Hostname: page-component-8448b6f56d-xtgtn Total loading time: 0 Render date: 2024-04-23T14:01:21.043Z Has data issue: false hasContentIssue false

Investigation of a Fused in Sarcoma Splicing Mutation in a Chinese Amyotrophic Lateral Sclerosis Patient

Published online by Cambridge University Press:  12 December 2022

Wanli Yang
Affiliation:
The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, Shanghai, China
Xi Chen
Affiliation:
Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
Yu Zhou
Affiliation:
The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, Shanghai, China
Xuelin Tang
Affiliation:
The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, Shanghai, China
Yimin Sun
Affiliation:
Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
Yi Dong
Affiliation:
Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
Hong Yang
Affiliation:
The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, Shanghai, China
Yan Chen*
Affiliation:
Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
Ming Zhang*
Affiliation:
The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, Shanghai, China Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China Institute for Advanced Study, Tongji University, Shanghai, China
*
Corresponding authors: Yan Chen. E-mail: chenyanhs@fudan.edu.cn; Ming Zhang. E-mail: mingzhang@tongji.edu.cn
Corresponding authors: Yan Chen. E-mail: chenyanhs@fudan.edu.cn; Ming Zhang. E-mail: mingzhang@tongji.edu.cn

Abstract:

Objective:

Genetic mutations of fused in sarcoma (FUS) causing amyotrophic lateral sclerosis (ALS) may disrupt mRNA splicing events. For example, the FUS c.1394-2delA variant was reported in two western ALS patients, but its molecular mechanism is unclear. In this study, we aim to investigate FUS splice site mutations in Chinese ALS patients.

Methods:

Sanger sequencing was used to identify FUS splicing mutations in Chinese ALS patients. We combined a deep learning tool (SpliceAI), RNA sequencing, and RT-PCR/RT-qPCR to analyze the effect of FUS c.1394-2delA mutation on RNA splicing and expression. AlphaFold was used to predict the protein structure of mutant FUS. In transfected cell lines, we used immunofluorescence to assess cytoplasmic mislocalization of mutant FUS protein.

Results:

We identified a de novo FUS splice acceptor site mutation (c.1394-2delA, p. Gly466Valfs*14) in one Chinese sporadic ALS patient, which is linked to exon 14 skipping, and upregulated total FUS mRNA expression. The FUS splice site mutation was predicted to be translated into a truncated protein product at C-terminal. In vitro studies revealed that the FUS mutation increased cytoplasmic mislocalization in both HEK293T and SH-SY5Y cells.

Conclusions:

We identified a de novo FUS splicing mutation (c.1394-2delA, p. Gly466Valfs*14) in 1 out of 233 Chinese ALS patients. It caused abnormal RNA splicing, upregulated gene expression, truncated FUS translation, and cytosolic mislocalization. Our findings suggested that FUS splice site mutation is rare in Chinese ALS patients and extended our knowledge of molecular mechanisms of the FUS c.1394-2delA mutation.

Résumé :

RÉSUMÉ :

Étude d’une mutation affectant des sites d’épissage dans le cas de protéines de fusion d’un sarcome chez un patient chinois atteint de sclérose latérale amyotrophique.

Objectif :

Les mutations génétiques des protéines de fusion d’un sarcome (PFS), lesquelles sont à l’origine de la sclérose latérale amyotrophique (SLA), peuvent perturber les sites d’épissage de l’ARN messager. À titre d’exemple, la variante c.1394-2delA des PFS a été signalée chez deux patients occidentaux atteints de SLA mais son mécanisme moléculaire demeure nébuleux. Dans la présente étude, nous entendons étudier les mutations des sites d’épissage des PFS chez des patients chinois atteints de SLA.

