Publikationsserver der Universitätsbibliothek Marburg

Titel:Activity-dependent regulation of miRNAs in different subcellular compartments of neurons and its implications for neuronal morphogenesis and plasticity
Autor:Khudayberdiev, Sharof Abdumalikovich
Weitere Beteiligte: Schratt, Gerhard (Prof. Dr. )
Veröffentlicht:2014
URI:https://archiv.ub.uni-marburg.de/diss/z2014/0210
DOI: https://doi.org/10.17192/z2014.0210
URN: urn:nbn:de:hebis:04-z2014-02109
DDC: Naturwissenschaften
Titel (trans.):Aktivitätsabhängige Regulation von miRNAs in verschiedenen subzellulären Kompartimenten von Neuronen und deren Einfluß auf die neuronale Morphogenese und Plastizität
Publikationsdatum:2014-03-11
Lizenz:https://rightsstatements.org/vocab/InC-NC/1.0/

Dokument

Schlagwörter:
Nervenzelle, Transkriptionsfaktor, Dendrite, Neuron, Neuronal activity, Zellkern, Plasticity, Plastizität, Dendrit, Dendritogenese, microRNA, isomiR, miRNS, Dendritogenesis, Neuronale Aktivität, microRNA, Transcription factor, Neuronen, Nucleus, Dendriten

Summary:
The activity-dependent spatiotemporal regulation of gene expression in neurons is essential for the formation and function of neuronal circuits within the brain. Recently microRNAs, a new class of post-transcriptional regulators of gene expression were implicated in the regulation of neuronal differentiation and development. Furthermore, in mature fully developed neurons, miRNAs (e.g. miR-134) were shown to be involved in the control of local protein synthesis in the vicinity of dendritic spines (Schratt et al., 2006). Activity-dependent local protein synthesis is required for synaptic plasticity, which is believed to be one of the molecular substrates of learning and memory. Nonetheless, the molecular mechanisms underlying the function and regulation of miRNAs during synaptic plasticity are poorly understood. In a previous publication from our lab, it was shown that the activity of the brain-enriched miRNA - miR-134 is regulated by brain-derived neurotrophic factor, which is released upon synaptic stimulation in neurons (Schratt et al, 2006). Interestingly, in the mouse genome this miRNA is encoded in a large miRNA cluster (miR379-410 cluster) consisting of 39 miRNAs. The expression of the miR379-410 cluster is induced upon neuronal activity by virtue of myocyte-enhancing factor 2, a transcription factor that binds to a regulatory region upstream of this cluster (Fiore et al., 2009). The transcriptional upregulation of a subset of miRNAs from the miR379-410 cluster (miR-134, -381 and -329) is necessary for activity-dependent dendritic development of rat hippocampal neurons. Furthermore, we found that the post-transcriptional regulation of the RNA-binding protein Pumilio 2 by miR-134 is essential for activity-dependent dendritogenesis. Taken together, we defined a novel MEF2-miRNA-PUM2 pathway involved in the activity-dependent regulation of dendritogenesis in primary neurons. MiR-134 localizes within dendrites of hippocampal neurons, where it can regulate the local translation of proteins important for spine structure and plasticity. However, at the beginning of this project, it was unknown how this miRNA is targeted to dendrites. I was involved in a project that aimed at identifying and characterizing the transport mechanism of miR-134 to dendrites. We found that the dendritic localization of miR-134 is mediated by the DEAH-box helicase DHX36 protein, which binds to a cis-acting element located within the loop region of the miR-134 precursor (pre-miR-134; Bicker et al., 2013). Furthermore, we showed that depletion of DHX36 increased protein levels of LIM kinase 1, a dendritically localized target of miR-134 (Schratt et al, 2006). Moreover, the depletion of DHX36 led to an increase in dendritic spine size, a similar phenotype as observed upon inhibition of miR-134 activity. In summary, we described a novel mechanism for dendritic targeting of pre-miR-134 relevant for the function of miR-134 in spine morphogenesis. Activity-dependent regulation of gene expression in the nucleus is important for the development and function of the nervous system, including synaptic plasticity and memory formation. Interestingly, several recent reports suggested that miRNAs (and/or siRNAs) might be involved in the regulation of epigenetic modifications and alternative mRNA splicing events in the nucleus of non-neuronal cells. However, whether miRNAs employ this mechanism to regulate gene expression in the neuronal nucleus was not known. A prerequisite for the study of miRNA function in the nucleus of post-mitotic neurons is the a priori knowledge of the nuclear miRNA repertoire. Therefore, using microarray and deep sequencing technologies, I identified miRNAs which are enriched in the nuclei of rat primary cortical neurons (Khudayberdiev et al. 2013; Frontiers in Mol. Neurosci, accepted for publication). Subsequently, I validated differential expression of specific nuclear-enriched miRNAs by Northern blot, quantitative real-time PCR and fluorescence in situ hybridization. By cross-comparison to published reports, I found that nuclear accumulation of miRNAs might be linked to a down-regulation of their expression during in vitro development of cortical neurons. Importantly, I found a significant overrepresentation of guanine nucleotides at the 3’ terminus of nuclear-enriched miRNA isoforms (isomiRs), suggesting the presence of neuron-specific mechanisms involved in miRNA nuclear localization. In conclusion, these results provide a starting point for future studies addressing the nuclear function of specific miRNAs and the detailed mechanisms underlying subcellular localization of miRNAs in neurons. Taken together, the results presented in my cumulative PhD thesis demonstrate that activity-dependent regulation of specific miRNAs in different subcellular neuronal compartments (dendrites, nucleus, and soma) plays an important role in neuronal morphogenesis (dendrite and spine development) and plasticity.

