Somatic Mutations in the MTOR gene cause focal cortical dysplasia type IIb
Mitsuko Nakashima MD, PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
These authors contributed equally to this work.
Search for more papers by this authorHirotomo Saitsu MD, PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
These authors contributed equally to this work.
Search for more papers by this authorNobuyuki Takei PhD
Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
Search for more papers by this authorJun Tohyama MD, PhD
Department of Child Neurology, Nishi-Niigata Chuo National Hospital, Niigata, Japan
Search for more papers by this authorMitsuhiro Kato MD, PhD
Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
Search for more papers by this authorHiroki Kitaura DDS, PhD
Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
Search for more papers by this authorMasaaki Shiina MD, PhD
Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama, Japan
Search for more papers by this authorHiroshi Shirozu MD, PhD
Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
Search for more papers by this authorHiroshi Masuda MD
Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
Search for more papers by this authorKeisuke Watanabe PhD
Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
Search for more papers by this authorChihiro Ohba MD, PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
Search for more papers by this authorYoshinori Tsurusaki PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
Search for more papers by this authorNoriko Miyake MD, PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
Search for more papers by this authorYingjun Zheng MD, PhD
Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
Search for more papers by this authorTatsuhiro Sato PhD
Division of Biochemistry, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
Search for more papers by this authorHirohide Takebayashi MD, PhD
Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
Search for more papers by this authorKazuhiro Ogata MD, PhD
Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama, Japan
Search for more papers by this authorShigeki Kameyama MD, PhD
Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
Search for more papers by this authorAkiyoshi Kakita MD, PhD
Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
These are co-last authors.
Search for more papers by this authorCorresponding Author
Naomichi Matsumoto MD, PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
These are co-last authors.
Address correspondence to Dr Naomichi Matsumoto, Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3–9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan. E-mail: [email protected]Search for more papers by this authorMitsuko Nakashima MD, PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
These authors contributed equally to this work.
Search for more papers by this authorHirotomo Saitsu MD, PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
These authors contributed equally to this work.
Search for more papers by this authorNobuyuki Takei PhD
Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
Search for more papers by this authorJun Tohyama MD, PhD
Department of Child Neurology, Nishi-Niigata Chuo National Hospital, Niigata, Japan
Search for more papers by this authorMitsuhiro Kato MD, PhD
Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
Search for more papers by this authorHiroki Kitaura DDS, PhD
Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
Search for more papers by this authorMasaaki Shiina MD, PhD
Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama, Japan
Search for more papers by this authorHiroshi Shirozu MD, PhD
Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
Search for more papers by this authorHiroshi Masuda MD
Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
Search for more papers by this authorKeisuke Watanabe PhD
Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
Search for more papers by this authorChihiro Ohba MD, PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
Search for more papers by this authorYoshinori Tsurusaki PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
Search for more papers by this authorNoriko Miyake MD, PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
Search for more papers by this authorYingjun Zheng MD, PhD
Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
Search for more papers by this authorTatsuhiro Sato PhD
Division of Biochemistry, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
Search for more papers by this authorHirohide Takebayashi MD, PhD
Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
Search for more papers by this authorKazuhiro Ogata MD, PhD
Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama, Japan
Search for more papers by this authorShigeki Kameyama MD, PhD
Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
Search for more papers by this authorAkiyoshi Kakita MD, PhD
Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
These are co-last authors.
Search for more papers by this authorCorresponding Author
Naomichi Matsumoto MD, PhD
Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
These are co-last authors.
Address correspondence to Dr Naomichi Matsumoto, Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3–9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan. E-mail: [email protected]Search for more papers by this authorAbstract
Objective
Focal cortical dysplasia (FCD) type IIb is a cortical malformation characterized by cortical architectural abnormalities, dysmorphic neurons, and balloon cells. It has been suggested that FCDs are caused by somatic mutations in cells in the developing brain. Here, we explore the possible involvement of somatic mutations in FCD type IIb.
Methods
We collected a total of 24 blood-brain paired samples with FCD, including 13 individuals with FCD type IIb, 5 with type IIa, and 6 with type I. We performed whole-exome sequencing using paired samples from 9 of the FCD type IIb subjects. Somatic MTOR mutations were identified and further investigated using all 24 paired samples by deep sequencing of the entire gene's coding region. Somatic MTOR mutations were confirmed by droplet digital polymerase chain reaction. The effect of MTOR mutations on mammalian target of rapamycin (mTOR) kinase signaling was evaluated by immunohistochemistry and Western blotting analyses of brain samples and by in vitro transfection experiments.
