Research Article

Upregulation of miR-25 and miR-181 Family Members Correlates with Reduced Expression of ATXN3 in Lymphocytes from SCA3 Patients

Author(s): Sybille Krauss*, Rohit Nalavade, Stephanie Weber, Katlynn Carter and Bernd O. Evert*

Volume 8, Issue 1, 2019

Page: [76 - 85] Pages: 10

DOI: 10.2174/2211536607666180821162403

Abstract

Background: Spinocerebellar ataxia type 3 (SCA3), the most common spinocerebellar ataxia, is caused by a polyglutamine (polyQ) expansion in the protein ataxin-3 (ATXN3). Silencing the expression of polyQ-expanded ATXN3 rescues the cellular disease phenotype.

Objective: This study investigated the differential expression of microRNAs (miRNAs), small noncoding RNAs targeting gene expression, in lymphoblastoid cells (LCs) from SCA3 patients and the capability of identified deregulated miRNAs to target and alter ATXN3 expression.

Methods: MiRNA profiling was performed by microarray hybridization of total RNA from control and SCA3-LCs. The capability of the identified miRNAs and their target sites to suppress ATXN3 expression was analyzed using mutagenesis, reverse transcription PCR, immunoblotting, luciferase reporter assays, mimics and precursors of the identified miRNAs.

Results: SCA3-LCs showed significantly decreased expression levels of ATXN3 and a significant upregulation of the ATXN3-3’UTR targeting miRNAs, miR-32 and miR-181c and closely related members of the miR-25 and miR-181 family, respectively. MiR-32 and miR-181c effectively targeted the 3’UTR of ATXN3 and suppressed the expression of ATXN3.

Conclusions: The simultaneous upregulation of closely related miRNAs targeting the 3’UTR of ATXN3 and the significantly reduced ATXN3 expression levels in SCA3-LCs suggests that miR-25 and miR-181 family members cooperatively bind to the 3’UTR to suppress the expression of ATXN3. The findings further suggest that the upregulation of miR-25 and miR-181 family members in SCA3- LCs reflects a cell type-specific, protective mechanism to diminish polyQ-mediated cytotoxic effects. Thus, miRNA mimics of miR-25 and miR-181 family members may prove useful for the treatment of SCA3.

Keywords: Ataxin-3, Machado-Joseph disease, miRNA, polyglutamine diseases, protein expression, spinocerebellar ataxia type 3.

