5.9
CiteScore
5.9
Impact Factor
Volume 49 Issue 9
Sep.  2022
Turn off MathJax
Article Contents

Loss of Wtap results in cerebellar ataxia and degeneration of Purkinje cells

doi: 10.1016/j.jgg.2022.03.001
Funds:

This study was supported by the National Natural Science Foundation of China (82121003, 81970841, and 81790643), the Department of Science and Technology of Sichuan Province (2021YFS0386, 2021YFS0369, 20ZYD038, 20ZYD037, 2020JDZH0026, 2021JDZH0022), the CAMS Innovation Fund for Medical Sciences (2019–12M-5-032), Huanhua Distingished Scholar grant, and the Department of Chengdu Science and Technology (2021-YF05-01316-SN). The funders had no role in the study design, data collection and analysis, or preparation of the manuscript. The authors would like to thank Chengdu LiLai Biotechnology Co., Ltd., for technical assistance with histology analysis.

  • Received Date: 2021-12-06
  • Accepted Date: 2022-03-04
  • Rev Recd Date: 2022-03-03
  • Publish Date: 2022-03-15
  • N6-methyladenosine (m6A) modification, which is achieved by the METTL3/METTL14/WTAP methyltransferase complex, is the most abundant internal mRNA modification. Although recent evidence indicates that m6A can regulate neurodevelopment as well as synaptic function, the roles of m6A modification in the cerebellum and related synaptic connections are not well established. Here, we report that Purkinje cell (PC)-specific WTAP knockout mice display early-onset ataxia concomitant with cerebellar atrophy due to extensive PC degeneration and apoptotic cell death. Loss of Wtap also causes the aberrant degradation of multiple PC synapses. WTAP depletion leads to decreased expression levels of METTL3/14 and reduced m6A methylation in PCs. Moreover, the expression of GFAP and NF-L in the degenerating cerebellum is increased, suggesting severe neuronal injuries. In conclusion, this study demonstrates the critical role of WTAP-mediated m6A modification in cerebellar PCs, thus providing unique insights related to neurodegenerative disorders.
  • loading
  • Ashizawa, T., Oz, G. and Paulson, H. L., 2018. Spinocerebellar ataxias:prospects and challenges for therapy development. Nat. Rev. Neurol. 14, 590-605.
    Barski, J. J., Dethleffsen, K. and Meyer, M., 2000. Cre recombinase expression in cerebellar Purkinje cells. Genesis. 28, 93-98.
    Buffo, A., Rolando, C. and Ceruti, S., 2010. Astrocytes in the damaged brain:molecular and cellular insights into their reactive response and healing potential. Biochem. Pharmacol. 79, 77-89.
    Cao, Y. H., Zhuang, Y. L., Chen, J. C., Xu, W. Z., Shou, Y. K., Huang, X. L., Shu, Q. and Li, X. K., 2020. Dynamic effects of Fto in regulating the proliferation and differentiation of adult neural stem cells of mice. Hum. Mol. Genet. 29, 727-735.
    Cerminara, N. L., Lang, E. J., Sillitoe, R. V. and Apps, R., 2015. Redefining the cerebellar cortex as an assembly of non-uniform Purkinje cell microcircuits. Nat. Rev. Neurosci. 16, 79-93.
    Chang, M. Q., Lv, H. Y., Zhang, W. L., Ma, C. H., He, X., Zhao, S. L., Zhang, Z. W., Zeng, Y. X., Song, S. H. and Niu, Y. M., 2017. Region-specific RNA m6A methylation represents a new layer of control in the gene regulatory network in the mouse brain. Open Biol. 7, 170166.
