296.
  • Linardic CM. PAX3-FOXO1 fusion gene in rhabdomyosarcoma. Cancer Lett. 2008;270:10–18.
  • Su X, Ling Y, Liu C, et al. Isolation, culture, differentiation, and nuclear reprogramming of Mongolian sheep fetal bone marrow-derived mesenchymal stem cells. Cell Reprogram. 2015;17:288–296.
  • Liu -Z-Z, Lin-Gai W, Xiao-Hu S, et al. Differentiation of Mongolia horse adipose tissue-derived mesenchymal stem cells into adipocytes and osteoblasts in Vitro. J South China Agric Univ. 2011;32:1.
  • Ling YH, Sui MH, Zheng Q, et al. miR-27b regulates myogenic proliferation and differentiation by targeting Pax3 in goat. Sci Rep. 2018;8:3909.
  • Beillard E, Ong SC, Giannakakis A, et al. miR-Sens–a retroviral dual-luciferase reporter to detect microRNA activity in primary cells. Rna. 2012;18:1091–1100.
  • Babiceanu M, Qin F, Xie Z, et al. Recurrent chimeric fusion RNAs in non-cancer tissues and cells. Nucleic Acids Res. 2016;44:2859–2872.
  • Magrangeas F, Pitiot G, Dubois S, et al. Cotranscription and intergenic splicing of human galactose-1-phosphate uridylyltransferase and interleukin-11 receptor alpha-chain genes generate a fusion mRNA in normal cells. Implication for the production of multidomain proteins during evolution. J Biol Chem. 1998;273:16005–16010.
  • Frenkel-Morgenstern M, Lacroix V, Ezkurdia I, et al. Chimeras taking shape: potential functions of proteins encoded by chimeric RNA transcripts. Genome Res. 2012;22:1231–1242.
  • Carrara M, Beccuti M, Cavallo F, et al. State of art fusion-finder algorithms are suitable to detect transcription-induced chimeras in normal tissues? BMC Bioinformatics. 2013;14(Suppl 7):S2.
  • Finta C, Zaphiropoulos PG. Intergenic mRNA molecules resulting from trans-splicing. J Biol Chem. 2002;277:5882–5890.
  • Chase A, Ernst T, Fiebig A, et al. TFG, a target of chromosome translocations in lymphoma and soft tissue tumors, fuses to GPR128 in healthy individuals. Haematologica. 2010;95:20–26.
  • Qin F, Song Z, Babiceanu M, et al. Discovery of CTCF-Sensitive Cis-Spliced fusion RNAs between adjacent genes in human prostate cells. PLoS Genet. 2015;11:e1005001.
  • Wu CS, Yu CY, Chuang CY, et al. Integrative transcriptome sequencing identifies trans-splicing events with important roles in human embryonic stem cell pluripotency. Genome Res. 2014;24:25–36.
  • Li H, Ma X, Wang J, et al. Effects of rearrangement and allelic exclusion of JJAZ1/SUZ12 on cell proliferation and survival. Proc Natl Acad Sci U S A. 2007;104:20001–20006.
  • Gingeras TR. Implications of chimaeric non-co-linear transcripts. Nature. 2009;461:206–211.
  • Chuang TJ, Wu CS, Chen CY, et al. NCLscan: accurate identification of non-co-linear transcripts (fusion, trans-splicing and circular RNA) with a good balance between sensitivity and precision. Nucleic Acids Res. 2016;44:e29.
  • Chung EY, Dews M, Cozma D, et al. c-Myb oncoprotein is an essential target of the dleu2 tumor suppressor microRNA cluster. Cancer Biol Ther. 2008;7:1758–1764.
  • Zhao H, Kalota A, Jin S, et al. The c-myb proto-oncogene and microRNA-15a comprise an active autoregulatory feedback loop in human hematopoietic cells. Blood. 2009;113:505–516.
  • Persson M, Andren Y, Mark J, et al. Recurrent fusion of MYB and NFIB transcription factor genes in carcinomas of the breast and head and neck. Proc Natl Acad Sci USA. 2009;106:18740–18744.
  • Parker BC, Annala MJ, Cogdell DE, et al. The tumorigenic FGFR3-TACC3 gene fusion escapes miR-99a regulation in glioblastoma. J Clin Invest. 2013;123:855–865.
  • Snyder CM, Rice AL, Estrella NL, et al. MEF2A regulates the Gtl2-Dio3 microRNA mega-cluster to modulate WNT signaling in skeletal muscle regeneration. Development. 2013;140:31–42.
  • Clark AL, Naya FJ. MicroRNAs in the myocyte enhancer factor 2 (MEF2)-regulated Gtl2-Dio3 noncoding RNA locus promote cardiomyocyte proliferation by targeting the transcriptional coactivator cited2. J Biol Chem. 2015;290:23162–23172.
  • Muir S, Nathanson J, Wilbert M, et al. The role of miRNA in PAX3-FKHR positive rhabdomyosarcoma. Cancer Res. 2014;74(19 Suppl):Abstract nr 3558.
  • Modrek B, Lee CJ. Alternative splicing in the human, mouse and rat genomes is associated with an increased frequency of exon creation and/or loss. Nat Genet. 2003;34:177–180.
  • Nurtdinov RN, Artamonova II, Mironov AA, et al. Low conservation of alternative splicing patterns in the human and mouse genomes. Hum Mol Genet. 2003;12:1313–1320.
  • Pan Q, Bakowski MA, Morris Q, et al. Alternative splicing of conserved exons is frequently species-specific in human and mouse. Trends Genet. 2005;21:73–77.
  • Yeo GW, Van Nostrand E, Holste D, et al. Identification and analysis of alternative splicing events conserved in human and mouse. Proc Natl Acad Sci U S A. 2005;102:2850–2855.
  • Chuang TJ, Chen YJ, Chen CY, et al. Integrative transcriptome sequencing reveals extensive alternative trans-splicing and cis-backsplicing in human cells. Nucleic Acids Res. 2018;46:3671–3691.
  • Qin P, Parlak M, Kuscu C, et al. Live cell imaging of low- and non-repetitive chromosome loci using CRISPR-Cas9. Nat Commun. 2017;8:14725.

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.