201
|
Lehalle D, Vabres P, Sorlin A, Bierhals T, Avila M, Carmignac V, Chevarin M, Torti E, Abe Y, Bartolomaeus T, Clayton-Smith J, Cogné B, Cusco I, Duplomb L, De Bont E, Duffourd Y, Duijkers F, Elpeleg O, Fattal A, Geneviève D, Guillen Sacoto MJ, Guimier A, Harris DJ, Hempel M, Isidor B, Jouan T, Kuentz P, Koshimizu E, Lichtenbelt K, Loik Ramey V, Maik M, Miyakate S, Murakami Y, Pasquier L, Pedro H, Simone L, Sondergaard-Schatz K, St-Onge J, Thevenon J, Valenzuela I, Abou Jamra R, van Gassen K, van Haelst MM, van Koningsbruggen S, Verdura E, Whelan Habela C, Zacher P, Rivière JB, Thauvin-Robinet C, Betschinger J, Faivre L. De novo mutations in the X-linked TFE3 gene cause intellectual disability with pigmentary mosaicism and storage disorder-like features. J Med Genet 2020; 57:808-819. [PMID: 32409512 DOI: 10.1136/jmedgenet-2019-106508] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 02/16/2020] [Accepted: 02/22/2020] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Pigmentary mosaicism (PM) manifests by pigmentation anomalies along Blaschko's lines and represents a clue toward the molecular diagnosis of syndromic intellectual disability (ID). Together with new insights on the role for lysosomal signalling in embryonic stem cell differentiation, mutations in the X-linked transcription factor 3 (TFE3) have recently been reported in five patients. Functional analysis suggested these mutations to result in ectopic nuclear gain of functions. MATERIALS AND METHODS Subsequent data sharing allowed the clustering of de novo TFE3 variants identified by exome sequencing on DNA extracted from leucocytes in patients referred for syndromic ID with or without PM. RESULTS We describe the detailed clinical and molecular data of 17 individuals harbouring a de novo TFE3 variant, including the patients that initially allowed reporting TFE3 as a new disease-causing gene. The 12 females and 5 males presented with pigmentation anomalies on Blaschko's lines, severe ID, epilepsy, storage disorder-like features, growth retardation and recognisable facial dysmorphism. The variant was at a mosaic state in at least two male patients. All variants were missense except one splice variant. Eleven of the 13 variants were localised in exon 4, 2 in exon 3, and 3 were recurrent variants. CONCLUSION This series further delineates the specific storage disorder-like phenotype with PM ascribed to de novo TFE3 mutation in exons 3 and 4. It confirms the identification of a novel X-linked human condition associated with mosaicism and dysregulation within the mechanistic target of rapamycin (mTOR) pathway, as well as a link between lysosomal signalling and human development.
Collapse
Affiliation(s)
- Daphné Lehalle
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France .,UF de Génétique Médicale, Département de Génétique, Groupe Hospitalier Pitié-Salpêtrière, APHP Sorbonne Université, Paris, France.,INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| | - Pierre Vabres
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France.,INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France.,Centre de Référence MAGEC, Service de Dermatologie, Centre Hospitalier Universitaire Dijon Bourgogne, Dijon, Bourgogne, France
| | - Arthur Sorlin
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France.,INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| | - Tatjana Bierhals
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, Germany
| | - Magali Avila
- INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| | - Virginie Carmignac
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France.,INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| | - Martin Chevarin
- INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| | | | - Yuichi Abe
- Division of Neurology, National Center for Child Health and Development, Tokyo, Japan
| | - Tobias Bartolomaeus
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Jill Clayton-Smith
- Genomic Medicine, Manchester Centre for Genomic Medicine, Manchester, Manchester, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, Greater Manchester, UK
| | - Benjamin Cogné
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Nantes, Nantes, France.,L'institut du thorax, INSERM, CNRS, Université de Nantes, Nantes, France
| | - Ivon Cusco
- Department of Clinical and Molecular Genetics and Rare Disease Unit, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Laurence Duplomb
- INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| | - Eveline De Bont
- Department of Pediatric Oncology, Ommelander Hospital Groningen, Scheemda, Groningen, The Netherlands
| | - Yannis Duffourd
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France.,INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| | - Floor Duijkers
- Department of Genetics, Amsterdam University Medical Centres, Amsterdam, Noord-Holland, The Netherlands
| | - Orly Elpeleg
- Monique and Jacques Roboh Department of Genetic Research, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Aviva Fattal
- Pediatric Neurology Institute, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - David Geneviève
- Departement de Génétique Medicale, Hôpital Arnaud de Villeneuve, CHRU Montpellier, Montpellier, France
| | | | - Anne Guimier
- Department of Genetics, Necker-Enfants Malades Hospitals, Paris, Île-de-France, France
| | - David J Harris
- Division of Genomics and Genetics, Boston Children s Hospital, Boston, Massachusetts, USA
| | - Maja Hempel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, Germany
| | - Bertrand Isidor
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Nantes, Nantes, France.,L'institut du thorax, INSERM, CNRS, Université de Nantes, Nantes, France
| | - Thibaud Jouan
- INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| | - Paul Kuentz
- INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France.,Génétique Biologique Histologie, PCBio, Centre Hospitalier Universitaire de Besancon, Besancon, France
| | - Eriko Koshimizu
- Department of Human Genetics, Yokohama City University School of Medicine Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Klaske Lichtenbelt
- Department of Genetics, University Medical Centre Utrecht Brain Centre, Utrecht, Utrecht, The Netherlands
| | - Valerie Loik Ramey
- Division of Genomics and Genetics, Boston Children s Hospital, Boston, Massachusetts, USA
| | - Miriam Maik
- Hackensack Meridian Health Inc, Edison, New Jersey, USA
| | - Sakoto Miyakate
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Yoshiko Murakami
- Yabumoto Department of Intractable Disease Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Laurent Pasquier
- Service de Génétique Clinique, CLAD Ouest, CHU Rennes, Rennes, France
| | - Helio Pedro
- Hackensack Meridian Health Inc, Edison, New Jersey, USA
| | - Laurie Simone
- Hackensack Meridian Health Inc, Edison, New Jersey, USA
| | - Krista Sondergaard-Schatz
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Judith St-Onge
- INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France.,Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Julien Thevenon
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France.,INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France.,Département de Génétique et Procréation, CHU Grenoble Alpes, Université Grenoble Alpes, Grenoble, France
| | - Irene Valenzuela
- Department of Clinical and Molecular Genetics and Rare Disease Unit, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Rami Abou Jamra
- Institute of Human Genetics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Koen van Gassen
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mieke M van Haelst
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Silvana van Koningsbruggen
- Department of Clinical Genetics, University of Amsterdam, Academic Medical Centre, Amsterdam, The Netherlands
| | - Edgard Verdura
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Christa Whelan Habela
- Department of Neurology, John M. Freeman Pediatric Epilepsy Center, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Pia Zacher
- The Saxon Epilepsy Center Kleinwachau, Radeberg, Germany
| | - Jean-Baptiste Rivière
- INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France.,Department of Human Genetics, McGill University Health Centre, Montreal, Quebec, Canada
| | - Christel Thauvin-Robinet
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France.,INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| | - Joerg Betschinger
- Friedrich Miescher Institute for Biomedical Research, Basel, Basel-Stadt, Switzerland
| | - Laurence Faivre
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France.,INSERM LNC UMR 1231, Faculté de Médecine, Université de Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
202
|
Abstract
Epilepsy encompasses a group of heterogeneous brain diseases that affect more than 50 million people worldwide. Epilepsy may have discernible structural, infectious, metabolic, and immune etiologies; however, in most people with epilepsy, no obvious cause is identifiable. Based initially on family studies and later on advances in gene sequencing technologies and computational approaches, as well as the establishment of large collaborative initiatives, we now know that genetics plays a much greater role in epilepsy than was previously appreciated. Here, we review the progress in the field of epilepsy genetics and highlight molecular discoveries in the most important epilepsy groups, including those that have been long considered to have a nongenetic cause. We discuss where the field of epilepsy genetics is moving as it enters a new era in which the genetic architecture of common epilepsies is starting to be unraveled.
Collapse
Affiliation(s)
- Piero Perucca
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria 3000, Australia.,Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, Victoria 3050, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria 3000, Australia
| | - Melanie Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Victoria 3084, Australia;
| |
Collapse
|
203
|
Tarkowski B, Kuchcinska K, Blazejczyk M, Jaworski J. Pathological mTOR mutations impact cortical development. Hum Mol Genet 2020; 28:2107-2119. [PMID: 30789219 DOI: 10.1093/hmg/ddz042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 02/03/2023] Open
Abstract
Several mosaic mutations of the mammalian/mechanistic target of rapamycin (mTOR) have recently been found in patients with cortical malformations, such as hemimegalencephaly (HME) and focal cortical dysplasia (FCD). Although all of them should activate mTOR signaling, comparisons of the impact of different mTOR mutations on brain development have been lacking. Also it remains unknown if any potential differences these mutations may have on cortical development are directly related to a degree of mTOR signaling increase. The present study assessed levels of mTORC1 pathway activity in cell lines and rat primary neurons overexpressing several mTOR mutants that were previously found in HME, FCD, cancer patients and in vitro mutagenesis screens. Next we introduced the mutants, enhancing mTORC1 signaling most potently, into developing mouse brains and assessed electroporated cell morphology and migratory phenotype using immunofluorescent staining. We observed the differential inhibition of neuronal progenitor cortical migration, which partly corresponded with a degree of mTORC1 signaling enhancement these mutants induced in cultured cells. The most potent quadruple mutant prevented most of the progenitors from entering the cortical plate. Cells that expressed less potent, single-point, mTOR mutants entered the cortical plate but failed to reach its upper layers and had enlarged soma. Our findings suggest a correlation between the potency of mTOR mutation to activate mTORC1 pathway and disruption of cortical migration.
Collapse
Affiliation(s)
- Bartosz Tarkowski
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Kinga Kuchcinska
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | | | - Jacek Jaworski
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| |
Collapse
|
204
|
Zhang Z, Gao K, Liu Q, Zhou J, Li X, Lang N, Liu M, Wang T, Zhang J, Wang H, Dong Y, Ji T, Wang S, Liu X, Jiang Y, Cai L, Wu Y. Somatic variants in new candidate genes identified in focal cortical dysplasia type II. Epilepsia 2020; 61:667-678. [PMID: 32216069 DOI: 10.1111/epi.16481] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 02/25/2020] [Accepted: 02/25/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Focal cortical dysplasia type II (FCDII) is a malformation of cortex development commonly found in children with drug-resistant epilepsy. FCDII has been associated with somatic mutations in mammalian target of rapamycin (mTOR)-related pathway genes and an upregulation of mTOR. Somatic mutations were found in 10%-63% of FCDII samples; the frequency of the mutant allele was 0.93%-33.5%. This study aimed to find new candidate genes involved in FCDII. METHODS We collected resected FCD lesions, perilesional brain tissues, and peripheral blood from 17 children with pathologically confirmed FCDII. We performed whole exome sequencing and followed a set of screening and analysis strategies to identify potentially deleterious somatic variants (PDSVs) in brain-expressed genes. We performed site-specific amplicon sequencing to validate the results. We also performed an in vitro functional study on an IRS1 variant. RESULTS In six of 17 samples, we identified seven PDSVs in seven genes, including two frameshift variants and five missense variants. The frequencies of the variant allele were 1.29%-5.50%. The genes were MTOR, TSC2, IRS1, RAB6B, RALA, HTR6, and ZNF337. PDSVs in IRS1, RAB6B, ZNF337, RALA, and HTR6 had not been previously associated with FCD. In one lesion, two PDSVs were found in two genes. In a transfected cell line, we demonstrated that the c.1791dupG (identified in FCDII from Patient 1) led to a truncated IRS1 and significant mTOR hyperactivation compared to cells that carried wild-type IRS1. mTOR was also activated in FCDII tissue from Patient 1. SIGNIFICANCE Seven PDSVs were identified in FCDII lesions in six of 17 children. Five variant genes had not been previously associated with cortical malformations. We demonstrated that the IRS1 variant led to mTOR hyperactivation in vitro. Although functional experiments are needed, the results provide evidence for novel candidate genes in the pathogenesis of FCDII.
Collapse
Affiliation(s)
- Zhongbin Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Qingzhu Liu
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Jiapeng Zhou
- College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiyuan Li
- Institute of Computing Technology, Chinese Academy of Science, Beijing, China
| | - Na Lang
- College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ming Liu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Tianshuang Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jie Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Hui Wang
- Department of Pathology, Peking University First Hospital, Beijing, China
| | - Ying Dong
- Department of Pathology, Peking University First Hospital, Beijing, China
| | - Taoyun Ji
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Shuang Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Xiaoyan Liu
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Lixin Cai
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| |
Collapse
|
205
|
Kabadi A, McDonnell E, Frank CL, Drowley L. Applications of Functional Genomics for Drug Discovery. SLAS DISCOVERY 2020; 25:823-842. [PMID: 32026742 DOI: 10.1177/2472555220902092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Many diseases, such as diabetes, autoimmune diseases, cancer, and neurological disorders, are caused by a dysregulation of a complex interplay of genes. Genome-wide association studies have identified thousands of disease-linked polymorphisms in the human population. However, detailing the causative gene expression or functional changes underlying those associations has been elusive in many cases. Functional genomics is an emerging field of research that aims to deconvolute the link between genotype and phenotype by making use of large -omic data sets and next-generation gene and epigenome editing tools to perturb genes of interest. Here we review how functional genomic tools can be used to better understand the biological interplay between genes, improve disease modeling, and identify novel drug targets. Incorporation of functional genomic capabilities into conventional drug development pipelines is predicted to expedite the development of first-in-class therapeutics.