    Chen, J. C., Zhang, Y. C., Huang, C. M., Shen, H., Sun, B. F., Cheng, X. J., Zhang, Y. J., Yang, Y. G., Shu, Q. and Yang, Y., 2019. m6A regulates neurogenesis and neuronal development by modulating histone methyltransferase Ezh2. Genom. Proteomics Bioinformatics. 17, 154-168.
    Hsu, P. J., Zhu, Y. F., Ma, H. H., Guo, Y. S., Shi, X. D., Liu, Y. Y., Qi, M. J., Lu, Z. K., Shi, H. L. and Wang, J. Y., 2017. Ythdc2 is an N 6-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res. 27, 1115-1127.
    Jayadev, S. and Bird, T. D., 2013. Hereditary ataxias:overview. Genet. Med. 15, 673-683.
    Li, L. P., Zang, L. Q., Zhang, F. R., Chen, J. C., Shen, H., Shu, L., Liang, F., Feng, C. Y., Chen, D. and Tao, H. K., 2017. Fat mass and obesity-associated (FTO) protein regulates adult neurogenesis. Hum. Mol. Genet. 26, 2398-2411.
    Li, M. M., Zhao, X., Wang, W., Shi, H. L., Pan, Q. F., Lu, Z. K., Perez, S. P., Suganthan, R., He, C. and Bjoeras, M., 2018. Ythdf2-mediated m 6 A mRNA clearance modulates neural development in mice. Genome Biol. 19, 1-16.
    Liu, C. Y., Mei, M., Li, Q. L., Roboti, P., Pang, Q. Q., Ying, Z. Z., Gao, F., Lowe, M. and Bao, S. L., 2017. Loss of the golgin GM130 causes Golgi disruption, Purkinje neuron loss, and ataxia in mice. Proc. Natl. Acad. Sci. Unit. States Am. 114, 346-351.
    Ma, C. H., Chang, M. Q., Lv, H. Y., Zhang, Z. W., Zhang, W. L., He, X., Wu, G. L., Zhao, S. L., Zhang, Y. and Wang, D., 2018. RNA m 6 A methylation participates in regulation of postnatal development of the mouse cerebellum. Genome Biol. 19, 1-18.
    Merkurjev, D., Hong, W. T., Iida, K., Oomoto, I., Goldie, B. J., Yamaguti, H., Ohara, T., Kawaguchi, S. Y., Hirano, T. and Martin, K. C., 2018. Synaptic N 6-methyladenosine (m 6 A) epitranscriptome reveals functional partitioning of localized transcripts. Nat. Neurosci. 21, 1004-1014.
    Meyer, K. D., Saletore, Y., Zumbo, P., Elemento, O., Mason, C. E. and Jaffrey, S. R., 2012. Comprehensive analysis of mRNA methylation reveals enrichment in 3'UTRs and near stop codons. Cell. 149, 1635-1646.
    Ping, X. L., Sun, B. F., Wang, L., Xiao, W., Yang, X., Wang, W. J., Adhikari, S., Shi, Y., Lv, Y. and Chen, Y. S., 2014. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 24, 177-189.
    Schols, L., Bauer, P., Schmidt, T., Schulte, T. and Riess, O., 2004. Autosomal dominant cerebellar ataxias:clinical features, genetics, and pathogenesis. Lancet Neurol. 3, 291-304.
    Shakkottai, V. and Paulson, H., 2019. Expanding the genetic basis of ataxia. Nat. Genet. 51, 580-581.
    Shi, H. L., Zhang, X. L., Weng, Y. L., Lu, Z. Y., Liu, Y. J., Lu, Z. K., Li, J. N., Hao, P. L., Zhang, Y. and Zhang, F., 2018. m 6 A facilitates hippocampus-dependent learning and memory through YTHDF1. Nature. 563, 249-253.
    Walters, B. J., Mercaldo, V., Gillon, C. J., Yip, M., Neve, R. L., Boyce, F. M., Frankland, P. W. and Josselyn, S. A., 2017. The role of the RNA demethylase FTO (fat mass and obesity-associated) and mRNA methylation in hippocampal memory formation. Neuropsychopharmacology. 42, 1502-1510.
    Wang, C., Zou, P., Yang, C., Liu, L., Cheng, L., He, X., Zhang, L., Zhang, Y., Jiang, H. and Chen, P. R., 2019. Dynamic modifications of biomacromolecules:mechanism and chemical interventions. Sci. China Life Sci. 62, 1459-1471.