Méthodes :

Le séquençage génétique de Sanger a été utilisé pour identifier les mutations des sites d’épissage des PFS chez des patients chinois atteints de SLA. Nous avons aussi combiné un outil d’apprentissage profond (SpliceAI), le séquençage de l’ARN messager et la méthode dite « RT-PCR/RT-qPCR » pour analyser l’effet de la mutation des PFS (variante c.1394-2delA) sur l’épissage et l’expression de l’ARN messager. Le programme AlphaFold a été par ailleurs utilisé pour prédire la structure protéique des PFS mutantes. Dans les lignées cellulaires transfectées, nous avons utilisé l’immunofluorescence pour évaluer la mauvaise localisation cytoplasmique des PFS mutantes.

Résultats :

Nous avons identifié une mutation de novo du site accepteur d’épissage des PFS (c.1394-2delA, p.Gly466Valfs*14) chez un patient chinois atteint de SLA sporadique, laquelle est liée à un saut de l’exon 14, ce qui a augmenté l’expression totale de l’ARN messager des PFS. La mutation des sites d’épissage des PFS est supposée se traduire par un produit protéique tronqué à l'extrémité C-terminale. Des études in vitro ont aussi révélé que les mutations des PFS augmentait la mauvaise localisation cytoplasmique dans les cellules HEK293T et SH-SY5Y.

Conclusions :

Nous avons identifié une mutation de novo de l’épissage des PFS (c.1394-2delA, p.Gly466Valfs*14) chez un des 233 patients chinois atteints de SLA. Cette mutation a provoqué un épissage anormal de l’ARN messager, une expression génétique régulée, une traduction tronquée des PFS ainsi qu’une mauvaise localisation cytoplasmique. En somme, nos résultats suggèrent que les mutations des sites d’épissage des PFS sont rares chez les patients chinois atteints de SLA. Ils permettent également d’élargir nos connaissances quant aux mécanismes moléculaires de la mutation c.1394-2delA des PFS.

Type
Original Article
Copyright
© The Author(s), 2022. Published by Cambridge University Press on behalf of Canadian Neurological Sciences Federation