Collapse
Affiliation(s)
- Ami Kabadi
- Element Genomics, a UCB company, Durham, NC, USA
| | | | | | | |
Collapse
|
206
|
Ganesan H, Balasubramanian V, Iyer M, Venugopal A, Subramaniam MD, Cho SG, Vellingiri B. mTOR signalling pathway - A root cause for idiopathic autism? BMB Rep 2020. [PMID: 31186084 PMCID: PMC6675248 DOI: 10.5483/bmbrep.2019.52.7.137] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental monogenic disorder with a strong genetic influence. Idiopathic autism could be defined as a type of autism that does not have a specific causative agent. Among signalling cascades, mTOR signalling pathway plays a pivotal role not only in cell cycle, but also in protein synthesis and regulation of brain homeostasis in ASD patients. The present review highlights, underlying mechanism of mTOR and its role in altered signalling cascades as a triggering factor in the onset of idiopathic autism. Further, this review discusses how distorted mTOR signalling pathway stimulates truncated translation in neuronal cells and leads to downregulation of protein synthesis at dendritic spines of the brain. This review concludes by suggesting downstream regulators such as p70S6K, eIF4B, eIF4E of mTOR signalling pathway as promising therapeutic targets for idiopathic autistic individuals. [BMB Reports 2019; 52(7): 424-433].
Collapse
Affiliation(s)
- Harsha Ganesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Venkatesh Balasubramanian
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Mahalaxmi Iyer
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | - Anila Venugopal
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Mohana Devi Subramaniam
- Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai 600006, Tamil Nadu, India
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| |
Collapse
|
207
|
Lee S, Kim SH, Kim B, Lee ST, Choi JR, Kim HD, Lee JS, Kang HC. Clinical Implementation of Targeted Gene Sequencing for Malformation of Cortical Development. Pediatr Neurol 2020; 103:27-34. [PMID: 31481326 DOI: 10.1016/j.pediatrneurol.2019.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 07/16/2019] [Accepted: 07/21/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND Malformations of cortical development comprise phenotypically heterogeneous conditions, and the diagnostic value of genetic testing in blood still remains to be elucidated. We used targeted gene sequencing to identify malformations of cortical development caused by germline mutations and characteristics associated with pathogenic mutations. METHODS A total of 81 patients with malformations of cortical development were included. Genomic DNA was isolated from peripheral blood. Ninety-six genes were assessed using a targeted next-generation sequencing panel. Single-nucleotide variants and exonic and chromosomal copy number variations were examined with our customized pipeline. RESULTS Genetic causes were identified from blood in 19 (23.5%) patients with malformations of cortical development; 14 patients had pathogenic or likely pathogenic single-nucleotide variants in seven genes, including DCX (n = 5), DEPDC5 (n = 2), PAFAH1B1 (n = 3), TUBA1A (n = 1), TUBA8 (n = 1), TUBB2B (n = 1), and TUBB3 (n = 1). Five patients had pathogenic copy number variations. Multifocal involvement of the lesion (tangential distribution, P < 0.001) and concurrent involvement of multiple structures such as the cortex, white matter, and ventricle (radial distribution, P = 0.003) were more commonly found in patients with identified genetic causes. Intellectual disability was also more commonly associated with pathogenic mutations (P = 0.048). In a multivariable regression analysis, both tangential and radial radiological distribution of malformations of cortical development were independently associated with positive germline test results. CONCLUSION We identified germline mutations in almost one-fourth of our patients with malformations of cortical development by using targeted gene sequencing. Germline abnormalities were more likely found in patients who had multifocal malformations of cortical development.
Collapse
Affiliation(s)
- Sangbo Lee
- Division of Pediatric Neurology, Epilepsy Research Institute, Severance Children's Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se Hee Kim
- Division of Pediatric Neurology, Epilepsy Research Institute, Severance Children's Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Borahm Kim
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heung Dong Kim
- Division of Pediatric Neurology, Epilepsy Research Institute, Severance Children's Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Soo Lee
- Division of Pediatric Neurology, Epilepsy Research Institute, Severance Children's Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hoon-Chul Kang
- Division of Pediatric Neurology, Epilepsy Research Institute, Severance Children's Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
208
|
Bonazzi S, Goold CP, Gray A, Thomsen NM, Nunez J, Karki RG, Gorde A, Biag JD, Malik HA, Sun Y, Liang G, Lubicka D, Salas S, Labbe-Giguere N, Keaney EP, McTighe S, Liu S, Deng L, Piizzi G, Lombardo F, Burdette D, Dodart JC, Wilson CJ, Peukert S, Curtis D, Hamann LG, Murphy LO. Discovery of a Brain-Penetrant ATP-Competitive Inhibitor of the Mechanistic Target of Rapamycin (mTOR) for CNS Disorders. J Med Chem 2020; 63:1068-1083. [PMID: 31955578 DOI: 10.1021/acs.jmedchem.9b01398] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recent clinical evaluation of everolimus for seizure reduction in patients with tuberous sclerosis complex (TSC), a disease with overactivated mechanistic target of rapamycin (mTOR) signaling, has demonstrated the therapeutic value of mTOR inhibitors for central nervous system (CNS) indications. Given that everolimus is an incomplete inhibitor of the mTOR function, we sought to develop a new mTOR inhibitor that has improved properties and is suitable for CNS disorders. Starting from an in-house purine-based compound, optimization of the physicochemical properties of a thiazolopyrimidine series led to the discovery of the small molecule 7, a potent and selective brain-penetrant ATP-competitive mTOR inhibitor. In neuronal cell-based models of mTOR hyperactivity, 7 corrected the mTOR pathway activity and the resulting neuronal overgrowth phenotype. The new mTOR inhibitor 7 showed good brain exposure and significantly improved the survival rate of mice with neuronal-specific ablation of the Tsc1 gene. These results demonstrate the potential utility of this tool compound to test therapeutic hypotheses that depend on mTOR hyperactivity in the CNS.
Collapse
Affiliation(s)
- Simone Bonazzi
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Carleton P Goold
- Neuroscience , Novartis Institutes for BioMedical Research , 22 Windsor Street , Cambridge , Massachusetts 02139 , United States
| | - Audrey Gray
- Neuroscience , Novartis Institutes for BioMedical Research , 22 Windsor Street , Cambridge , Massachusetts 02139 , United States
| | - Noel M Thomsen
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Jill Nunez
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Rajeshri G Karki
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Aakruti Gorde
- Neuroscience , Novartis Institutes for BioMedical Research , 22 Windsor Street , Cambridge , Massachusetts 02139 , United States
| | - Jonathan D Biag
- Neuroscience , Novartis Institutes for BioMedical Research , 22 Windsor Street , Cambridge , Massachusetts 02139 , United States
| | - Hasnain A Malik
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Yingchuan Sun
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Guiqing Liang
- Pharmacokinetic Sciences , Novartis Institutes for BioMedical Research , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Danuta Lubicka
- Global Drug Development/Technical Research and Development , Novartis Institutes for BioMedical Research , 700 Main Street , Cambridge , Massachusetts 02139 , United States
| | - Sarah Salas
- Neuroscience , Novartis Institutes for BioMedical Research , 22 Windsor Street , Cambridge , Massachusetts 02139 , United States
| | - Nancy Labbe-Giguere
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Erin P Keaney
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Stephanie McTighe
- Neuroscience , Novartis Institutes for BioMedical Research , 22 Windsor Street , Cambridge , Massachusetts 02139 , United States
| | - Shanming Liu
- Chemical Biology and Therapeutics , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Lin Deng
- Pharmacokinetic Sciences , Novartis Institutes for BioMedical Research , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Grazia Piizzi
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Franco Lombardo
- Pharmacokinetic Sciences , Novartis Institutes for BioMedical Research , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Doug Burdette
- Pharmacokinetic Sciences , Novartis Institutes for BioMedical Research , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Jean-Cosme Dodart
- Neuroscience , Novartis Institutes for BioMedical Research , 22 Windsor Street , Cambridge , Massachusetts 02139 , United States
| | - Christopher J Wilson
- Neuroscience , Novartis Institutes for BioMedical Research , 22 Windsor Street , Cambridge , Massachusetts 02139 , United States
| | - Stefan Peukert
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Daniel Curtis
- Neuroscience , Novartis Institutes for BioMedical Research , 22 Windsor Street , Cambridge , Massachusetts 02139 , United States
| | - Lawrence G Hamann
- Global Discovery Chemistry , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| | - Leon O Murphy
- Chemical Biology and Therapeutics , Novartis Institutes for BioMedical Research , 181 Massachusetts Ave , Cambridge , Massachusetts 02139 , United States
| |
Collapse
|
209
|
Subramanian L, Calcagnotto ME, Paredes MF. Cortical Malformations: Lessons in Human Brain Development. Front Cell Neurosci 2020; 13:576. [PMID: 32038172 PMCID: PMC6993122 DOI: 10.3389/fncel.2019.00576] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Creating a functional cerebral cortex requires a series of complex and well-coordinated developmental steps. These steps have evolved across species with the emergence of cortical gyrification and coincided with more complex behaviors. The presence of diverse progenitor cells, a protracted timeline for neuronal migration and maturation, and diverse neuronal types are developmental features that have emerged in the gyrated cortex. These factors could explain how the human brain has expanded in size and complexity. However, their complex nature also renders new avenues of vulnerability by providing additional cell types that could contribute to disease and longer time windows that could impact the composition and organization of the cortical circuit. We aim to discuss the unique developmental steps observed in human corticogenesis and propose how disruption of these species-unique processes could lead to malformations of cortical development.
Collapse
Affiliation(s)
- Lakshmi Subramanian
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
| | - Maria Elisa Calcagnotto
- Neurophysiology and Neurochemistry of Neuronal Excitability and Synaptic Plasticity Laboratory, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mercedes F. Paredes
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
- Neuroscience Graduate Division, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
210
|
Abstract
PURPOSE OF REVIEW There has been rapid progress in defining novel causative gene variants responsible for a large spectrum of human epilepsy syndromes and subtypes. Of particular interest is the discovery that somatic mutations, for example, noninherited mutations occurring in neuroglial progenitor cells during embryonic brain development, are highly linked to malformations of cortical development (MCD) such as focal cortical dysplasia (FCD) type II and hemimegalencephaly. RECENT FINDINGS Somatic gene variants have been identified in genes encoding regulatory proteins within the mechanistic target of rapamycin (mTOR) signaling cascade and have thus comprised the group classified as mTORopathies. FCD II and hemimegalencephaly often result from mutations in identical genes suggesting that these are spectrum disorders. An exciting recent development has been the identification of somatic mutations causing both FCD Ia and nonlesional neocortical epilepsy. SUMMARY Defining somatic gene mutations in brain tissue specimens has shed new light on how MCD form and the mechanisms of epileptogenesis associated with MCD. Trials of mTOR inhibitors in tuberous sclerosis complex have demonstrated that inhibition of mTOR activation in mTORopathies can reduce seizure frequency. New somatic mutations found for a variety of epilepsy syndromes may provide new targets for clinical therapeutics.
Collapse
|
211
|
Campbell C, Cavalleri GL, Delanty N. Exploring the genetic overlap between psychiatric illness and epilepsy: A review. Epilepsy Behav 2020; 102:106669. [PMID: 31785486 DOI: 10.1016/j.yebeh.2019.106669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
Abstract
There is a long-documented epidemiological link between epilepsy and psychiatric disorders. People with epilepsy are at an increased risk for a variety of psychiatric illnesses, as are their family members, and people with epilepsy may experience psychiatric side effects because of their antiepileptic drugs (AEDs). In recent years, large-scale, collaborative international studies have begun to shed light on the role of genetic variation in both epilepsy and psychiatric illnesses, such as schizophrenia, depression, and anxiety. But so far, finding shared genetic links between epilepsy and psychiatric illness has proven surprisingly difficult. This review will discuss the prevalence of psychiatric comorbidities in epilepsy, recent advances in genetic research into both epilepsy and psychiatric illness, and the extent of our current knowledge of the genetic overlap between these two important neurobiological conditions.