    Wang, C. X., Cui, G. S., Liu, X. Y., Xu, K., Wang, M., Zhang, X. X., Jiang, L. Y., Li, A., Yang, Y. and Lai, W. Y., 2018. METTL3-mediated m6A modification is required for cerebellar development. PLoS Biol. 16, e2004880.
    Wang, P., Doxtader, K. A. and Nam, Y. S., 2016a. Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol. Cell. 63, 306-317.
    Wang, S. S. H., Kloth, A. D. and Badura, A., 2014a. The cerebellum, sensitive periods, and autism. Neuron. 83, 518-532.
    Wang, X., Feng, J., Xue, Y., Guan, Z. Y., Zhang, D. L., Liu, Z., Gong, Z., Wang, Q., Huang, J. B. and Tang, C., 2016b. Structural basis of N 6-adenosine methylation by the METTL3-METTL14 complex. Nature. 534, 575-578.
    Wang, X., Lu, Z. K., Gomez, A., Hon, G. C., Yue, Y. N., Han, D. L., Fu, Y., Parisien, M., Dai, Q. and Jia, G. F., 2014b. N 6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 505, 117-120.
    Weng, Y. L., Wang, X., An, R., Cassin, J., Vissers, C., Liu, Y. Y., Liu, Y. J., Xu, T. L., Wang, X. Y. and Wong, S. Z. H., 2018. Epitranscriptomic m6A regulation of axon regeneration in the adult mammalian nervous system. Neuron. 97, 313-325. e316.
    Widagdo, J., Zhao, Q. Y., Kempen, M.-J., Tan, M. C., Ratnu, V. S., Wei, W., Leighton, L., Spadaro, P. A., Edson, J. and Anggono, V., 2016. Experience-dependent accumulation of N6-methyladenosine in the prefrontal cortex is associated with memory processes in mice. J. Neurosci. 36, 6771-6777.
    Yang, Y., Yamada, T. and Bonni, A., 2019. Epigenetic Regulation of the Cerebellum. Cham:Springer International Publishing.
    Yang, Y. M., Sun, K. X., Liu, W. J., Zhang, L., Peng, K., Zhang, S. S., Li, S. J., Yang, M., Jiang, Z. L. and Lu, F., 2018. Disruption of Tmem30a results in cerebellar ataxia and degeneration of Purkinje cells. Cell Death Dis. 9, 1-13.
    Yoon, K. J., Ringeling, F. R., Vissers, C., Jacob, F., Pokrass, M., Jimenez-Cyrus, D., Su, Y. J., Kim, N. S., Zhu, Y. H. and Zheng, L., 2017. Temporal control of mammalian cortical neurogenesis by m6A methylation. Cell. 171, 877-889. e817.
    Yu, J., Chen, M. X., Huang, H. J., Zhu, J. D., Song, H. X., Zhu, J., Park, J. and Ji, S. J., 2018. Dynamic m6A modification regulates local translation of mRNA in axons. Nucleic Acids Res. 46, 1412-1423.
    Yuan, A. D., Rao, M. V. and Nixon, R. A., 2017. Neurofilaments and neurofilament proteins in health and disease. Cold Spring Harbor Perspect. Biol. 9, a018309.
    Zhang, Z. Y., Wang, M., Xie, D. F., Huang, Z. H., Zhang, L. S., Yang, Y., Ma, D. X., Li, W. G., Zhou, Q. and Yang, Y. G., 2018. METTL3-mediated N 6-methyladenosine mRNA modification enhances long-term memory consolidation. Cell Res. 28, 1050-1061.
    Zhao, B. S., Roundtree, I. A. and He, C., 2017. Post-transcriptional gene regulation by mRNA modifications. Nat. Rev. Mol. Cell Biol. 18, 31-42.
    Zhuang, M. R., Li, X. B., Zhu, J. D., Zhang, J., Niu, F. G., Liang, F. H., Chen, M. X., Li, D., Han, P. and Ji, S. J., 2019. The m6A reader YTHDF1 regulates axon guidance through translational control of Robo3. 1 expression. Nucleic Acids Res. 47, 4765-4777
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Article Metrics

    Article views (227) PDF downloads (26) Cited by ()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return