Graphical Abstract
[1]
Costa Mdo C, Paulson HL. Toward understanding Machado-Joseph disease. Prog Neurobiol 2012; 97(2): 239-57.
[2]
Kawaguchi Y, Okamoto T, Taniwaki M, et al. CAG expansions in a novel gene for Machado-Joseph disease at chromosome 14q32.1. Nat Genet 1994; 8(3): 221-8.
[3]
Lima M, Costa MC, Montiel R, et al. Population genetics of wild-type CAG repeats in the Machado-Joseph disease gene in Portugal. Hum Hered 2005; 60(3): 156-63.
[4]
Maciel P, Costa MC, Ferro A, et al. Improvement in the molecular diagnosis of Machado-Joseph disease. Arch Neurol 2001; 58(11): 1821-7.
[5]
Burnett B, Li F, Pittman RN. The polyglutamine neurodegenerative protein ataxin-3 binds polyubiquitylated proteins and has ubiquitin protease activity. Hum Mol Genet 2003; 12(23): 3195-205.
[6]
Chai Y, Berke SS, Cohen RE, Paulson HL. Poly-ubiquitin binding by the polyglutamine disease protein ataxin-3 links its normal function to protein surveillance pathways. J Biol Chem 2004; 279(5): 3605-11.
[7]
Araujo J, Breuer P, Dieringer S, et al. FOXO4-dependent upregulation of superoxide dismutase-2 in response to oxidative stress is impaired in spinocerebellar ataxia type 3. Hum Mol Genet 2011; 20(15): 2928-41.
[8]
Sowa ME, Bennett EJ, Gygi SP, Harper JW. Defining the human deubiquitinating enzyme interaction landscape. Cell 2009; 138(2): 389-403.
[9]
Rodrigues AJ, Coppola G, Santos C, et al. Functional genomics and biochemical characterization of the C. elegans orthologue of the Machado-Joseph disease protein ataxin-3. FASEB J 2007; 21(4): 1126-36.
[10]
Schmitt I, Linden M, Khazneh H, et al. Inactivation of the mouse Atxn3 (ataxin-3) gene increases protein ubiquitination. Biochem Biophys Res Commun 2007; 362(3): 734-9.
[11]
Bichelmeier U, Schmidt T, Hubener J, et al. Nuclear localization of ataxin-3 is required for the manifestation of symptoms in SCA3: in vivo evidence. J Neurosci 2007; 27(28): 7418-28.
[12]
Mueller T, Breuer P, Schmitt I, Walter J, Evert BO, Wullner U. CK2-dependent phosphorylation determines cellular localization and stability of ataxin-3. Hum Mol Genet 2009; 18(17): 3334-43.
[13]
Antony PM, Mantele S, Mollenkopf P, et al. Identification and functional dissection of localization signals within ataxin-3. Neurobiol Dis 2009; 36(2): 280-92.
[14]
Macedo-Ribeiro S, Cortes L, Maciel P, Carvalho AL. Nucleocytoplasmic shuttling activity of ataxin-3. PLoS One 2009; 4(6): e5834.
[15]
Teixeira-Castro A, Ailion M, Jalles A, et al. Neuron-specific proteotoxicity of mutant ataxin-3 in C. elegans: rescue by the DAF-16 and HSF-1 pathways. Hum Mol Genet 2011; 20(15): 2996-3009.
[16]
Koch P, Breuer P, Peitz M, et al. Excitation-induced ataxin-3 aggregation in neurons from patients with Machado-Joseph disease. Nature 2011; 480(7378): 543-6.
[17]
Simoes AT, Goncalves N, Koeppen A, et al. Calpastatin-mediated inhibition of calpains in the mouse brain prevents mutant ataxin 3 proteolysis, nuclear localization and aggregation, relieving Machado-Joseph disease. Brain 2012; 135(Pt 8): 2428-39.
[18]
Takahashi T, Katada S, Onodera O. Polyglutamine diseases: where does toxicity come from? what is toxicity? where are we going? J Mol Cell Biol 2010; 2(4): 180-91.
[19]
Alves S, Nascimento-Ferreira I, Auregan G, et al. Allele-specific RNA silencing of mutant ataxin-3 mediates neuroprotection in a rat model of Machado-Joseph disease. PLoS One 2008; 3(10): e3341.
[20]
Alves S, Nascimento-Ferreira I, Dufour N, et al. Silencing ataxin-3 mitigates degeneration in a rat model of Machado-Joseph disease: no role for wild-type ataxin-3? Hum Mol Genet 2010; 19(12): 2380-94.
[21]
Rodriguez-Lebron E, Costa M, Luna-Cancalon K, et al. Silencing mutant ATXN3 expression resolves molecular phenotypes in SCA3 transgenic mice. Mol Ther 2013; 21(10): 1909-18.
[22]
Goodall EF, Heath PR, Bandmann O, Kirby J, Shaw PJ. Neuronal dark matter: the emerging role of microRNAs in neurodegeneration. Front Cell Neurosci 2013; 7: 178.
[23]
Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009; 136(2): 215-33.
[24]
Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005; 120(1): 15-20.
[25]
Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB. Prediction of mammalian microRNA targets. Cell 2003; 115(7): 787-98.
[26]
Sonntag KC. MicroRNAs and deregulated gene expression networks in neurodegeneration. Brain Res 2010; 1338: 48-57.
[27]
Persengiev S, Kondova I, Otting N, Koeppen AH, Bontrop RE. Genome-wide analysis of miRNA expression reveals a potential role for miR-144 in brain aging and spinocerebellar ataxia pathogenesis. Neurobiol Aging 2011; 32(12): 2316.e17-27.
[http://dx.doi.org/10.1016/j.neurobiolaging.2010.03.014]
[28]
Lee ST, Chu K, Im WS, et al. Altered microRNA regulation in Huntington’s disease models. Exp Neurol 2011; 227(1): 172-9.
[29]
Lee Y, Samaco RC, Gatchel JR, Thaller C, Orr HT, Zoghbi HY. miR-19, miR-101 and miR-130 co-regulate ATXN1 levels to potentially modulate SCA1 pathogenesis. Nat Neurosci 2008; 11(10): 1137-9.
[30]
Huang F, Zhang L, Long Z, et al. miR-25 alleviates polyQ-mediated cytotoxicity by silencing ATXN3. FEBS Lett 2014; 588(24): 4791-8.
[31]
Koscianska E, Krzyzosiak WJ. Current understanding of the role of microRNAs in spinocerebellar ataxias. Cerebellum Ataxias 2014; 1: 7.
[32]
Carmona V, Cunha-Santos J, Onofre I, et al. Unravelling endogenous microRNA system dysfunction as a new pathophysiological mechanism in Machado-Joseph disease. Mol Ther 2017; 25(4): 1038-55.
[33]
Bilen J, Liu N, Burnett BG, Pittman RN, Bonini NM. MicroRNA pathways modulate polyglutamine-induced neurodegeneration. Mol Cell 2006; 24(1): 157-63.
[34]
Stappert L, Borghese L, Roese-Koerner B, et al. MicroRNA-based promotion of human neuronal differentiation and subtype specification. PLoS One 2013; 8(3): e59011.
[35]
Saetrom P, Heale BS, Snove O Jr, Aagaard L, Alluin J, Rossi JJ. Distance constraints between microRNA target sites dictate efficacy and cooperativity. Nucleic Acids Res 2007; 35(7): 2333-42.
[36]
Denzler R, McGeary SE, Title AC, Agarwal V, Bartel DP, Stoffel M. Impact of microrna levels, target-site complementarity, and cooperativity on competing endogenous RNA-regulated gene expression. Mol Cell 2016; 64(3): 565-79.
[37]
Wienholds E, Plasterk RH. MicroRNA function in animal development. FEBS Lett 2005; 579(26): 5911-22.
[38]
Landgraf P, Rusu M, Sheridan R. al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 2007; 129(7): 1401-14.
[39]
Gurtner A, Falcone E, Garibaldi F, Piaggio G. Dysregulation of microRNA biogenesis in cancer: the impact of mutant p53 on Drosha complex activity. J Exp Clin Cancer Res 2016; 35: 45.
[40]
Onofre I, Mendonca N, Lopes S, et al. Fibroblasts of Machado Joseph disease patients reveal autophagy impairment. Sci Rep 2016; 6: 28220.
[41]
Shi Y, Huang F, Tang B, et al. MicroRNA profiling in the serums of SCA3/MJD patients. Int J Neurosci 2014; 124(2): 97-101.
[42]
Wu W, Yang J, Feng X, et al. MicroRNA-32 (miR-32) regulates phosphatase and tensin homologue (PTEN) expression and promotes growth, migration, and invasion in colorectal carcinoma cells. Mol Cancer 2013; 12: 30.
[43]
Ji J, Yamashita T, Budhu A, et al. Identification of microRNA-181 by genome-wide screening as a critical player in EpCAM-positive hepatic cancer stem cells. Hepatology 2009; 50(2): 472-80.
[44]
Pichiorri F, Suh SS, Ladetto M, et al. MicroRNAs regulate critical genes associated with multiple myeloma pathogenesis. Proc Natl Acad Sci USA 2008; 105(35): 12885-90.
[45]
Zhang S, Chen H, Zhao X, et al. REV3L 3'UTR 460 T>C polymorphism in microRNA target sites contributes to lung cancer susceptibility. Oncogene 2013; 32(2): 242-50.
[46]
Wang L, Pal S, Sif S. Protein arginine methyltransferase 5 suppresses the transcription of the RB family of tumor suppressors in leukemia and lymphoma cells. Mol Cell Biol 2008; 28(20): 6262-77.
[47]
Ambs S, Prueitt RL, Yi M, et al. Genomic profiling of microRNA and messenger RNA reveals deregulated microRNA expression in prostate cancer. Cancer Res 2008; 68(15): 6162-70.
[48]
Suh SS, Yoo JY, Nuovo GJ, et al. MicroRNAs/TP53 feedback circuitry in glioblastoma multiforme. Proc Natl Acad Sci USA 2012; 109(14): 5316-21.

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