Collapse
Affiliation(s)
- Ciarán Campbell
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Molecular and Cellular Therapeutics, RCSI Dublin, Ireland
| | - Gianpiero L Cavalleri
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Molecular and Cellular Therapeutics, RCSI Dublin, Ireland
| | - Norman Delanty
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland; Department of Molecular and Cellular Therapeutics, RCSI Dublin, Ireland; Department of Neurology, Beaumont Hospital, Dublin, Ireland.
| |
Collapse
|
212
|
|
213
|
Donkels C, Peters M, Fariña Núñez MT, Nakagawa JM, Kirsch M, Vlachos A, Scheiwe C, Schulze-Bonhage A, Prinz M, Beck J, Haas CA. Oligodendrocyte lineage and myelination are compromised in the gray matter of focal cortical dysplasia type IIa. Epilepsia 2019; 61:171-184. [PMID: 31872870 DOI: 10.1111/epi.16415] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 01/26/2023]
Abstract
OBJECTIVES Focal cortical dysplasias (FCDs) are local malformations of the human neocortex and a leading cause of medically intractable epilepsy. FCDs are characterized by local architectural disturbances of the neocortex and often by a blurred gray-white matter boundary indicating abnormal white matter myelination. We have recently shown that myelination is also compromised in the gray matter of dysplastic areas, since transcripts encoding factors for oligodendrocyte differentiation and myelination are downregulated and myelin fibers appear fractured and disorganized. METHODS Here, we characterized the gray matter-associated myelination pathology in detail by in situ hybridization, immunohistochemistry, and electron microscopy with markers for myelin, mature oligodendrocytes, and oligodendrocyte precursor cells in tissue sections of FCD IIa and control cortices. In addition, we isolated oligodendrocyte precursor cells from resected dysplastic tissue and performed proliferation assays. RESULTS We show that the proportion of myelinated gray matter is similar in the dysplastic cortex to that in controls and myelinated fibers extend up to layer III. On the ultrastructural level, however, we found that the myelin sheaths of layer V axons are thinner in dysplastic specimens than in controls. In addition, the density of oligodendrocyte precursor cells and of mature oligodendrocytes was reduced. Finally, we show for the first time that oligodendrocyte precursor cells isolated from resected dysplastic cortex have a reduced proliferation capacity in comparison to controls. SIGNIFICANCE These results indicate that proliferation and differentiation of oligodendrocyte precursor cells and the formation of myelin sheaths are compromised in FCD and might contribute to the epileptogenicity of this cortical malformation.
Collapse
Affiliation(s)
- Catharina Donkels
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Myriam Peters
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mateo T Fariña Núñez
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia M Nakagawa
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matthias Kirsch
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Scheiwe
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Schulze-Bonhage
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Epilepsy Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BrainLinks-BrainTools, Cluster of Excellence, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Neuropathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Jürgen Beck
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BrainLinks-BrainTools, Cluster of Excellence, University of Freiburg, Freiburg, Germany
| |
Collapse
|
214
|
Neal A, Ostrowsky-Coste K, Jung J, Lagarde S, Maillard L, Kahane P, Touraine R, Catenoix H, Montavont A, Isnard J, Arzimanoglou A, Bartolomei F, Guenot M, Rheims S. Epileptogenicity in tuberous sclerosis complex: A stereoelectroencephalographic study. Epilepsia 2019; 61:81-95. [PMID: 31860139 DOI: 10.1111/epi.16410] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/19/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE In tuberous sclerosis complex (TSC)-associated drug-resistant epilepsy, the optimal invasive electroencephalographic (EEG) and operative approach remains unclear. We examined the role of stereo-EEG in TSC and used stereo-EEG data to investigate tuber and surrounding cortex epileptogenicity. METHODS We analyzed 18 patients with TSC who underwent stereo-EEG (seven adults). One hundred ten seizures were analyzed with the epileptogenicity index (EI). In 13 patients with adequate tuber sampling, five anatomical regions of interest (ROIs) were defined: dominant tuber (tuber with highest median EI), perituber cortex, secondary tuber (tuber with second highest median EI), nearby cortex (normal-appearing cortex in the same lobe as dominant tuber), and distant cortex (in other lobes). At the seizure level, epileptogenicity of ROIs was examined by comparing the highest EI recorded within each anatomical region. At the patient level, epileptogenic zone (EZ) organization was separated into focal tuber (EZ confined to dominant tuber) and complex (all other patterns). RESULTS The most epileptogenic ROI was the dominant tuber, with higher EI than perituber cortex, secondary tuber, nearby cortex, and distant cortex (P < .001). A focal tuber EZ organization was identified in seven patients. This group had 80% Engel IA postsurgical outcome and distinct dominant tuber characteristics: continuous interictal discharges (IEDs; 100%), fluid-attenuated inversion recovery (FLAIR) hypointense center (86%), center-to-rim EI gradient, and stimulation-induced seizures (71%). In contrast, six patients had a complex EZ organization, characterized by nearby cortex as the most epileptogenic region and 40% Engel IA outcome. At the intratuber level, the combination of FLAIR hypointense center, continuous IEDs, and stimulation-induced seizures offered 98% specificity for a focal tuber EZ organization. SIGNIFICANCE Tubers with focal EZ organization have a striking similarity to type II focal cortical dysplasia. The presence of distinct EZ organizations has significant implications for EZ hypothesis generation, invasive EEG approach, and resection strategy.
Collapse
Affiliation(s)
- Andrew Neal
- National Institute of Health and Medical Research U1028/National Center for Scientific Research, Mixed Unit of Research 5292, Lyon Neuroscience Research Center, Lyon, France.,Department of Functional Neurology and Epileptology, Member of the ERN EpiCARE Lyon University Hospital and Lyon 1 University, Lyon, France.,Department of Neuroscience, Faculty of Medicine, Nursing, and Health Sciences, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Karine Ostrowsky-Coste
- Department of Functional Neurology and Epileptology, Member of the ERN EpiCARE Lyon University Hospital and Lyon 1 University, Lyon, France.,Department of Pediatric Clinical Epileptology, Sleep Disorders, and Functional Neurology, Member of the ERN EpiCARE Lyon University Hospital, Lyon, France
| | - Julien Jung
- National Institute of Health and Medical Research U1028/National Center for Scientific Research, Mixed Unit of Research 5292, Lyon Neuroscience Research Center, Lyon, France.,Department of Functional Neurology and Epileptology, Member of the ERN EpiCARE Lyon University Hospital and Lyon 1 University, Lyon, France
| | - Stanislas Lagarde
- Epileptology Department, Public Assistance Hospitals of Marseille, National Institute of Health and Medical Research, Institute of Systems Neuroscience, Timone Hospital, Aix Marseille University, Marseille, France
| | - Louis Maillard
- Neurology Department, University Hospital of Nancy, Nancy, France
| | - Philippe Kahane
- Department of Neurology, Grenoble-Alpes University Hospital, Grenoble Institute of Neurosciences, National Institute of Health and Medical Research U1216, Grenoble Alpes University, Grenoble, France
| | - Renaud Touraine
- Department of Genetics, University Hospital Center-North Hospital, Saint Etienne, France
| | - Helene Catenoix
- National Institute of Health and Medical Research U1028/National Center for Scientific Research, Mixed Unit of Research 5292, Lyon Neuroscience Research Center, Lyon, France.,Department of Functional Neurology and Epileptology, Member of the ERN EpiCARE Lyon University Hospital and Lyon 1 University, Lyon, France
| | - Alexandra Montavont
- National Institute of Health and Medical Research U1028/National Center for Scientific Research, Mixed Unit of Research 5292, Lyon Neuroscience Research Center, Lyon, France.,Department of Functional Neurology and Epileptology, Member of the ERN EpiCARE Lyon University Hospital and Lyon 1 University, Lyon, France
| | - Jean Isnard
- National Institute of Health and Medical Research U1028/National Center for Scientific Research, Mixed Unit of Research 5292, Lyon Neuroscience Research Center, Lyon, France.,Department of Functional Neurology and Epileptology, Member of the ERN EpiCARE Lyon University Hospital and Lyon 1 University, Lyon, France
| | - Alexis Arzimanoglou
- National Institute of Health and Medical Research U1028/National Center for Scientific Research, Mixed Unit of Research 5292, Lyon Neuroscience Research Center, Lyon, France.,Department of Pediatric Clinical Epileptology, Sleep Disorders, and Functional Neurology, Member of the ERN EpiCARE Lyon University Hospital, Lyon, France
| | - Fabrice Bartolomei
- Epileptology Department, Public Assistance Hospitals of Marseille, National Institute of Health and Medical Research, Institute of Systems Neuroscience, Timone Hospital, Aix Marseille University, Marseille, France
| | - Marc Guenot
- National Institute of Health and Medical Research U1028/National Center for Scientific Research, Mixed Unit of Research 5292, Lyon Neuroscience Research Center, Lyon, France.,Department of Functional Neurosurgery, Member of the ERN EpiCARE Lyon University Hospital and Lyon 1 University, Lyon, France
| | - Sylvain Rheims
- National Institute of Health and Medical Research U1028/National Center for Scientific Research, Mixed Unit of Research 5292, Lyon Neuroscience Research Center, Lyon, France.,Department of Functional Neurology and Epileptology, Member of the ERN EpiCARE Lyon University Hospital and Lyon 1 University, Lyon, France.,Idée Epilepsy Institute, Lyon, France
| |
Collapse
|
215
|
Extracellular Vesicles in the Forebrain Display Reduced miR-346 and miR-331-3p in a Rat Model of Chronic Temporal Lobe Epilepsy. Mol Neurobiol 2019; 57:1674-1687. [PMID: 31813125 DOI: 10.1007/s12035-019-01797-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/22/2019] [Indexed: 12/20/2022]
Abstract
An initial precipitating injury in the brain, such as after status epilepticus (SE), evolves into chronic temporal lobe epilepsy (TLE). We investigated changes in the miRNA composition of extracellular vesicles (EVs) in the forebrain after the establishment of SE-induced chronic TLE. We induced SE in young Fischer 344 rats through graded intraperitoneal injections of kainic acid, which resulted in consistent spontaneous recurrent seizures at ~ 3 months post-SE. We isolated EVs from the entire forebrain of chronically epileptic rats and age-matched naïve control animals through an ultracentrifugation method and performed miRNA-sequencing studies to discern changes in the miRNA composition of forebrain-derived EVs in chronic epilepsy. EVs from both naïve and epileptic forebrains displayed spherical or cup-shaped morphology, a comparable size range, and CD63 expression but lacked the expression of a deep cellular marker GM130. However, miRNA-sequencing studies suggested downregulation of 3 miRNAs (miR-187-5p, miR-346, and miR-331-3p) and upregulation of 4 miRNAs (miR-490-5p, miR-376b-3p, miR-493-5p, and miR-124-5p) in EVs from epileptic forebrains with fold changes ranging from 1.5 to 2.4 (p < 0.0006; FDR < 0.05). By using geNorm and Normfinder software, we identified miR-487 and miR-221 as the best combination of reference genes for measurement of altered miRNAs found in the epileptic forebrain through qRT-PCR studies. The validation revealed that only miR-346 and miR-331-3p were significantly downregulated in EVs from the epileptic forebrain. The enrichment pathway analysis of these miRNAs showed an overrepresentation of signaling pathways that are linked to molecular mechanisms underlying chronic epilepsy, including GABA-ergic (miR-346 targets) and mTOR (miR-331-3p targets) systems. Thus, the packaging of two miRNAs into EVs in neural cells is considerably altered in chronic epilepsy. Functional studies on these two miRNAs may uncover their role in the pathophysiology and treatment of TLE.
Collapse
|
216
|
Precise detection of low-level somatic mutation in resected epilepsy brain tissue. Acta Neuropathol 2019; 138:901-912. [PMID: 31377847 DOI: 10.1007/s00401-019-02052-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/11/2019] [Accepted: 07/29/2019] [Indexed: 01/16/2023]
Abstract
Low-level somatic mutations have been shown to be the major genetic etiology of intractable epilepsy. The extents thereof, however, have yet to be systematically and accurately explored in a large cohort of resected epilepsy brain tissues. Moreover, clinically useful and precise analysis tools for detecting low-level somatic mutations from unmatched formalin-fixed paraffin-embedded (FFPE) brain samples, the most clinically relevant samples, are still lacking. In total, 446 tissues samples from 232 intractable epilepsy patients with various brain pathologies were analyzed using deep sequencing (average read depth, 1112x) of known epilepsy-related genes (up to 28 genes) followed by confirmatory site-specific amplicon sequencing. Pathogenic mutations were discovered in 31.9% (74 of 232) of the resected epilepsy brain tissues and were recurrently found in only eight major focal epilepsy genes, including AKT3, DEPDC5, MTOR, PIK3CA, TSC1, TSC2, SCL35A2, and BRAF. Somatic mutations, two-hit mutations, and germline mutations accounted for 22.0% (51), 0.9% (2), and 9.1% (21) of the patients with intractable epilepsy, respectively. The majority of pathogenic somatic mutations (62.3%, 33 of 53) had a low variant allelic frequency of less than 5%. The use of deep sequencing replicates in the eight major focal epilepsy genes robustly increased PPVs to 50-100% and sensitivities to 71-100%. In an independent FCDII cohort of only unmatched FFPE brain tissues, deep sequencing replicates in the eight major focal epilepsy genes identified pathogenic somatic mutations in 33.3% (5 of 15) of FCDII individuals (similar to the genetic detecting rate in the entire FCDII cohort) without any false-positive calls. Deep sequencing replicates of major focal epilepsy genes in unmatched FFPE brain tissues can be used to accurately and efficiently detect low-level somatic mutations, thereby improving overall patient care by enriching genetic counseling and informing treatment decisions.