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Brown, RH, Al-Chalabi, A. Amyotrophic lateral sclerosis. N Engl J Med. 2017;377:162–72. DOI 10.1056/NEJMra1603471.CrossRefGoogle ScholarPubMed
Zou, ZY, Zhou, ZR, Che, CH, Liu, CY, He, RL, Huang, HP. Genetic epidemiology of amyotrophic lateral sclerosis: a systematic review and meta-analysis. J Neurol Neurosurg Psychiatry. 2017;88:540–9. DOI 10.1136/jnnp-2016-315018.CrossRefGoogle ScholarPubMed
Chen, C, Ding, X, Akram, N, Xue, S, Luo, SZ. Fused in sarcoma: properties, self-assembly and correlation with neurodegenerative diseases. Molecules. 2019;24:1622. DOI 10.3390/molecules24081622.CrossRefGoogle ScholarPubMed
Qiu, H, Lee, S, Shang, Y, et al. ALS-associated mutation FUS-R521C causes DNA damage and RNA splicing defects. J Clin Invest. 2021;131:e149564. DOI 10.1172/jci149564.CrossRefGoogle ScholarPubMed
Shang, Y, Huang, EJ. Mechanisms of FUS mutations in familial amyotrophic lateral sclerosis. Brain Res. 2016;1647:6578. DOI 10.1016/j.brainres.2016.03.036.CrossRefGoogle ScholarPubMed
Reber, S, Stettler, J, Filosa, G, et al. Minor intron splicing is regulated by FUS and affected by ALS-associated FUS mutants. EMBO J. 2016;35:1504–21. DOI 10.15252/embj.201593791.CrossRefGoogle ScholarPubMed
Dormann, D, Rodde, R, Edbauer, D, et al. ALS-associated fused in sarcoma (FUS) mutations disrupt Transportin-mediated nuclear import. EMBO J. 2010;29:2841–57. DOI 10.1038/emboj.2010.143.CrossRefGoogle ScholarPubMed
Baralle, D, Baralle, M. Splicing in action: assessing disease causing sequence changes. J Med Genet. 2005;42:737–48. DOI 10.1136/jmg.2004.029538.CrossRefGoogle ScholarPubMed
Skjørringe, T, Tümer, Z, Møller, LB. Splice site mutations in the ATP7A gene. PLoS One. 2011;6:e18599. DOI 10.1371/journal.pone.0018599.CrossRefGoogle ScholarPubMed
Krawczak, M, Reiss, J, Cooper, DN. The mutational spectrum of single base-pair substitutions in mRNA splice junctions of human genes: causes and consequences. Hum Genet. 1992;90:4154. DOI 10.1007/bf00210743.CrossRefGoogle ScholarPubMed
Cooper, TA, Wan, L, Dreyfuss, G. RNA and disease. Cell. 2009;136:777–93. DOI 10.1016/j.cell.2009.02.011.CrossRefGoogle ScholarPubMed
DeJesus-Hernandez, M, Kocerha, J, Finch, N, et al. De novo truncating FUS gene mutation as a cause of sporadic amyotrophic lateral sclerosis. Hum Mutat. 2010;31:E137789. DOI 10.1002/humu.21241.CrossRefGoogle ScholarPubMed
Belzil, VV, St-Onge, J, Daoud, H, et al. Identification of a FUS splicing mutation in a large family with amyotrophic lateral sclerosis. J Hum Genet. 2011;56:247–9. DOI 10.1038/jhg.2010.162.CrossRefGoogle Scholar
Canosa, A, Lomartire, A, De Marco, G, et al. A novel splice site FUS mutation in a familial ALS case: effects on protein expression. Amyotroph Lateral Scler Frontotemporal Degener. 2022;23:128–36. DOI 10.1080/21678421.2021.1909065.CrossRefGoogle Scholar
Müller, K, Brenner, D, Weydt, P, et al. Comprehensive analysis of the mutation spectrum in 301 German ALS families. J Neurol Neurosurg Psychiatry. 2018;89:817–27. DOI 10.1136/jnnp-2017-317611.CrossRefGoogle ScholarPubMed
Cady, J, Allred, P, Bali, T, et al. Amyotrophic lateral sclerosis onset is influenced by the burden of rare variants in known amyotrophic lateral sclerosis genes. Ann Neurol. 2015;77:100–13. DOI 10.1002/ana.24306.CrossRefGoogle ScholarPubMed
Ludolph, A, Drory, V, Hardiman, O, et al. A revision of the El Escorial criteria – 2015. Amyotroph Lateral Scler Frontotemporal Degener. 2015;16:291–2. DOI 10.3109/21678421.2015.1049183.CrossRefGoogle Scholar
Cedarbaum, JM, Stambler, N, Malta, E, et al. The ALSFRS-R: a revised ALS functional rating scale that incorporates assessments of respiratory function. BDNF ALS Study Group (Phase III). J Neurol Sci. 1999;169:1321. DOI 10.1016/s0022-510x(99)00210-5.CrossRefGoogle ScholarPubMed