Results
We identified four lesion-specific somatic MTOR mutations in 6 of 13 (46%) individuals with FCD type IIb showing mutant allele rates of 1.11% to 9.31%. Functional analyses showed that phosphorylation of ribosomal protein S6 in FCD type IIb brain tissues with MTOR mutations was clearly elevated, compared to control samples. Transfection of any of the four MTOR mutants into HEK293T cells led to elevated phosphorylation of 4EBP, the direct target of mTOR kinase.
Interpretation
We found low-prevalence somatic mutations in MTOR in FCD type IIb, indicating that activating somatic mutations in MTOR cause FCD type IIb. Ann Neurol 2015;78:375–386
Supporting Information
Additional Supporting Information can be found in the online version of this article.
Filename | Description |
---|---|
ana24444-sup-0001-suppinfo.docx32.6 KB |
Supplementary Information |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
References
- 1Taylor DC, Falconer MA, Bruton CJ, Corsellis JA. Focal dysplasia of the cerebral cortex in epilepsy. J Neurol Neurosurg Psychiatry 1971; 34: 369–387.
- 2Marin-Valencia I, Guerrini R, Gleeson JG. Pathogenetic mechanisms of focal cortical dysplasia. Epilepsia 2014; 55: 970–978.
- 3Blumcke I, Thom M, Aronica E, et al. The clinicopathologic spectrum of focal cortical dysplasias: a consensus classification proposed by an ad hoc Task Force of the ILAE Diagnostic Methods Commission. Epilepsia 2011; 52: 158–174.
- 4Sisodiya SM, Fauser S, Cross JH, Thom M. Focal cortical dysplasia type II: biological features and clinical perspectives. Lancet Neurol 2009; 8: 830–843.
- 5Baybis M, Yu J, Lee A, et al. mTOR cascade activation distinguishes tubers from focal cortical dysplasia. Ann Neurol 2004; 56: 478–487.
- 6Sato T, Nakashima A, Guo L, Coffman K, Tamanoi F. Single amino-acid changes that confer constitutive activation of mTOR are discovered in human cancer. Oncogene 2010; 29: 2746–2752.
- 7Ljungberg MC, Bhattacharjee MB, Lu Y, et al. Activation of mammalian target of rapamycin in cytomegalic neurons of human cortical dysplasia. Ann Neurol 2006; 60: 420–429.
- 8Takei N, Nawa H. mTOR signaling and its roles in normal and abnormal brain development. Front Mol Neurosci 2014; 7: 28.
- 9Crino PB. mTOR: a pathogenic signaling pathway in developmental brain malformations. Trends Mol Med 2011; 17: 734–742.
- 10Poduri A, Evrony GD, Cai X, Walsh CA. Somatic mutation, genomic variation, and neurological disease. Science 2013; 341: 1237758.
- 11Schick V, Majores M, Koch A, et al. Alterations of phosphatidylinositol 3-kinase pathway components in epilepsy-associated glioneuronal lesions. Epilepsia 2007; 48(Suppl 5): 65–73.
- 12Riviere JB, Mirzaa GM, O'Roak BJ, et al. De novo germline and postzygotic mutations in AKT3, PIK3R2 and PIK3CA cause a spectrum of related megalencephaly syndromes. Nat Genet 2012; 44: 934–940.
- 13Lee JH, Huynh M, Silhavy JL, et al. De novo somatic mutations in components of the PI3K-AKT3-mTOR pathway cause hemimegalencephaly. Nat Genet 2012; 44: 941–945.
- 14Lindhurst MJ, Sapp JC, Teer JK, et al. A mosaic activating mutation in AKT1 associated with the Proteus syndrome. N Engl J Med 2011; 365: 611–619.
- 15Poduri A, Evrony GD, Cai X, et al. Somatic activation of AKT3 causes hemispheric developmental brain malformations. Neuron 2012; 74: 41–48.
- 16D'Gama AM, Geng Y, Couto JA, et al. Mammalian target of rapamycin pathway mutations cause hemimegalencephaly and focal cortical dysplasia. Ann Neurol. 2015; 77: 720–725.