Collapse
|
217
|
Baldassari S, Ribierre T, Marsan E, Adle-Biassette H, Ferrand-Sorbets S, Bulteau C, Dorison N, Fohlen M, Polivka M, Weckhuysen S, Dorfmüller G, Chipaux M, Baulac S. Dissecting the genetic basis of focal cortical dysplasia: a large cohort study. Acta Neuropathol 2019; 138:885-900. [PMID: 31444548 PMCID: PMC6851393 DOI: 10.1007/s00401-019-02061-5] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/05/2019] [Accepted: 08/11/2019] [Indexed: 12/24/2022]
Abstract
Genetic malformations of cortical development (MCDs), such as mild MCDs (mMCD), focal cortical dysplasia (FCD), and hemimegalencephaly (HME), are major causes of severe pediatric refractory epilepsies subjected to neurosurgery. FCD2 are characterized by neuropathological hallmarks that include enlarged dysmorphic neurons (DNs) and balloon cells (BCs). Here, we provide a comprehensive assessment of the contribution of germline and somatic variants in a large cohort of surgical MCD cases. We enrolled in a monocentric study 80 children with drug-resistant epilepsy and a postsurgical neuropathological diagnosis of mMCD, FCD1, FCD2, or HME. We performed targeted gene sequencing ( ≥ 2000X read depth) on matched blood-brain samples to search for low-allele frequency variants in mTOR pathway and FCD genes. We were able to elucidate 29% of mMCD/FCD1 patients and 63% of FCD2/HME patients. Somatic loss-of-function variants in the N-glycosylation pathway-associated SLC35A2 gene were found in mMCD/FCD1 cases. Somatic gain-of-function variants in MTOR and its activators (AKT3, PIK3CA, RHEB), as well as germline, somatic and two-hit loss-of-function variants in its repressors (DEPDC5, TSC1, TSC2) were found exclusively in FCD2/HME cases. We show that panel-negative FCD2 cases display strong pS6-immunostaining, stressing that all FCD2 are mTORopathies. Analysis of microdissected cells demonstrated that DNs and BCs carry the pathogenic variants. We further observed a correlation between the density of pathological cells and the variant-detection likelihood. Single-cell microdissection followed by sequencing of enriched pools of DNs unveiled a somatic second-hit loss-of-heterozygosity in a DEPDC5 germline case. In conclusion, this study indicates that mMCD/FCD1 and FCD2/HME are two distinct genetic entities: while all FCD2/HME are mosaic mTORopathies, mMCD/FCD1 are not caused by mTOR-pathway-hyperactivating variants, and ~ 30% of the cases are related to glycosylation defects. We provide a framework for efficient genetic testing in FCD/HME, linking neuropathology to genetic findings and emphasizing the usefulness of molecular evaluation in the pediatric epileptic neurosurgical population.
Collapse
Affiliation(s)
- Sara Baldassari
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, Paris, France
- INSERM, U1127, Paris, France
- CNRS, UMR 7225, Paris, France
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière-47, bd de l'hôpital, 75013, Paris, France
| | - Théo Ribierre
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, Paris, France
- INSERM, U1127, Paris, France
- CNRS, UMR 7225, Paris, France
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière-47, bd de l'hôpital, 75013, Paris, France
| | - Elise Marsan
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, Paris, France
- INSERM, U1127, Paris, France
- CNRS, UMR 7225, Paris, France
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière-47, bd de l'hôpital, 75013, Paris, France
| | - Homa Adle-Biassette
- INSERM UMR 1141, Hôpital Robert-Debré, 75019, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
- Service d'Anatomie et de Cytologie Pathologiques, Hôpital Lariboisière, APHP, 75010, Paris, France
| | - Sarah Ferrand-Sorbets
- Department of Pediatric Neurosurgery, Rothschild Foundation Hospital, 75019, Paris, France
| | - Christine Bulteau
- Department of Pediatric Neurosurgery, Rothschild Foundation Hospital, 75019, Paris, France
| | - Nathalie Dorison
- Department of Pediatric Neurosurgery, Rothschild Foundation Hospital, 75019, Paris, France
| | - Martine Fohlen
- Department of Pediatric Neurosurgery, Rothschild Foundation Hospital, 75019, Paris, France
| | - Marc Polivka
- Service d'Anatomie et de Cytologie Pathologiques, Hôpital Lariboisière, APHP, 75010, Paris, France
| | - Sarah Weckhuysen
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, Paris, France
- INSERM, U1127, Paris, France
- CNRS, UMR 7225, Paris, France
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière-47, bd de l'hôpital, 75013, Paris, France
- Department of Neurology, University Hospital Antwerp, Antwerp, Belgium
| | - Georg Dorfmüller
- Department of Pediatric Neurosurgery, Rothschild Foundation Hospital, 75019, Paris, France
| | - Mathilde Chipaux
- Department of Pediatric Neurosurgery, Rothschild Foundation Hospital, 75019, Paris, France
| | - Stéphanie Baulac
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, Paris, France.
- INSERM, U1127, Paris, France.
- CNRS, UMR 7225, Paris, France.
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière-47, bd de l'hôpital, 75013, Paris, France.
| |
Collapse
|
218
|
Pelorosso C, Watrin F, Conti V, Buhler E, Gelot A, Yang X, Mei D, McEvoy-Venneri J, Manent JB, Cetica V, Ball LL, Buccoliero AM, Vinck A, Barba C, Gleeson JG, Guerrini R, Represa A. Somatic double-hit in MTOR and RPS6 in hemimegalencephaly with intractable epilepsy. Hum Mol Genet 2019; 28:3755-3765. [PMID: 31411685 PMCID: PMC6935386 DOI: 10.1093/hmg/ddz194] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 01/19/2023] Open
Abstract
Single germline or somatic activating mutations of mammalian target of rapamycin (mTOR) pathway genes are emerging as a major cause of type II focal cortical dysplasia (FCD), hemimegalencephaly (HME) and tuberous sclerosis complex (TSC). A double-hit mechanism, based on a primary germline mutation in one allele and a secondary somatic hit affecting the other allele of the same gene in a small number of cells, has been documented in some patients with TSC or FCD. In a patient with HME, severe intellectual disability, intractable seizures and hypochromic skin patches, we identified the ribosomal protein S6 (RPS6) p.R232H variant, present as somatic mosaicism at ~15.1% in dysplastic brain tissue and ~11% in blood, and the MTOR p.S2215F variant, detected as ~8.8% mosaicism in brain tissue, but not in blood. Overexpressing the two variants independently in animal models, we demonstrated that MTOR p.S2215F caused neuronal migration delay and cytomegaly, while RPS6 p.R232H prompted increased cell proliferation. Double mutants exhibited a more severe phenotype, with increased proliferation and migration defects at embryonic stage and, at postnatal stage, cytomegalic cells exhibiting eccentric nuclei and binucleation, which are typical features of balloon cells. These findings suggest a synergistic effect of the two variants. This study indicates that, in addition to single activating mutations and double-hit inactivating mutations in mTOR pathway genes, severe forms of cortical dysplasia can also result from activating mutations affecting different genes in this pathway. RPS6 is a potential novel disease-related gene.
Collapse
Affiliation(s)
- Cristiana Pelorosso
- Paediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children’s Hospital A. Meyer, University of Florence, Florence 50139, Italy
| | - Françoise Watrin
- INMED, Aix-Marseille University, INSERM UMR1249, Marseille 13009, France
| | - Valerio Conti
- Paediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children’s Hospital A. Meyer, University of Florence, Florence 50139, Italy
| | - Emmanuelle Buhler
- INMED, Aix-Marseille University, INSERM UMR1249, Marseille 13009, France
| | - Antoinette Gelot
- Service d'Anatomie Pathologique, Hôpital Trousseau, Hôpitaux Universitaires de l'Est Parisien, Université Pierre et Marie Curie, Paris 75012, France
| | - Xiaoxu Yang
- Department of Neuroscience, Howard Hughes Medical Institute, Rady Children’s Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Davide Mei
- Paediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children’s Hospital A. Meyer, University of Florence, Florence 50139, Italy
| | - Jennifer McEvoy-Venneri
- Department of Neuroscience, Howard Hughes Medical Institute, Rady Children’s Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | | | - Valentina Cetica
- Paediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children’s Hospital A. Meyer, University of Florence, Florence 50139, Italy
| | - Laurel L Ball
- Department of Neuroscience, Howard Hughes Medical Institute, Rady Children’s Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Anna Maria Buccoliero
- Pathology Unit, Children’s Hospital A. Meyer, University of Florence, Florence 50139, Italy
| | - Antonin Vinck
- INMED, Aix-Marseille University, INSERM UMR1249, Marseille 13009, France
| | - Carmen Barba
- Paediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children’s Hospital A. Meyer, University of Florence, Florence 50139, Italy
| | - Joseph G Gleeson
- Department of Neuroscience, Howard Hughes Medical Institute, Rady Children’s Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Renzo Guerrini
- Paediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Children’s Hospital A. Meyer, University of Florence, Florence 50139, Italy
- IRCCS Fondazione Stella Maris, Pisa 56126, Italy
| | - Alfonso Represa
- INMED, Aix-Marseille University, INSERM UMR1249, Marseille 13009, France
| |
Collapse
|
219
|
Fernández-Marmiesse A, Roca I, Díaz-Flores F, Cantarín V, Pérez-Poyato MS, Fontalba A, Laranjeira F, Quintans S, Moldovan O, Felgueroso B, Rodríguez-Pedreira M, Simón R, Camacho A, Quijada P, Ibanez-Mico S, Domingno MR, Benito C, Calvo R, Pérez-Cejas A, Carrasco ML, Ramos F, Couce ML, Ruiz-Falcó ML, Gutierrez-Solana L, Martínez-Atienza M. Rare Variants in 48 Genes Account for 42% of Cases of Epilepsy With or Without Neurodevelopmental Delay in 246 Pediatric Patients. Front Neurosci 2019; 13:1135. [PMID: 31780880 PMCID: PMC6856296 DOI: 10.3389/fnins.2019.01135] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/08/2019] [Indexed: 12/21/2022] Open
Abstract
In order to characterize the genetic architecture of epilepsy in a pediatric population from the Iberian Peninsula (including the Canary Islands), we conducted targeted exome sequencing of 246 patients with infantile-onset seizures with or without neurodevelopmental delay. We detected 107 variants in 48 different genes, which were implicated in neuronal excitability, neurodevelopment, synaptic transmission, and metabolic pathways. In 104 cases (42%) we detected variant(s) that we classified as pathogenic or likely pathogenic. Of the 48 mutated genes, 32 were dominant, 8 recessive and 8 X-linked. Of the patients for whom family studies could be performed and in whom pathogenic variants were identified in dominant or X-linked genes, 82% carried de novo mutations. The involvement of small copy number variations (CNVs) is 9%. The use of progressively updated custom panels with high mean vertical coverage enabled establishment of a definitive diagnosis in a large proportion of cases (42%) and detection of CNVs (even duplications) with high fidelity. In 10.5% of patients we detected associations that are pending confirmation via functional and/or familial studies. Our findings had important consequences for the clinical management of the probands, since a large proportion of the cohort had been clinically misdiagnosed, and their families were subsequently able to avail of genetic counseling. In some cases, a more appropriate treatment was selected for the patient in question, or an inappropriate treatment discontinued. Our findings suggest the existence of modifier genes that may explain the incomplete penetrance of some epilepsy-related genes. We discuss possible reasons for non-diagnosis and future research directions. Further studies will be required to uncover the roles of structural variants, epimutations, and oligogenic inheritance in epilepsy, thereby providing a more complete molecular picture of this disease. In summary, given the broad phenotypic spectrum of most epilepsy-related genes, efficient genomic tools like the targeted exome sequencing panel described here are essential for early diagnosis and treatment, and should be implemented as first-tier diagnostic tools for children with epilepsy without a clear etiologic basis.