Bibliographie / References

  1. Benjamini, Y., Drai, D., Elmer, G., Kafkafi, N., and Golani, I. (2001). Controlling the false discovery rate in behavior genetics research. Behav Brain Res 125, 279-284. Nuclear miRNAs in neurons (References)
  2. Khudayberdiev S.A, Zampa F, Rajman M, Schratt G (2013) A comprehensive characterization of the nuclear microRNA repertoire of post- mitotic neurons. Front. Mol. Neurosci. 6:43. doi: 10.3389/fnmol.2013.00043 (accepted for publication).
  3. MicroRNA function in the nervous system. Prog Mol Biol Transl Sci 102, 47-100.
  4. Benhamed, M., Herbig, U., Ye, T., Dejean, A., and Bischof, O. (2012). Senescence is an endogenous trigger for microRNA-directed transcriptional gene silencing in human cells. Nature Cell Biology 14, 266-+.
  5. Ohrt, T., Merkle, D., Birkenfeld, K., Echeverri, C.J., and Schwille, P. (2006). In situ fluorescence analysis demonstrates active siRNA exclusion from the nucleus by Exportin 5. Nucleic Acids Res 34, 1369-1380.
  6. Kim, Y.K., Yu, J., Han, T.S., Park, S.Y., Namkoong, B., Kim, D.H., Hur, K., Yoo, M.W., Lee, H.J., Yang, H.K., and Kim, V.N. (2009). Functional links between clustered microRNAs: suppression of cell-cycle inhibitors by microRNA clusters in gastric cancer. Nucleic Acids Res 37, 1672-1681.
  7. Pantano, L., Estivill, X., and Marti, E. (2010). SeqBuster, a bioinformatic tool for the processing and analysis of small RNAs datasets, reveals ubiquitous miRNA modifications in human embryonic cells. Nucleic Acids Res 38, e34.
  8. Tan, G.S., Garchow, B.G., Liu, X., Yeung, J., Morris, J.P.T., Cuellar, T.L., Mcmanus, M.T., and Kiriakidou, M. (2009). Expanded RNA-binding activities of mammalian Argonaute 2. Nucleic Acids Res 37, 7533-7545.
  9. Nishi, K., Nishi, A., Nagasawa, T., and Ui-Tei, K. (2013). Human TNRC6A is an Argonaute-navigator protein for microRNA-mediated gene silencing in the nucleus. RNA 19, 17-35.
  10. Politz, J.C., Hogan, E.M., and Pederson, T. (2009). MicroRNAs with a nucleolar location. RNA 15, 1705-1715.
  11. Jeffries, C.D., Fried, H.M., and Perkins, D.O. (2011). Nuclear and cytoplasmic localization of neural stem cell microRNAs. RNA 17, 675-686.
  12. Lee, L.W., Zhang, S., Etheridge, A., Ma, L., Martin, D., Galas, D., and Wang, K. (2010). Complexity of the microRNA repertoire revealed by next-generation sequencing. RNA 16, 2170-2180.
  13. Rodriguez-Aznar, E., Barrallo-Gimeno, A., and Nieto, M.A. (2013). Scratch2 prevents cell cycle re-entry by repressing miR-25 in postmitotic primary neurons. J Neurosci 33, 5095- 5105.
  14. Statistical significance was determined using Welch's t-test (unequal variance) with Bonferroni correction (*, p<0.05; **, p<0.001; ***, p<0.00001; ****, p<1.0-E10). (D) rNEnS for canonical isomiRs containing different 3' terminal nucleotides. Bar plots show mean ± SD (canon_G, n=57; canon_C, n=62; canon_A, n=75; canon_U, n=112). Statistical significance was determined using Welch's t-test (unequal variance) with Bonferroni correction (*, p<0.05; **, p<0.001; ***, p<1.0-E7). (E, F) Impact of trimming on the nuclear localization of isomiRs. (E) rNEnS for canonical isomiR_G that underwent 3' trimming, thereby exposing the indicated nucleotide. Bar plots show mean ± SD (canon_G, n=57; trim_G>G, n=12; trim_G>C, n=14; trim_G>A, n=17; trim_G>U, n=9).
