Post-transcriptional Regulation of α-Synuclein Expression by mir-7 and mir-153

Genetic and biochemical studies have established a central role for α-synuclein accumulation in the pathogenesis of Parkinson disease. Here, two microRNAs, namely mir-7 and mir-153, have been identified to regulate α-synuclein levels post-transcriptionally. These microRNAs bind specifically to the 3′-untranslated region of α-synuclein and down-regulate its mRNA and protein levels, with their effect being additive. They are expressed predominantly in the brain with a pattern that mirrors synuclein expression in different tissues as well as during neuronal development, indicating that they play a tuning role in the amount of α-synuclein produced. Overexpression of mir-7 and mir-153 significantly reduces endogenous α-synuclein levels, whereas inhibition of mir-7 and mir-153 enhances translation of a luciferase construct bearing the α-synuclein 3′-untranslated region in primary neurons. These findings reveal a significant additional mechanism by which α-synuclein is regulated and point toward new therapeutic regimes for lowering endogenous α-synuclein levels in patients with familial or sporadic Parkinson disease.

[1]  N. Rajewsky,et al.  Cell-type-specific signatures of microRNAs on target mRNA expression. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[2]  Jan Krüger,et al.  RNAhybrid: microRNA target prediction easy, fast and flexible , 2006, Nucleic Acids Res..

[3]  P. Bork,et al.  mRNA degradation by miRNAs and GW182 requires both CCR4:NOT deadenylase and DCP1:DCP2 decapping complexes. , 2006, Genes & development.

[4]  T. Chase,et al.  Degradation of α-Synuclein by Proteasome* , 1999, The Journal of Biological Chemistry.

[5]  Janel O. Johnson,et al.  α-Synuclein Locus Triplication Causes Parkinson's Disease , 2003, Science.

[6]  John Q. Trojanowski,et al.  Induction of α-Synuclein Aggregation by Intracellular Nitrative Insult , 2001, The Journal of Neuroscience.

[7]  R. Miranda,et al.  Competing Interactions between Micro-RNAs Determine Neural Progenitor Survival and Proliferation after Ethanol Exposure: Evidence from an Ex Vivo Model of the Fetal Cerebral Cortical Neuroepithelium , 2007, The Journal of Neuroscience.

[8]  P. Rudolph,et al.  Simultaneous introduction of multiple mutations using overlap extension PCR. , 1997, BioTechniques.

[9]  M. McPeek,et al.  MicroRNAs and metazoan macroevolution: insights into canalization, complexity, and the Cambrian explosion , 2009, BioEssays : news and reviews in molecular, cellular and developmental biology.

[10]  K. Grant,et al.  Peripheral blood alpha-synuclein mRNA levels are elevated in cynomolgus monkeys that chronically self-administer ethanol. , 2006, Alcohol.

[11]  J. Kordower,et al.  Age-associated increases of α-synuclein in monkeys and humans are associated with nigrostriatal dopamine depletion: Is this the target for Parkinson's disease? , 2007, Neurobiology of Disease.

[12]  R. Plasterk,et al.  MicroRNA function in animal development , 2005, FEBS letters.

[13]  D. Bartel,et al.  Micromanagers of gene expression: the potentially widespread influence of metazoan microRNAs , 2004, Nature Reviews Genetics.

[14]  A. Silahtaroglu,et al.  Antagonism of microRNA-122 in mice by systemically administered LNA-antimiR leads to up-regulation of a large set of predicted target mRNAs in the liver , 2007, Nucleic acids research.

[15]  Justin J. Cassidy,et al.  A MicroRNA Imparts Robustness against Environmental Fluctuation during Development , 2009, Cell.

[16]  E. Lai miRNAs: Whys and Wherefores of miRNA-Mediated Regulation , 2005, Current Biology.

[17]  Takeshi Iwatsubo,et al.  Fatal attractions: abnormal protein aggregation and neuron death in Parkinson's disease and Lewy body dementia , 1998, Cell Death and Differentiation.

[18]  Jeremy N. Skepper,et al.  α-Synuclein Is Degraded by Both Autophagy and the Proteasome* , 2003, Journal of Biological Chemistry.

[19]  J. Rehwinkel,et al.  MicroRNAs silence gene expression by repressing protein expression and/or by promoting mRNA decay. , 2006, Cold Spring Harbor symposia on quantitative biology.

[20]  Brian S. Roberts,et al.  Simple, quantitative primer-extension PCR assay for direct monitoring of microRNAs and short-interfering RNAs. , 2005, RNA.

[21]  L. Lim,et al.  MicroRNA targeting specificity in mammals: determinants beyond seed pairing. , 2007, Molecular cell.

[22]  Kostas Vekrellis,et al.  Wild Type α-Synuclein Is Degraded by Chaperone-mediated Autophagy and Macroautophagy in Neuronal Cells* , 2008, Journal of Biological Chemistry.

[23]  Noam Shomron,et al.  Canalization of development by microRNAs , 2006, Nature Genetics.

[24]  H. Inoko,et al.  Multiple candidate gene analysis identifies alpha-synuclein as a susceptibility gene for sporadic Parkinson's disease. , 2006, Human molecular genetics.

[25]  Sonja W. Scholz,et al.  Genome-Wide Association Study reveals genetic risk underlying Parkinson’s disease , 2009, Nature Genetics.

[26]  Alexander F. Schier,et al.  Differential Regulation of Germline mRNAs in Soma and Germ Cells by Zebrafish miR-430 , 2006, Current Biology.

[27]  E. Huang,et al.  Homeodomain-Interacting Protein Kinase-2 Regulates Apoptosis in Developing Sensory and Sympathetic Neurons , 2004, Current Biology.

[28]  K. Gunsalus,et al.  Combinatorial microRNA target predictions , 2005, Nature Genetics.

[29]  K. Livak,et al.  Multiplexing RT-PCR for the detection of multiple miRNA species in small samples. , 2006, Biochemical and biophysical research communications.

[30]  Anton J. Enright,et al.  MicroRNA targets in Drosophila , 2003, Genome Biology.

[31]  N. Rajewsky,et al.  Silencing of microRNAs in vivo with ‘antagomirs’ , 2005, Nature.

[32]  H. Hausen,et al.  Conserved Sensory-Neurosecretory Cell Types in Annelid and Fish Forebrain: Insights into Hypothalamus Evolution , 2007, Cell.

[33]  S. Kauppinen,et al.  LNA-mediated microRNA silencing in non-human primates , 2008, Nature.

[34]  Elisa Izaurralde,et al.  Deadenylation is a widespread effect of miRNA regulation. , 2008, RNA.

[35]  R. Crowther,et al.  α-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with Lewy bodies , 1998 .

[36]  M. Farrer,et al.  α-Synuclein and Parkinson disease susceptibility , 2007, Neurology.