Collapse
Affiliation(s)
- Ana Fernández-Marmiesse
- Unit for the Diagnosis and Treatment of Congenital Metabolic Diseases, Clinical University Hospital of Santiago de Compostela, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.,Genomes & Disease Group, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), Santiago de Compostela University-IDIS, Santiago de Compostela, Spain
| | - Iria Roca
- Unit for the Diagnosis and Treatment of Congenital Metabolic Diseases, Clinical University Hospital of Santiago de Compostela, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain.,Genomes & Disease Group, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), Santiago de Compostela University-IDIS, Santiago de Compostela, Spain
| | - Felícitas Díaz-Flores
- Molecular Genetics Unit, Clinical University Hospital of Canarias, Santa Cruz de Tenerife, Spain
| | - Verónica Cantarín
- Neuropediatrics Unit, Niño Jesús Clinical University Hospital, Madrid, Spain
| | | | - Ana Fontalba
- Genetics Unit, Marqués de Valdecilla Clinical University Hospital, Santander, Spain
| | - Francisco Laranjeira
- Centro de Genética Médica Jacinto Magalhães, Centro Hospitalar Do Porto, Porto, Portugal
| | - Sofia Quintans
- Neuropediatrics Unit, Santa María Hospital, Lisbon, Portugal
| | - Oana Moldovan
- Genetics Unit, Santa María Hospital, Lisbon, Portugal
| | - Blanca Felgueroso
- Neuropediatrics Unit, Teresa Herrera Child's Hospital, A Coruña, Spain
| | | | - Rogelio Simón
- Neuropediatrics Unit, 12 de Octubre Clinical University Hospital, Madrid, Spain
| | - Ana Camacho
- Neuropediatrics Unit, 12 de Octubre Clinical University Hospital, Madrid, Spain.,Department of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Pilar Quijada
- Metabolic Disorders Unit, 12 de Octubre Clinical University Hospital, Madrid, Spain
| | - Salvador Ibanez-Mico
- Neuropediatrics Unit, Virgen de la Arrixaca Clinical University Hospital, Murcia, Spain
| | - Mª Rosario Domingno
- Neuropediatrics Unit, Virgen de la Arrixaca Clinical University Hospital, Murcia, Spain
| | - Carmen Benito
- Genetics Unit, Clinical University Hospital of Málaga, Málaga, Spain
| | - Rocío Calvo
- Neuropediatrics Unit, Clinical University Hospital of Málaga, Málaga, Spain
| | - Antonia Pérez-Cejas
- Molecular Genetics Unit, Clinical University Hospital of Canarias, Santa Cruz de Tenerife, Spain
| | - Mª Llanos Carrasco
- Neuropediatrics Unit, Clinical University Hospital Severo Ochoa, Leganés, Madrid, Spain
| | - Feliciano Ramos
- Clinical Genetics Unit, Pediatrics, Clinical University Hospital of Zaragoza, Zaragoza, Spain
| | - Mª Luz Couce
- Unit for the Diagnosis and Treatment of Congenital Metabolic Diseases, Clinical University Hospital of Santiago de Compostela, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Mª Luz Ruiz-Falcó
- Neuropediatrics Unit, Niño Jesús Clinical University Hospital, Madrid, Spain
| | | | - Margarita Martínez-Atienza
- Genomes & Disease Group, Molecular Medicine and Chronic Diseases Research Centre (CiMUS), Santiago de Compostela University-IDIS, Santiago de Compostela, Spain.,Molecular Genetics Unit, Clinical University Hospital of Canarias, Santa Cruz de Tenerife, Spain.,Molecular Genetics Unit, Virgen de las Nieves Clinical University Hospital, Granada, Spain
| |
Collapse
|
220
|
Iffland PH, Carson V, Bordey A, Crino PB. GATORopathies: The role of amino acid regulatory gene mutations in epilepsy and cortical malformations. Epilepsia 2019; 60:2163-2173. [PMID: 31625153 PMCID: PMC7155771 DOI: 10.1111/epi.16370] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/20/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
The mechanistic target of rapamycin (mTOR) pathway has been implicated in a growing number of malformations of cortical development (MCD) associated with intractable epilepsy. Mutations in single genes encoding mTOR pathway regulatory proteins have been linked to MCD such as focal cortical dysplasia (FCD) types IIa and IIb, hemimegalencephaly (HME), and megalencephaly. Recent studies have demonstrated that the GATOR1 protein complex, comprised of DEPDC5, NPRL3, and NPRL2, plays a pivotal role in regulating mTOR signaling in response to cellular amino acid levels and that mutations in DEPDC5, NPRL3, or NPRL2 are linked to FCD, HME, and seizures. Histopathological analysis of FCD and HME tissue specimens resected from individuals harboring DEPDC5, NPRL3, or NPRL2 gene mutations reveals hyperactivation of mTOR pathway signaling. Family pedigrees carrying mutations in either DEPDC5 or NPRL3 share clinical phenotypes of epilepsy and MCD, as well as intellectual and neuropsychiatric disabilities. Interestingly, some individuals with seizures associated with DEPDC5, NPRL3, or NPRL2 variants exhibit normal brain imaging suggesting either occult MCD or a role for these genes in non-lesional neocortical epilepsy. Mouse models resulting from knockdown or knockout of either Depdc5 or Nprl3 exhibit altered cortical lamination, neuronal dysmorphogenesis, and enhanced neuronal excitability as reported in models resulting from direct mTOR activation through expression of its canonical activator RHEB. The role of the GATOR1 proteins in regulating mTOR signaling suggest plausible options for mTOR inhibition in the treatment of epilepsy associated with mutations in DEPDC5, NPRL3, or NPRL2.
Collapse
Affiliation(s)
- Philip H. Iffland
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Vincent Carson
- The Clinic for Special Children, Strasburg, Pennsylvania
| | - Angelique Bordey
- Department of Neurosurgery, Yale University School of Medicine, New Haven, Connecticut
| | - Peter B. Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
221
|
Salinas V, Vega P, Piccirilli MV, Chicco C, Ciraolo C, Christiansen S, Consalvo D, Perez-Maturo J, Medina N, González-Morón D, Novaro V, Perrone C, García MDC, Agosta G, Silva W, Kauffman M. Identification of a somatic mutation in the RHEB gene through high depth and ultra-high depth next generation sequencing in a patient with Hemimegalencephaly and drug resistant Epilepsy. Eur J Med Genet 2019; 62:103571. [DOI: 10.1016/j.ejmg.2018.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/18/2018] [Accepted: 11/04/2018] [Indexed: 10/27/2022]
|
222
|
Miller ZA, Spina S, Pakvasa M, Rosenberg L, Watson C, Mandelli ML, Paredes MF, Joie RL, Rabinovici GD, Rosen HJ, Grinberg LT, Huang EJ, Miller BL, Seeley WW, Gorno-Tempini ML. Cortical developmental abnormalities in logopenic variant primary progressive aphasia with dyslexia. Brain Commun 2019; 1:fcz027. [PMID: 32699834 PMCID: PMC7364264 DOI: 10.1093/braincomms/fcz027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/13/2019] [Accepted: 08/22/2019] [Indexed: 01/01/2023] Open
Abstract
An increased prevalence of dyslexia has been observed in individuals diagnosed with primary progressive aphasia, most notably the logopenic variant primary progressive aphasia. The underlying pathology most commonly associated with logopenic variant primary progressive aphasia is Alzheimer's disease. In this clinical case report series, we describe the neuropathological findings of three patients with logopenic variant primary progressive aphasia and developmental dyslexia, each demonstrating a pattern of cerebrocortical microdysgenesis, reminiscent of findings first reported in dyslexic individuals, alongside expected Alzheimer's disease pathology. Neurodevelopmental and most severe Alzheimer's disease pathological changes overlapped within perisylvian brain regions, areas associated with phonological deficits in both logopenic variant primary progressive aphasia and dyslexia. These three cases with pathological findings support the hypothesis that early-life neurodevelopmental changes might influence later-life susceptibility to neurodegenerative disease and could contribute to non-amnestic, early age-of-onset presentations of Alzheimer's disease. Larger studies investigating neurobiological vulnerability across the lifespan are needed.
Collapse
Affiliation(s)
- Zachary A Miller
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Salvatore Spina
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Mikhail Pakvasa
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Lynne Rosenberg
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Christa Watson
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Maria Luisa Mandelli
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Mercedes F Paredes
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Gil D Rabinovici
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA.,Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA
| | - Howard J Rosen
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Lea T Grinberg
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA.,Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA.,Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Maria Luisa Gorno-Tempini
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
223
|
Sapir T, Barakat TS, Paredes MF, Lerman-Sagie T, Aronica E, Klonowski W, Nguyen L, Ben Zeev B, Bahi-Buisson N, Leventer R, Rachmian N, Reiner O. Building Bridges Between the Clinic and the Laboratory: A Meeting Review - Brain Malformations: A Roadmap for Future Research. Front Cell Neurosci 2019; 13:434. [PMID: 31611776 PMCID: PMC6776596 DOI: 10.3389/fncel.2019.00434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/09/2019] [Indexed: 01/08/2023] Open
Abstract
In the middle of March 2019, a group of scientists and clinicians (as well as those who wear both hats) gathered in the green campus of the Weizmann Institute of Science to share recent scientific findings, to establish collaborations, and to discuss future directions for better diagnosis, etiology modeling and treatment of brain malformations. One hundred fifty scientists from twenty-two countries took part in this meeting. Thirty-eight talks were presented and as many as twenty-five posters were displayed. This review is aimed at presenting some of the highlights that the audience was exposed to during the three-day meeting.
Collapse
Affiliation(s)
- Tamar Sapir
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Mercedes F. Paredes
- Department of Neurology and Neuroscience Graduate Division, University of California, San Francisco, San Francisco, CA, United States
| | - Tally Lerman-Sagie
- Pediatric Neurology Unit, Fetal Neurology Clinic, Wolfson Medical Center, Holon and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eleonora Aronica
- Department of (Neuro-)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Zwolle, Netherlands
| | - Wlodzimierz Klonowski
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Laurent Nguyen
- GIGA-Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), C.H.U. Sart Tilman, University of Liège, Liège, Belgium
| | - Bruria Ben Zeev
- Sackler School of Medicine and Pediatric Neurology Unit, Edmond and Lilly Safra Pediatric Hospital, Tel Aviv University, Tel Aviv, Israel
| | - Nadia Bahi-Buisson
- INSERM UMR 1163, Imagine Institute, Paris Descartes University, Paris, France
- Necker Enfants Malades Hospital, Pediatrric Neurology APHP, Paris, France
| | - Richard Leventer
- Department of Neurology, Royal Children’s Hospital, Murdoch Children’s Research Institute, University of Melbourne, Parkville, VIC, Australia
- Department of Pediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Noa Rachmian
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
224
|
Pan YH, Wu N, Yuan XB. Toward a Better Understanding of Neuronal Migration Deficits in Autism Spectrum Disorders. Front Cell Dev Biol 2019; 7:205. [PMID: 31620440 PMCID: PMC6763556 DOI: 10.3389/fcell.2019.00205] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/06/2019] [Indexed: 11/13/2022] Open
Abstract
Newborn neurons in developing brains actively migrate from germinal zones to designated regions before being wired into functional circuits. The motility and trajectory of migrating neurons are regulated by both extracellular factors and intracellular signaling cascades. Defects in the molecular machinery of neuronal migration lead to mis-localization of affected neurons and are considered as an important etiology of multiple developmental disorders including epilepsy, dyslexia, schizophrenia (SCZ), and autism spectrum disorders (ASD). However, the mechanisms that link neuronal migration deficits to the development of these diseases remain elusive. This review focuses on neuronal migration deficits in ASD. From a translational perspective, we discuss (1) whether neuronal migration deficits are general neuropathological characteristics of ASD; (2) how the phenotypic heterogeneity of neuronal migration disorders is generated; (3) how neuronal migration deficits lead to functional defects of brain circuits; and (4) how therapeutic intervention of neuronal migration deficits can be a potential treatment for ASD.
Collapse
Affiliation(s)
- Yi-Hsuan Pan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China
| | - Nan Wu
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China
| | - Xiao-Bing Yuan
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), Institute of Brain Functional Genomics, School of Life Sciences and the Collaborative Innovation Center for Brain Science, East China Normal University, Shanghai, China.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
225
|
Tahmasebi S, Khoutorsky A, Mathews MB, Sonenberg N. Translation deregulation in human disease. Nat Rev Mol Cell Biol 2019; 19:791-807. [PMID: 30038383 DOI: 10.1038/s41580-018-0034-x] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Advances in sequencing and high-throughput techniques have provided an unprecedented opportunity to interrogate human diseases on a genome-wide scale. The list of disease-causing mutations is expanding rapidly, and mutations affecting mRNA translation are no exception. Translation (protein synthesis) is one of the most complex processes in the cell. The orchestrated action of ribosomes, tRNAs and numerous translation factors decodes the information contained in mRNA into a polypeptide chain. The intricate nature of this process renders it susceptible to deregulation at multiple levels. In this Review, we summarize current evidence of translation deregulation in human diseases other than cancer. We discuss translation-related diseases on the basis of the molecular aberration that underpins their pathogenesis (including tRNA dysfunction, ribosomopathies, deregulation of the integrated stress response and deregulation of the mTOR pathway) and describe how deregulation of translation generates the phenotypic variability observed in these disorders.