- 17Baulac S, Ishida S, Marsan E, et al. Familial focal epilepsy with focal cortical dysplasia due to DEPDC5 mutations. Ann Neurol 2015; 77: 675–683.
- 18Schick V, Majores M, Engels G, et al. Activation of Akt independent of PTEN and CTMP tumor-suppressor gene mutations in epilepsy-associated Taylor-type focal cortical dysplasias. Acta Neuropathol 2006; 112: 715–725.
- 19Jansen LA, Mirzaa GM, Ishak GE, et al. PI3K/AKT pathway mutations cause a spectrum of brain malformations from megalencephaly to focal cortical dysplasia. Brain 2015; 138(Pt 6): 1613–1628.
- 20Cibulskis K, Lawrence MS, Carter SL, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 2013; 31: 213–219.
- 21Koboldt DC, Zhang Q, Larson DE, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res 2012; 22: 568–576.
- 22Genomes Project C, Abecasis GR, Altshuler D, et al. A map of human genome variation from population-scale sequencing. Nature 2010; 467: 1061–1073.
- 23Genomes Project C, Abecasis GR, Auton A, et al. An integrated map of genetic variation from 1,092 human genomes. Nature 2012; 491: 56–65.
- 24Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res 2010; 38: e164.
- 25Thorvaldsdottir H, Robinson JT, Mesirov JP. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform 2013; 14: 178–192.
- 26Miyahara H, Natsumeda M, Shiga A, et al. Suppressed expression of autophagosomal protein LC3 in cortical tubers of tuberous sclerosis complex. Brain Pathol 2013; 23: 254–262.
- 27Guerois R, Nielsen JE, Serrano L. Predicting changes in the stability of proteins and protein complexes: a study of more than 1000 mutations. J Mol Biol 2002; 320: 369–387.
- 28Mizushima S, Nagata S. pEF-BOS, a powerful mammalian expression vector. Nucleic Acids Res 1990; 18: 5322.
- 29Eguchi S, Tokunaga C, Hidayat S, et al. Different roles for the TOS and RAIP motifs of the translational regulator protein 4E-BP1 in the association with raptor and phosphorylation by mTOR in the regulation of cell size. Genes Cells 2006; 11: 757–766.
- 30Takei N, Inamura N, Kawamura M, et al. Brain-derived neurotrophic factor induces mammalian target of rapamycin-dependent local activation of translation machinery and protein synthesis in neuronal dendrites. J Neurosci 2004; 24: 9760–9769.
- 31Yang H, Rudge DG, Koos JD, et al. mTOR kinase structure, mechanism and regulation. Nature 2013; 497: 217–223.
- 32Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell 2012; 149: 274–293.
- 33.Urano J, Sato T, Matsuo T, et al. Point mutations in TOR confer Rheb-independent growth in fission yeast and nutrient-independent mammalian TOR signaling in mammalian cells. Proc Natl Acad Sci U S A 2007; 104: 3514–3519.
- 34Ohne Y, Takahara T, Hatakeyama R, et al. Isolation of hyperactive mutants of mammalian target of rapamycin. J Biol Chem 2008; 283: 31861–31870.
- 35Reinke A, Chen JC, Aronova S, Powers T. Caffeine targets TOR complex I and provides evidence for a regulatory link between the FRB and kinase domains of Tor1p. J Biol Chem 2006; 281: 31616–31626.
- 36Lim JS, Kim WI, Kang HC, et al. Brain somatic mutations in MTOR cause focal cortical dysplasia type II leading to intractable epilepsy. Nat Med 2015; 21: 395–400.
- 37Scheffer IE, Heron SE, Regan BM, et al. Mutations in mammalian target of rapamycin regulator DEPDC5 cause focal epilepsy with brain malformations. Ann Neurol 2014; 75: 782–787.
- 38Salmena L, Carracedo A, Pandolfi PP. Tenets of PTEN tumor suppression. Cell 2008; 133: 403–414.
- 39Bar-Peled L, Chantranupong L, Cherniack AD, et al. A Tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1. Science 2013; 340: 1100–1106.
- 40Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol 2011; 12: 21–35.
- 41Loewith R, Jacinto E, Wullschleger S, et al. Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control. Mol Cell 2002; 10: 457–468.
- 42Grabiner BC, Nardi V, Birsoy K, et al. A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity. Cancer Discov 2014; 4: 554–563.