  15. Statistical significance was determined using Welch's t-test (unequal variance) with Bonferroni correction (*, p<0.05; **, p<0.001; ***, p<1.0-E5). (F) rNEnS for canonical isomiR_C that underwent 3' trimming, thereby exposing the indicated nucleotide. Bar plots show mean ± SD (canon_U, n=112; trim_U>G, n=32; trim_U>C, n=29; trim_U>A, n=10; trim_U>U, n=20). Statistical significance was determined using Welch's t-test (unequal variance) with Bonferroni correction (*, p<0.05; **, p<0.001; ***, p<1.0-E5). APPENDIX: Curriculum Vitae 6. APPENDIX 148
  16. Control of alternative splicing through siRNA-mediated transcriptional gene silencing. Nat Struct Mol Biol 16, 717-724.
  17. Nuclear miRNAs in neurons (References)
  18. Morris, K.V., Chan, S.W., Jacobsen, S.E., and Looney, D.J. (2004). Small interfering RNA-induced transcriptional gene silencing in human cells. Science 305, 1289-1292.
  19. *with equal contribution
  20. Krol, J., Loedige, I., and Filipowicz, W. (2010). The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet 11, 597-610.
  21. Sinkkonen, L., Hugenschmidt, T., Filipowicz, W., and Svoboda, P. (2010). Dicer is associated with ribosomal DNA chromatin in mammalian cells. PLoS One 5, e12175.
  22. Schmid, M., and Jensen, T.H. (2008). The exosome: a multipurpose RNA-decay machine. Trends Biochem Sci 33, 501-510.
  23. Role of the miR-106b-25 microRNA cluster in hepatocellular carcinoma. Cancer Sci 100, 1234-1242.
  24. Jovicic, A., Roshan, R., Moisoi, N., Pradervand, S., Moser, R., Pillai, B., and Luthi-Carter, R. (2013). Comprehensive expression analyses of neural cell-type-specific miRNAs identify new determinants of the specification and maintenance of neuronal phenotypes. J Neurosci 33, 5127-5137.
  25. Llorens, F., Hummel, M., Pantano, L., Pastor, X., Vivancos, A., Castillo, E., Mattlin, H., Ferrer, A., Ingham, M., Noguera, M., Kofler, R., Dohm, J.C., Pluvinet, R., Bayes, M., Himmelbauer, H., Del Rio, J.A., Marti, E., and Sumoy, L. (2013). Microarray and deep sequencing cross-platform analysis of the mirRNome and isomiR variation in response to epidermal growth factor. BMC Genomics 14, 371.
  26. Laing, E., and Smith, C.P. (2010). RankProdIt: A web-interactive Rank Products analysis tool. BMC Res Notes 3, 221.
  27. Till, S., Lejeune, E., Thermann, R., Bortfeld, M., Hothorn, M., Enderle, D., Heinrich, C., Hentze, M.W., and Ladurner, A.G. (2007). A conserved motif in Argonaute-interacting proteins mediates functional interactions through the Argonaute PIWI domain. Nat Struct Mol Biol 14, 897-903. Nuclear miRNAs in neurons (Figures) depicted. (B) Frequency of different nucleotides in the 3' terminal 5 nts of 100 isomiRs with highest (upper panel), lowest (middle panel) and random rNEnS (lower panel). (C) Impact of 3' non-templated (NTA) and templated additions (TA) on the relative nuclear localization. Bar plots show mean ± SD (n= from 31 (TA_G) to 306 (canonical)).
  28. Ameyar-Zazoua, M., Rachez, C., Souidi, M., Robin, P., Fritsch, L., Young, R., Morozova, N., Fenouil, R., Descostes, N., Andrau, J.C., Mathieu, J., Hamiche, A., Ait-Si-Ali, S., Muchardt, C., Batsche, E., and Harel-Bellan, A. (2012). Argonaute proteins couple chromatin silencing to alternative splicing. Nat Struct Mol Biol 19, 998-1004.