Collapse
Affiliation(s)
- Soroush Tahmasebi
- Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada. .,Department of Biochemistry, McGill University, Montreal, Quebec, Canada. .,Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA.
| | - Arkady Khoutorsky
- Department of Anesthesia and Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Michael B Mathews
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Nahum Sonenberg
- Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada. .,Department of Biochemistry, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
226
|
The Autophagy-Cilia Axis: An Intricate Relationship. Cells 2019; 8:cells8080905. [PMID: 31443299 PMCID: PMC6721705 DOI: 10.3390/cells8080905] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 01/19/2023] Open
Abstract
Primary cilia are microtubule-based organelles protruding from the surface of almost all vertebrate cells. This organelle represents the cell’s antenna which acts as a communication hub to transfer extracellular signals into intracellular responses during development and in tissue homeostasis. Recently, it has been shown that loss of cilia negatively regulates autophagy, the main catabolic route of the cell, probably utilizing the autophagic machinery localized at the peri-ciliary compartment. On the other side, autophagy influences ciliogenesis in a context-dependent manner, possibly to ensure that the sensing organelle is properly formed in a feedback loop model. In this review we discuss the recent literature and propose that the autophagic machinery and the ciliary proteins are functionally strictly related to control both autophagy and ciliogenesis. Moreover, we report examples of diseases associated with autophagic defects which cause cilia abnormalities, and propose and discuss the hypothesis that, at least some of the clinical manifestations observed in human diseases associated to ciliary disfunction may be the result of a perturbed autophagy.
Collapse
|
227
|
Murugan AK. mTOR: Role in cancer, metastasis and drug resistance. Semin Cancer Biol 2019; 59:92-111. [PMID: 31408724 DOI: 10.1016/j.semcancer.2019.07.003] [Citation(s) in RCA: 310] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/14/2019] [Accepted: 07/03/2019] [Indexed: 02/09/2023]
Abstract
Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that gets inputs from the amino acids, nutrients, growth factor, and environmental cues to regulate varieties of fundamental cellular processes which include protein synthesis, growth, metabolism, aging, regeneration, autophagy, etc. The mTOR is frequently deregulated in human cancer and activating somatic mutations of mTOR were recently identified in several types of human cancer and hence mTOR is therapeutically targeted. mTOR inhibitors were commonly used as immunosuppressors and currently, it is approved for the treatment of human malignancies. This review briefly focuses on the structure and biological functions of mTOR. It extensively discusses the genetic deregulation of mTOR including amplifications and somatic mutations, mTOR-mediated cell growth promoting signaling, therapeutic targeting of mTOR and the mechanisms of resistance, the role of mTOR in precision medicine and other recent advances in further understanding the role of mTOR in cancer.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, PO Box 3354, Research Center (MBC 03), Riyadh, 11211, Saudi Arabia.
| |
Collapse
|
228
|
Assessment of genetic variant burden in epilepsy-associated brain lesions. Eur J Hum Genet 2019; 27:1738-1744. [PMID: 31358956 DOI: 10.1038/s41431-019-0484-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 06/29/2019] [Accepted: 07/05/2019] [Indexed: 01/31/2023] Open
Abstract
It is challenging to estimate genetic variant burden across different subtypes of epilepsy. Herein, we used a comparative approach to assess the genetic variant burden and genotype-phenotype correlations in four most common brain lesions in patients with drug-resistant focal epilepsy. Targeted sequencing analysis was performed for a panel of 161 genes with a mean coverage of >400×. Lesional tissue was histopathologically reviewed and dissected from hippocampal sclerosis (n = 15), ganglioglioma (n = 16), dysembryoplastic neuroepithelial tumors (n = 8), and focal cortical dysplasia type II (n = 15). Peripheral blood (n = 12) or surgical tissue samples histopathologically classified as lesion-free (n = 42) were available for comparison. Variants were classified as pathogenic or likely pathogenic according to American College of Medical Genetics and Genomics guidelines. Overall, we identified pathogenic and likely pathogenic variants in 25.9% of patients with a mean coverage of 383×. The highest number of pathogenic/likely pathogenic variants was observed in patients with ganglioglioma (43.75%; all somatic) and dysembryoplastic neuroepithelial tumors (37.5%; all somatic), and in 20% of cases with focal cortical dysplasia type II (13.33% somatic, 6.67% germline). Pathogenic/likely pathogenic positive genes were disorder specific and BRAF V600E the only recurrent pathogenic variant. This study represents a reference for the genetic variant burden across the four most common lesion entities in patients with drug-resistant focal epilepsy. The observed large variability in variant burden by epileptic lesion type calls for whole exome sequencing of histopathologically well-characterized tissue in a diagnostic setting and in research to discover novel disease-associated genes.
Collapse
|
229
|
Zhao S, Li Z, Zhang M, Zhang L, Zheng H, Ning J, Wang Y, Wang F, Zhang X, Gan H, Wang Y, Zhang X, Luo H, Bu G, Xu H, Yao Y, Zhang YW. A brain somatic RHEB doublet mutation causes focal cortical dysplasia type II. Exp Mol Med 2019; 51:1-11. [PMID: 31337748 PMCID: PMC6802736 DOI: 10.1038/s12276-019-0277-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 03/27/2019] [Accepted: 04/10/2019] [Indexed: 01/16/2023] Open
Abstract
Focal cortical dysplasia type II (FCDII) is a cerebral cortex malformation characterized by local cortical structure disorganization, neuronal dysmorphology, and refractory epilepsy. Brain somatic mutations in several genes involved in the PI3K/AKT/mTOR pathway are associated with FCDII, but they are only found in a proportion of patients with FCDII. The genetic causes underlying the development FCDII in other patients remain unclear. Here, we carried out whole exome sequencing and targeted sequencing in paired brain–blood DNA from patients with FCDII and identified a brain somatic doublet mutation c.(A104T, C105A) in the Ras homolog, mTORC1 binding (RHEB) gene, which led to the RHEB p.Y35L mutation in one patient with FCDII. This RHEB mutation carrier had a dramatic increase of ribosomal protein S6 phosphorylation, indicating mTOR activation in the region of the brain lesion. The RHEB p.Y35L mutant protein had increased GTPλS-binding activity compared with wild-type RHEB. Overexpression of the RHEB p.Y35L variant in cultured cells also resulted in elevated S6 phosphorylation compared to wild-type RHEB. Importantly, in utero electroporation of the RHEB p.Y35L variant in mice induced S6 phosphorylation, cytomegalic neurons, dysregulated neuron migration, abnormal electroencephalogram, and seizures, all of which are found in patients with FCDII. Rapamycin treatment rescued abnormal electroencephalograms and alleviated seizures in these mice. These results demonstrate that brain somatic mutations in RHEB are also responsible for the pathogenesis of FCDII, indicating that aberrant activation of mTOR signaling is a primary driver and potential drug target for FCDII. Identifying a genetic mutation causing a congenital brain disorder that triggers difficult-to-treat epilepsy suggests a potential therapeutic target, according to Chinese scientists. Focal cortical dysplasia type II (FCDII) is one of a group of brain development abnormalities that cause intractable epilepsy. Scientists have identified gene mutations in some FCDII patients linked to a signaling pathway involved in cell proliferation and metabolism in the developing brain. Now, Yun-wu Zhang and Yi Yao at Xiamen University in Fujian, China, and co-workers have discovered a mutation on a gene which triggers abnormal activation of the same signaling pathway. The team found that the drug rapamycin, which inhibits this pathway, alleviated epileptic seizures in mice with the mutant gene. Their findings add weight to the theory that this pathway may be a viable target for future therapies.
Collapse
Affiliation(s)
- Shanshan Zhao
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Zhenghui Li
- Neuromedicine Center, the 174th Hospital of Chinese People's Liberation Army, Affiliated Chenggong Hospital, Xiamen University, Xiamen, 361003, Fujian, China.,Department of Neurosurgery, Kaifeng Central Hospital, Kaifeng, 475000, Henan, China
| | - Muxian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Lingliang Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Honghua Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Jinhuan Ning
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yanyan Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Fengpeng Wang
- Neuromedicine Center, the 174th Hospital of Chinese People's Liberation Army, Affiliated Chenggong Hospital, Xiamen University, Xiamen, 361003, Fujian, China.,XiaMen Humanity Hospital, No.3777 XianYue Road, HuLi District, XiaMen, 361015, FuJian, China
| | - Xiaobin Zhang
- Neuromedicine Center, the 174th Hospital of Chinese People's Liberation Army, Affiliated Chenggong Hospital, Xiamen University, Xiamen, 361003, Fujian, China.,XiaMen Humanity Hospital, No.3777 XianYue Road, HuLi District, XiaMen, 361015, FuJian, China
| | - Hexia Gan
- Neuromedicine Center, the 174th Hospital of Chinese People's Liberation Army, Affiliated Chenggong Hospital, Xiamen University, Xiamen, 361003, Fujian, China
| | - Yuanqing Wang
- Neuromedicine Center, the 174th Hospital of Chinese People's Liberation Army, Affiliated Chenggong Hospital, Xiamen University, Xiamen, 361003, Fujian, China
| | - Xian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Hong Luo
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Huaxi Xu
- Neuroscience Initiative, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Yi Yao
- Neuromedicine Center, the 174th Hospital of Chinese People's Liberation Army, Affiliated Chenggong Hospital, Xiamen University, Xiamen, 361003, Fujian, China. .,XiaMen Humanity Hospital, No.3777 XianYue Road, HuLi District, XiaMen, 361015, FuJian, China. .,Department of Pediatric Neurology, Shenzhen Children's Hospital, Shenzhen, 518026, Guangdong Province, China.
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
| |
Collapse
|
230
|
Abstract
Somatic mutation of the MTOR gene is a genetic etiology of focal malformations of cortical development. In this issue of Neuron, Park et al. (2018) identify defective autophagy-dependent ciliogenesis/Wnt signaling as an underlying mechanism affecting neuronal migration and cortical lamination.
Collapse
Affiliation(s)
- Alessia Di Nardo
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mustafa Sahin
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
231
|
Park JS, Lee J, Jung ES, Kim MH, Kim IB, Son H, Kim S, Kim S, Park YM, Mook-Jung I, Yu SJ, Lee JH. Brain somatic mutations observed in Alzheimer's disease associated with aging and dysregulation of tau phosphorylation. Nat Commun 2019; 10:3090. [PMID: 31300647 PMCID: PMC6626023 DOI: 10.1038/s41467-019-11000-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 06/14/2019] [Indexed: 01/24/2023] Open
Abstract
The role of brain somatic mutations in Alzheimer’s disease (AD) is not well understood. Here, we perform deep whole-exome sequencing (average read depth 584×) in 111 postmortem hippocampal formation and matched blood samples from 52 patients with AD and 11 individuals not affected by AD. The number of somatic single nucleotide variations (SNVs) in AD brain specimens increases significantly with aging, and the rate of mutation accumulation in the brain is 4.8-fold slower than that in AD blood. The putatively pathogenic brain somatic mutations identified in 26.9% (14 of 52) of AD individuals are enriched in PI3K-AKT, MAPK, and AMPK pathway genes known to contribute to hyperphosphorylation of tau. We show that a pathogenic brain somatic mutation in PIN1 leads to a loss-of-function mutation. In vitro mimicking of haploinsufficiency of PIN1 aberrantly increases tau phosphorylation and aggregation. This study provides new insights into the genetic architecture underlying the pathogenesis of AD. The role of brain somatic mutations in neurodegenerative diseases such as Alzheimer’s disease (AD) is not well understood. Here the authors carry out high-depth exome sequencing ~500× on brain tissue from patients with AD and controls, and identify mutations in a number of genes that are known to contribute to phosphorylation and aggregation of tau, including PIN1.
Collapse
Affiliation(s)
- Jun Sung Park
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Junehawk Lee
- Center for Supercomputing Applications, Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Republic of Korea
| | - Eun Sun Jung
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.,Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Myeong-Heui Kim
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Il Bin Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hyeonju Son
- Department of Biomedical Systems Informatics, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Sanghyeon Kim
- Laboratory of Brain Research, Stanley Medical Research Institute (SMRI), 9800 Medical Center Drive, Suite C-050, Rockville, MD, 20850, USA
| | - Young Mok Park
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, 34126, Republic of Korea
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.,Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seok Jong Yu
- Center for Supercomputing Applications, Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Republic of Korea.
| | - Jeong Ho Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea. .,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
232
|
Ye Z, McQuillan L, Poduri A, Green TE, Matsumoto N, Mefford HC, Scheffer IE, Berkovic SF, Hildebrand MS. Somatic mutation: The hidden genetics of brain malformations and focal epilepsies. Epilepsy Res 2019; 155:106161. [PMID: 31295639 DOI: 10.1016/j.eplepsyres.2019.106161] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 01/12/2023]
Abstract
Over the past decade there has been a substantial increase in genetic studies of brain malformations, fueled by the availability of improved technologies to study surgical tissue to address the hypothesis that focal lesions arise from focal, post-zygotic genetic disruptions. Traditional genetic studies of patients with malformations utilized leukocyte-derived DNA to search for germline variants, which are inherited or arise de novo in parental gametes. Recent studies have demonstrated somatic variants that arise post-zygotically also underlie brain malformations, and that somatic mutation explains a larger proportion of focal malformations than previously thought. We now know from studies of non-diseased individuals that somatic variation occurs routinely during cell division, including during early brain development when the rapid proliferation of neuronal precursor cells provides the ideal environment for somatic mutation to occur and somatic variants to accumulate. When confined to brain, pathogenic variants contribute to the "hidden genetics" of neurological diseases. With burgeoning novel high-throughput genetic technologies, somatic genetic variations are increasingly being recognized. Here we discuss accumulating evidence for the presence of somatic variants in normal brain tissue, review our current understanding of somatic variants in brain malformations associated with lesional epilepsy, and provide strategies to identify the potential contribution of somatic mutation to non-lesional epilepsies. We also discuss technologies that may improve detection of somatic variants in the future in these and other neurological conditions.