Jaganathan, K, Kyriazopoulou Panagiotopoulou, S, McRae, JF, et al. Predicting splicing from primary sequence with deep learning. Cell. 2019;176:535.e24–48. DOI 10.1016/j.cell.2018.12.015.CrossRefGoogle ScholarPubMed
Jumper, J, Evans, R, Pritzel, A, et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596:583–9. DOI 10.1038/s41586-021-03819-2.CrossRefGoogle ScholarPubMed
Li, YI, Knowles, DA, Humphrey, J, et al. Annotation-free quantification of RNA splicing using LeafCutter. Nat Genet. 2018;50:151–8. DOI 10.1038/s41588-017-0004-9.CrossRefGoogle ScholarPubMed
Dobin, A, Davis, CA, Schlesinger, F, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:1521. DOI 10.1093/bioinformatics/bts635.CrossRefGoogle ScholarPubMed
Dalby, B, Cates, S, Harris, A, et al. Advanced transfection with Lipofectamine 2000 reagent: primary neurons, siRNA, and high-throughput applications. Methods. 2004;33:95103. DOI 10.1016/j.ymeth.2003.11.023.CrossRefGoogle ScholarPubMed
An, H, Rabesahala de Meritens, C, Buchman, VL, Shelkovnikova, TA. Frameshift peptides alter the properties of truncated FUS proteins in ALS-FUS. Mol Brain. 2020;13:77. DOI 10.1186/s13041-020-00618-0.CrossRefGoogle ScholarPubMed
Niu, C, Zhang, J, Gao, F, et al. FUS-NLS/Transportin 1 complex structure provides insights into the nuclear targeting mechanism of FUS and the implications in ALS. PLoS One. 2012;7:e47056. DOI 10.1371/journal.pone.0047056.CrossRefGoogle ScholarPubMed
Zinszner, H, Sok, J, Immanuel, D, Yin, Y, Ron, D. TLS (FUS) binds RNA in vivo and engages in nucleo-cytoplasmic shuttling. J Cell Sci. 1997;110:1741–50. DOI 10.1242/jcs.110.15.1741.CrossRefGoogle ScholarPubMed
Lee, BJ, Cansizoglu, AE, Süel, KE, Louis, TH, Zhang, Z, Chook, YM. Rules for nuclear localization sequence recognition by karyopherin beta 2. Cell. 2006;126:543–58. DOI 10.1016/j.cell.2006.05.049.CrossRefGoogle ScholarPubMed
Daigle, JG, Lanson, NA Jr, Smith, RB, et al. RNA-binding ability of FUS regulates neurodegeneration, cytoplasmic mislocalization and incorporation into stress granules associated with FUS carrying ALS-linked mutations. Hum Mol Genet. 2013;22:1193–205. DOI 10.1093/hmg/dds526.CrossRefGoogle ScholarPubMed
Sainouchi, M, Hatano, Y, Tada, M, et al. A novel splicing variant of ANXA11 in a patient with amyotrophic lateral sclerosis: histologic and biochemical features. Acta Neuropathol Commun. 2021;9:106. DOI 10.1186/s40478-021-01202-w.CrossRefGoogle Scholar
Birve, A, Neuwirth, C, Weber, M, et al. A novel SOD1 splice site mutation associated with familial ALS revealed by SOD activity analysis. Hum Mol Genet. 2010;19:4201–6. DOI 10.1093/hmg/ddq338.CrossRefGoogle ScholarPubMed
Lu, YQ, Chen, JM, Lin, H, et al. Novel intronic mutations of TBK1 promote aberrant splicing modes in amyotrophic lateral sclerosis. Front Mol Neurosci. 2022;15:691534. DOI 10.3389/fnmol.2022.691534.CrossRefGoogle ScholarPubMed
Amrani, N, Sachs, MS, Jacobson, A. Early nonsense: mRNA decay solves a translational problem. Nat Rev Mol Cell Biol. 2006;7:415–25. DOI 10.1038/nrm1942.CrossRefGoogle Scholar
Zhou, Y, Liu, S, Liu, G, Oztürk, A, Hicks, GG. ALS-associated FUS mutations result in compromised FUS alternative splicing and autoregulation. PLos Genet. 2013;9:e1003895. DOI 10.1371/journal.pgen.1003895.CrossRefGoogle ScholarPubMed
Humphrey, J, Birsa, N, Milioto, C, et al. FUS ALS-causative mutations impair FUS autoregulation and splicing factor networks through intron retention. Nucleic Acids Res. 2020;48:6889–905. DOI 10.1093/nar/gkaa410.CrossRefGoogle ScholarPubMed
Supplementary material: File

Yang et al. supplementary material

Yang et al. supplementary material

Download Yang et al. supplementary material(File)
File 480.8 KB