  29. Place, R.F., Li, L.C., Pookot, D., Noonan, E.J., and Dahiya, R. (2008). MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci U S A 105, 1608-1613.
  30. Kim, D.H., Saetrom, P., Snove, O., Jr., and Rossi, J.J. (2008). MicroRNA-directed transcriptional gene silencing in mammalian cells. Proc Natl Acad Sci U S A 105, 16230- 16235.
  31. Addendum article 5. Khudayberdiev, S., Fiore, R., and Schratt, G. (2009). MicroRNA as modulators of neuronal responses. Commun Integr Biol 2, 411-413. Review article 6. Fiore, R*., Khudayberdiev, S.*, Saba, R.*, and Schratt, G. (2011).
  32. Liao, J.Y., Ma, L.M., Guo, Y.H., Zhang, Y.C., Zhou, H., Shao, P., Chen, Y.Q., and Qu, L.H. (2010). Deep sequencing of human nuclear and cytoplasmic small RNAs reveals an unexpectedly complex subcellular distribution of miRNAs and tRNA 3' trailers. PLoS One 5, e10563.
  33. Jeffries, C.D., Fried, H.M., and Perkins, D.O. (2010). Additional layers of gene regulatory complexity from recently discovered microRNA mechanisms. Int J Biochem Cell Biol 42, 1236-1242.
  34. Langmead, B. (2010). Aligning short sequencing reads with Bowtie. Curr Protoc Bioinformatics Chapter 11, Unit 11 17.
  35. Kumar, M., Lu, Z., Takwi, A.A., Chen, W., Callander, N.S., Ramos, K.S., Young, K.H., and Li, Y. (2011). Negative regulation of the tumor suppressor p53 gene by microRNAs. Oncogene 30, 843-853.
  36. Ansel, K.M., Pastor, W.A., Rath, N., Lapan, A.D., Glasmacher, E., Wolf, C., Smith, L.C., Papadopoulou, N., Lamperti, E.D., Tahiliani, M., Ellwart, J.W., Shi, Y., Kremmer, E., Rao, A., and Heissmeyer, V. (2008). Mouse Eri1 interacts with the ribosome and catalyzes 5.8S rRNA processing. Nat Struct Mol Biol 15, 523-530.
  37. Park, C.W., Zeng, Y., Zhang, X., Subramanian, S., and Steer, C.J. (2010). Mature microRNAs identified in highly purified nuclei from HCT116 colon cancer cells. RNA Biol 7, 606-614.
  38. Tang, R., Li, L., Zhu, D., Hou, D., Cao, T., Gu, H., Zhang, J., Chen, J., Zhang, C.Y., and Zen, K. (2012). Mouse miRNA-709 directly regulates miRNA-15a/16-1 biogenesis at the posttranscriptional level in the nucleus: evidence for a microRNA hierarchy system. Cell Res 22, 504-515.
  39. Siegel, G., Obernosterer, G., Fiore, R., Oehmen, M., Bicker, S., Christensen, M., Khudayberdiev, S., Leuschner, P.F., Busch, C.J., Kane, C., Hubel, K., Dekker, F., Hedberg, C., Rengarajan, B., Drepper, C., Waldmann, H., Kauppinen, S., Greenberg, M.E., Draguhn, A., Rehmsmeier, M., Martinez, J., and Schratt, G.M. (2009). A functional screen implicates microRNA-138-dependent regulation of the depalmitoylation enzyme APT1 in dendritic spine morphogenesis. Nature Cell Biology 11, 705-716.
  40. Bicker, S., Khudayberdiev, S., Weiss, K., Zocher, K., Baumeister, S., and Schratt, G. (2013). The DEAH-box helicase DHX36 mediates dendritic localization of the neuronal precursor-microRNA-134. Genes Dev 27, 991-996
  41. Ruegger, S., and Grosshans, H. (2012). MicroRNA turnover: when, how, and why. Trends Biochem Sci 37, 436-446.
  42. Norris, A.D., and Calarco, J.A. (2012). Emerging Roles of Alternative Pre-mRNA Splicing Regulation in Neuronal Development and Function. Front Neurosci 6, 122.


* Das Dokument ist im Internet frei zugänglich - Hinweise zu den Nutzungsrechten