Collapse
Affiliation(s)
- Zimeng Ye
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia
| | - Lara McQuillan
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia
| | - Annapurna Poduri
- Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Timothy E Green
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Heather C Mefford
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, United States
| | - Ingrid E Scheffer
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia; Department of Pediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia; Department of Neurology, Royal Children's Hospital, Parkville, Victoria, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Samuel F Berkovic
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia
| | - Michael S Hildebrand
- Department of Medicine (Austin Hospital), University of Melbourne, Heidelberg, Victoria, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.
| |
Collapse
|
233
|
Global Analysis of Intercellular Homeodomain Protein Transfer. Cell Rep 2019; 28:712-722.e3. [PMID: 31315049 PMCID: PMC9472292 DOI: 10.1016/j.celrep.2019.06.056] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 05/06/2019] [Accepted: 06/14/2019] [Indexed: 11/20/2022] Open
Abstract
The homeodomain is found in hundreds of transcription factors that play roles in fate determination via cell-autonomous regulation of gene expression. However, some homeodomain-containing proteins (HPs) are thought to be secreted and penetrate neighboring cells to affect the recipient cell fate. To determine whether this is a general characteristic of HPs, we carried out a large-scale validation for intercellular transfer of HPs. Our screening reveals that intercellular transfer is a general feature of HPs, but it occurs in a cell-context-sensitive manner. We also found the secretion is not solely a function of the homeodomain, but it is supported by external motifs containing hydrophobic residues. Thus, mutations of hydrophobic residues of HPs abrogate secretion and consequently interfere with HP function in recipient cells. Collectively, our study proposes that HP transfer is an intercellular communication method that couples the functions of interacting cells. Lee et al. evaluate capabilities of homeodomain proteins (HPs) for transfer between cells. They find that intercellular transfer is a general but cell-context-sensitive property of HP. Intercellular HP transfer can be an unconventional way for the cells to communicate with neighboring cells that associate structurally and functionally.
Collapse
|
234
|
Benova B, Jacques TS. Genotype-phenotype correlations in focal malformations of cortical development: a pathway to integrated pathological diagnosis in epilepsy surgery. Brain Pathol 2019; 29:473-484. [PMID: 30485578 PMCID: PMC8028510 DOI: 10.1111/bpa.12686] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/20/2018] [Indexed: 12/18/2022] Open
Abstract
Malformations of cortical development (MCD) comprise a broad spectrum of developmental brain abnormalities. Patients presenting with MCDs often suffer from drug-resistant focal epilepsy, and some become candidates for epilepsy surgery. Their likelihood of achieving freedom from seizures, however, remains uncertain, and depends in a major part on the underlying pathology. Tissue samples obtained in epilepsy surgery form the basis of definite histopathological diagnosis; however, new molecular genetic methods have not yet been implemented in diagnostic processes for MCD cases. Furthermore, it has not been completely understood how the underlying pathology affects patients' outcomes after epilepsy surgery. We performed a systematic literature review of studies describing both histopathological and molecular genetic findings in MCD, along with studies on epilepsy surgery outcomes. We aimed to correlate the genetic causes with the underlying morphological abnormalities in focal cortical malformations and to stress the importance of the underlying biology for patient management and counseling. From the summarized findings of multiple authors, it is obvious that MCD may have a diverse genetic background despite a similar or even identical histopathological picture. Even though most of their molecular genetic findings converge on various levels of the PI3K/AKT/mTOR pathway, the exact mechanisms underlying MCD formation have not yet been completely described or indeed how this pathway generates a diverse range of histological abnormalities. Based on our findings, we therefore propose that all patients diagnosed and operated for drug-resistant epilepsy should have an integrated molecular and pathological diagnosis similar to the current practice in brain tumor diagnostic processes that might lead to more accurate diagnosis and effective stratification of patients undergoing epilepsy surgery.
Collapse
Affiliation(s)
- Barbora Benova
- 2nd Faculty of Medicine, Department of Paediatric NeurologyCharles University and Motol University HospitalPragueCzech Republic
- 2nd Faculty of MedicineCharles UniversityPragueCzech Republic
- Developmental Biology and Cancer ProgrammeUCL GOS Institute of Child HealthLondonUK
| | - Thomas S. Jacques
- Developmental Biology and Cancer ProgrammeUCL GOS Institute of Child HealthLondonUK
- Department of HistopathologyGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| |
Collapse
|
235
|
Lee WS, Stephenson SEM, Howell KB, Pope K, Gillies G, Wray A, Maixner W, Mandelstam SA, Berkovic SF, Scheffer IE, MacGregor D, Harvey AS, Lockhart PJ, Leventer RJ. Second-hit DEPDC5 mutation is limited to dysmorphic neurons in cortical dysplasia type IIA. Ann Clin Transl Neurol 2019; 6:1338-1344. [PMID: 31353856 PMCID: PMC6649645 DOI: 10.1002/acn3.50815] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 05/22/2019] [Indexed: 12/13/2022] Open
Abstract
Focal cortical dysplasia (FCD) causes drug‐resistant epilepsy and is associated with pathogenic variants in mTOR pathway genes. How germline variants cause these focal lesions is unclear, however a germline + somatic “2‐hit” model is hypothesized. In a boy with drug‐resistant epilepsy, FCD, and a germline DEPDC5 pathogenic variant, we show that a second‐hit DEPDC5 variant is limited to dysmorphic neurons, and the somatic mutation load correlates with both dysmorphic neuron density and the epileptogenic zone. These findings provide new insights into the molecular and cellular correlates of FCD determining drug‐resistant epilepsy and refine conceptualization of the epileptogenic zone.
Collapse
Affiliation(s)
- Wei Shern Lee
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,University of Melbourne, Melbourne, Victoria, Australia
| | - Sarah E M Stephenson
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,University of Melbourne, Melbourne, Victoria, Australia
| | - Katherine B Howell
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,University of Melbourne, Melbourne, Victoria, Australia.,Royal Children's Hospital Department of Neurology, Melbourne, Victoria, Australia.,The Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia
| | - Kate Pope
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Greta Gillies
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Alison Wray
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Royal Children's Hospital Department of Neurosurgery, Melbourne, Victoria, Australia
| | - Wirginia Maixner
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Royal Children's Hospital Department of Neurosurgery, Melbourne, Victoria, Australia
| | - Simone A Mandelstam
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,University of Melbourne, Melbourne, Victoria, Australia.,The Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.,Royal Children's Hospital Department of Medical Imaging, Melbourne, Victoria, Australia
| | - Samuel F Berkovic
- University of Melbourne, Melbourne, Victoria, Australia.,The Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia
| | - Ingrid E Scheffer
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,University of Melbourne, Melbourne, Victoria, Australia.,Royal Children's Hospital Department of Neurology, Melbourne, Victoria, Australia.,The Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia
| | - Duncan MacGregor
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Royal Children's Hospital Department of Anatomical Pathology, Melbourne, Victoria, Australia
| | - Anthony Simon Harvey
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,University of Melbourne, Melbourne, Victoria, Australia.,Royal Children's Hospital Department of Neurology, Melbourne, Victoria, Australia
| | - Paul J Lockhart
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,University of Melbourne, Melbourne, Victoria, Australia
| | - Richard J Leventer
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,University of Melbourne, Melbourne, Victoria, Australia.,Royal Children's Hospital Department of Neurology, Melbourne, Victoria, Australia
| |
Collapse
|
236
|
Ahn JH, Ohk TG, Kim DW, Kim H, Song M, Lee TK, Lee JC, Yang GE, Shin MC, Cho JH, Choi SY, Won MH, Park JH. Fluoro-Jade B histofluorescence staining detects dentate granule cell death after repeated five-minute transient global cerebral ischemia. Metab Brain Dis 2019; 34:951-956. [PMID: 30830598 DOI: 10.1007/s11011-019-00404-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/25/2019] [Indexed: 02/06/2023]
Abstract
Transient global cerebral ischemia (tGCI)-induced neuronal damage is variable according to its duration and degree. There are many studies on the damage or death of pyramidal cells of the hippocampus proper (CA1-3) in rodent models of tGCI. However, studies on the death of granule cells in the hippocampal dentate gyrus (DG) following tGCI have not yet been addressed. In this study, we examined the damage/death of granule cells in the gerbil DG at 5 days after various durations (5, 10, and 15 min) of single tGCI and repeated tGCI (two 5-min tGCI with 1-h interval) using cresyl violet staining, NeuN immunohistochemistry and Fluoro-Jade B (F-J B) histofluorescence staining. Neuronal death was observed only in the polymorphic layer in all single tGCI-operated groups. However, in the repeated tGCI-operated group, massive neuronal death was observed in the granule cell layer as well as in the polymorphic layer by using F-J B histofluorescence staining. In addition, microgliosis in the DG was significantly increased in the repeated tGCI-operated group compared to the 15-min tGCI-operated group. Taken together, these findings indicate that repeated brief tGCI causes granule cell death in the DG which could not occur by a longer duration of single tGCI.
Collapse
Affiliation(s)
- Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, 1 Hallimdaehak-gil, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, and Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, Gangneung, Gangwon, 25457, Republic of Korea
| | - Hyunjung Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Minah Song
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Go Eun Yang
- Department of Radiology, Kangwon National University Hospital, Chuncheon, Gangwon, 24289, Republic of Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, 1 Hallimdaehak-gil, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea.
| | - Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, 1 Hallimdaehak-gil, Chuncheon, Gangwon, 24252, Republic of Korea.
| |
Collapse
|
237
|
FAN M, DONG S, ZOU X, ZHENG B, HUANG Y, WANG J, ZENG L. [Correlation of phosphorylated S6 protein expression in blood and brain tissue in mice and rats with kainic acid-induced seizure]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:303-309. [PMID: 31496163 PMCID: PMC8800752 DOI: 10.3785/j.issn.1008-9292.2019.06.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/15/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To determine the correlation of phosphorylated ribosomal S6 protein (P-S6) content in blood and brain tissue in mice and rats with seizure. METHODS Seizure models were induced by intraperitoric injection of kainic acid (KA) in C57BL/mice and SD rats. Flow cytometry was used to detect the content of P-S6 in blood; Western blot was used to detect the expression of P-S6 in brain tissues. The correlation between P-S6 expression in blood and in brain tissue was examine by Pearson analysis, and the correlation between P-S6 expression in blood and the severity of seizure was also observed. RESULTS Western blotting analysis showed that the expression of P-S6 was significantly increased in peripheral blood and brain tissue in mice 1 h after KA-induced seizure,and the expression levels increased to (1.49±0.45) times (P<0.05) and (2.55±0.66) times (P <0.01) of the control group, respectively. Flow cytometry showed that the positive percentage and average fluorescence intensity of P-S6 in the blood of mice increased significantly 1 h after KA-induced seizures (P<0.01), which was consistent with the expression of P-S6 in brain tissue (r=0.8474, P<0.01). Flow cytometry showed that the average fluorescence intensity of P-S6 in blood increased from 14.89±9.75 to 52.35±21.72 (P<0.01) in rats with seizure, which was consistent with the change of P-S6 in brain tissue (r=0.9385, P<0.01). Rats with higher levels of seizure were of higher levels of P-S6 in peripheral blood. CONCLUSIONS Consistent correlation of P-S6 expression is demonstrated in peripheral blood and in brain tissue after KA-induced seizure, suggesting that the expression of P-S6 in blood can accurately reflect the changes of mTOR signaling pathway in brain tissue.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianda WANG
- 王健达(1988-), 男, 硕士, 住院医师, 主要从事神经内科学研究; E-mail:
;
https://orcid.org/0000-0001-7148-3183
| | - Linghui ZENG
- 曾玲晖(1972-), 女, 博士, 教授, 博士生导师, 主要从事神经药理学研究; E-mail:
;
https://orcid.org/0000-0002-5924-4419
| |
Collapse
|
238
|
Zhang L, Huang T, Teaw S, Bordey A. Hypervascularization in mTOR-dependent focal and global cortical malformations displays differential rapamycin sensitivity. Epilepsia 2019; 60:1255-1265. [PMID: 31125447 DOI: 10.1111/epi.15969] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 01/16/2023]
Abstract
OBJECTIVES Patients with mammalian target of rapamycin (mTOR)-dependent malformations of cortical development (MCDs) associated with seizures display hyperperfusion and increased vessel density of the dysmorphic cortical tissue. Some studies have suggested that the vascular defect occurred independently of seizures. Here, we further examined whether hypervascularization occurs in animal models of global and focal MCD with and without seizures, and whether it is sensitive to the mTOR blocker, rapamycin, that is approved for epilepsy treatment in tuberous sclerosis complex. METHODS We used two experimental models of mTOR-dependent MCD consisting of conditional transgenic mice containing Tsc1null cells in the forebrain generating a global malformation associated with seizures and of wild-type mice containing a focal malformation in the somatosensory cortex generated by in utero electroporation (IUE) that does not lead to seizures. Alterations in blood vessels and the effects of a 2-week-long rapamycin treatment on these phenotypes were assessed in juvenile mice. RESULTS Blood vessels in both the focal and global MCDs of postnatal day 14 mice displayed significant increase in vessel density, branching index, total vessel length, and decreased tissue lacunarity. In addition, rapamycin treatment (0.5 mg/kg, every 2 days) partially rescued vessel abnormalities in the focal MCD model, but it did not ameliorate the vessel abnormalities in the global MCD model that required higher rapamycin dosage for a partial rescue. SIGNIFICANCE Here, we identified hypervascularization in mTOR-dependent MCD in the absence of seizures in young mice, suggesting that increased angiogenesis occurs during development in parallel to alterations in corticogenesis. In addition, a predictive functional outcome is that dysplastic neurons forming MCD will have better access to oxygen and metabolic supplies via their closer proximity to blood vessels. Finally, the difference in rapamycin sensitivity between a focal and global MCD suggest that rapamycin treatment will need to be titrated to match the type of MCD.
Collapse
Affiliation(s)
- Longbo Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Departments of Neurosurgery and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| | - Tianxiang Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Departments of Neurosurgery and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| | - Shannon Teaw
- Departments of Neurosurgery and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| | - Angélique Bordey
- Departments of Neurosurgery and Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
239
|
Blair JD, Bateup HS. New frontiers in modeling tuberous sclerosis with human stem cell-derived neurons and brain organoids. Dev Dyn 2019; 249:46-55. [PMID: 31070828 DOI: 10.1002/dvdy.60] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/16/2022] Open
Abstract
Recent advances in human stem cell and genome engineering have enabled the generation of genetically defined human cellular models for brain disorders. These models can be established from a patient's own cells and can be genetically engineered to generate isogenic, controlled systems for mechanistic studies. Given the challenges of obtaining and working with primary human brain tissue, these models fill a critical gap in our understanding of normal and abnormal human brain development and provide an important complement to animal models. Recently, there has been major progress in modeling the neuropathophysiology of the canonical "mTORopathy" tuberous sclerosis complex (TSC) with such approaches. Studies using two- and three-dimensional cultures of human neurons and glia have provided new insights into how mutations in the TSC1 and TSC2 genes impact human neural development and function. Here we discuss recent progress in human stem cell-based modeling of TSC and highlight challenges and opportunities for further efforts in this area.
Collapse
Affiliation(s)
- John D Blair
- Department of Molecular and Cell Biology, University of California, Berkeley, California
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,Helen Wills Neuroscience Institute, University of California, Berkeley, California.,Chan Zuckerberg Biohub, San Francisco, California
| |
Collapse
|
240
|
Lee KH, Lee YJ, Seo JH, Baumgartner JE, Westerveld M. Epilepsy Surgery in Children versus Adults. J Korean Neurosurg Soc 2019; 62:328-335. [PMID: 31085959 PMCID: PMC6514317 DOI: 10.3340/jkns.2019.0026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 02/24/2019] [Indexed: 11/29/2022] Open
Abstract
Epilepsy is one of the most common chronic neurological disorder affecting 6–7 per 1000 worldwide. Nearly one-third of patients with newly diagnosed epilepsy continue to have recurrent seizures despite adequate trial of more than two anti-seizure drugs : drug-resistant epilepsy (DRE). Children with DRE often experience cognitive and psychosocial co-morbidities requiring more urgent and aggressive treatment than adults. Epilepsy surgery can result in seizure-freedom in approximately two-third of children with improvement in cognitive development and quality of life. Understanding fundamental differences in etiology, co-morbidity, and neural plasticity between children and adults is critical for appropriate selection of surgical candidates, appropriate presurgical evaluation and surgical approach, and improved overall outcome.
Collapse
Affiliation(s)
- Ki Hyeong Lee
- Comprehensive Epilepsy Center, Advent Health for Children, Orlando, FL, USA
| | - Yun-Jin Lee
- Comprehensive Epilepsy Center, Advent Health for Children, Orlando, FL, USA.,Department of Pediatrics, Pusan University College of Medicine, Yangsan, Korea
| | - Joo Hee Seo
- Comprehensive Epilepsy Center, Advent Health for Children, Orlando, FL, USA
| | | | - Michael Westerveld
- Comprehensive Epilepsy Center, Advent Health for Children, Orlando, FL, USA
| |
Collapse
|
241
|
Phi JH, Cho BK. Epilepsy Surgery in 2019 : A Time to Change. J Korean Neurosurg Soc 2019; 62:361-365. [PMID: 31085963 PMCID: PMC6514313 DOI: 10.3340/jkns.2019.0078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023] Open
Abstract
Epilepsy has been known to humankind since antiquity. The surgical treatment of epilepsy began in the early days of neurosurgery and has developed greatly. Many surgical procedures have stood the test of time. However, clinicians treating epilepsy patients are now witnessing a huge tide of change. In 2017, the classification system for seizure and epilepsy types was revised nearly 36 years after the previous scheme was released. The actual difference between these systems may not be large, but there have been many conceptual changes, and clinicians must bid farewell to old terminology. Paradigms in drug discovery are changing, and novel antiseizure drugs have been introduced for clinical use. In particular, drugs that target genetic changes harbor greater therapeutic potential than previous screening-based compounds. The concept of focal epilepsy has been challenged, and now epilepsy is regarded as a network disorder. With this novel concept, stereotactic electroencephalography (SEEG) is becoming increasingly popular for the evaluation of dysfunctioning neuronal networks. Minimally invasive ablative therapies using SEEG electrodes and neuromodulatory therapies such as deep brain stimulation and vagus nerve stimulation are widely applied to remedy dysfunctional epilepsy networks. The use of responsive neurostimulation is currently off-label in children with intractable epilepsy.
Collapse
Affiliation(s)
- Ji Hoon Phi
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Korea
| | - Byung-Kyu Cho
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Korea.,Department of Neurosurgery, Armed Forces Capital Hospital, Seongnam, Korea
| |
Collapse
|
242
|
Kobow K, Ziemann M, Kaipananickal H, Khurana I, Mühlebner A, Feucht M, Hainfellner JA, Czech T, Aronica E, Pieper T, Holthausen H, Kudernatsch M, Hamer H, Kasper BS, Rössler K, Conti V, Guerrini R, Coras R, Blümcke I, El-Osta A, Kaspi A. Genomic DNA methylation distinguishes subtypes of human focal cortical dysplasia. Epilepsia 2019; 60:1091-1103. [PMID: 31074842 PMCID: PMC6635741 DOI: 10.1111/epi.14934] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/26/2022]
Abstract
Objectives Focal cortical dysplasia (FCD) is a major cause of drug‐resistant focal epilepsy in children, and the clinicopathological classification remains a challenging issue in daily practice. With the recent progress in DNA methylation–based classification of human brain tumors we examined whether genomic DNA methylation and gene expression analysis can be used to also distinguish human FCD subtypes. Methods DNA methylomes and transcriptomes were generated from massive parallel sequencing in 15 surgical FCD specimens, matched with 5 epilepsy and 6 nonepilepsy controls. Results Differential hierarchical cluster analysis of DNA methylation distinguished major FCD subtypes (ie, Ia, IIa, and IIb) from patients with temporal lobe epilepsy patients and nonepileptic controls. Targeted panel sequencing identified a novel likely pathogenic variant in DEPDC5 in a patient with FCD type IIa. However, no enrichment of differential DNA methylation or gene expression was observed in mechanistic target of rapamycin (mTOR) pathway–related genes. Significance Our studies extend the evidence for disease‐specific methylation signatures toward focal epilepsies in favor of an integrated clinicopathologic and molecular classification system of FCD subtypes incorporating genomic methylation.
Collapse
Affiliation(s)
- Katja Kobow
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Mark Ziemann
- Epigenetics in Human Health and Disease, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Harikrishnan Kaipananickal
- Epigenetics in Human Health and Disease, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ishant Khurana
- Epigenetics in Human Health and Disease, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Angelika Mühlebner
- Department of Pediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria.,Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Martha Feucht
- Department of Pediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria
| | | | - Thomas Czech
- Department of Neurosurgery, Medical University Vienna, Vienna, Austria
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Zwolle, The Netherlands
| | - Tom Pieper
- Department of Neuropaediatrics and Neurological Rehabilitation, Epilepsy Centre for Children and Adolescents, Schoen Clinic Vogtareuth, Vogtareuth, Germany
| | - Hans Holthausen
- Department of Neuropaediatrics and Neurological Rehabilitation, Epilepsy Centre for Children and Adolescents, Schoen Clinic Vogtareuth, Vogtareuth, Germany
| | - Manfred Kudernatsch
- Department of Neurosurgery and Epilepsy Surgery, Schoen Clinic Vogtareuth, Vogtareuth, Germany
| | - Hajo Hamer
- Department of Neurology, Erlangen Epilepsy Center, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Burkhard S Kasper
- Department of Neurology, Erlangen Epilepsy Center, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Karl Rössler
- Department of Neurosurgery, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Valerio Conti
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Roland Coras
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ingmar Blümcke
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Assam El-Osta
- Epigenetics in Human Health and Disease, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia.,Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong City, Hong Kong SAR
| | - Antony Kaspi
- Epigenetics in Human Health and Disease, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
243
|
Yang J, Kim SK, Kim KJ, Chae JH, Lim BC, Wang KC, Park SH, Phi JH. Satellite lesions of DNET: implications for seizure and tumor control after resection. J Neurooncol 2019; 143:437-445. [DOI: 10.1007/s11060-019-03174-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/25/2019] [Indexed: 01/09/2023]
|
244
|
Phi JH, Kim SK. Clinical Pearls and Advances in Molecular Researches of Epilepsy-Associated Tumors. J Korean Neurosurg Soc 2019; 62:313-320. [PMID: 31085957 PMCID: PMC6514318 DOI: 10.3340/jkns.2019.0033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/07/2019] [Accepted: 03/17/2019] [Indexed: 11/30/2022] Open
Abstract
Brain tumors are the second most common type of structural brain lesion that causes chronic epilepsy. Patients with low-grade brain tumors often experience chronic drug-resistant epilepsy starting in childhood, which led to the concept of long-term epilepsy-associated tumors (LEATs). Dysembryoplastic neuroepithelial tumor and ganglioglioma are representative LEATs and are characterized by young age of onset, frequent temporal lobe location, benign tumor biology, and chronic epilepsy. Although highly relevant in clinical epileptology, the concept of LEATs has been criticized in the neuro-oncology field. Recent genomic and molecular studies have challenged traditional views on LEATs and low-grade gliomas. Molecular studies have revealed that lowgrade gliomas can largely be divided into three groups : LEATs, pediatric-type diffuse low-grade glioma (DLGG; astrocytoma and oligodendroglioma), and adult-type DLGG. There is substantial overlap between conventional LEATs and pediatric-type DLGG in regard to clinical features, histology, and molecular characteristics. LEATs and pediatric-type DLGG are characterized by mutations in BRAF, FGFR1, and MYB/MYBL1, which converge on the RAS-RAF-MAPK pathway. Gene (mutation)-centered classification of epilepsyassociated tumors could provide new insight into these heterogeneous and diverse neoplasms and may lead to novel molecular targeted therapies for epilepsy in the near future.
Collapse
Affiliation(s)
- Ji Hoon Phi
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Korea
| | - Seung-Ki Kim
- Division of Pediatric Neurosurgery, Seoul National University Children's Hospital, Seoul, Korea
| |
Collapse
|
245
|
Kim JK, Lee JH. Mechanistic Target of Rapamycin Pathway in Epileptic Disorders. J Korean Neurosurg Soc 2019; 62:272-287. [PMID: 31085953 PMCID: PMC6514310 DOI: 10.3340/jkns.2019.0027] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/12/2019] [Indexed: 12/19/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) pathway coordinates the metabolic activity of eukaryotic cells through environmental signals, including nutrients, energy, growth factors, and oxygen. In the nervous system, the mTOR pathway regulates fundamental biological processes associated with neural development and neurodegeneration. Intriguingly, genes that constitute the mTOR pathway have been found to be germline and somatic mutation from patients with various epileptic disorders. Hyperactivation of the mTOR pathway due to said mutations has garnered increasing attention as culprits of these conditions : somatic mutations, in particular, in epileptic foci have recently been identified as a major genetic cause of intractable focal epilepsy, such as focal cortical dysplasia. Meanwhile, epilepsy models with aberrant activation of the mTOR pathway have helped elucidate the role of the mTOR pathway in epileptogenesis, and evidence from epilepsy models of human mutations recapitulating the features of epileptic patients has indicated that mTOR inhibitors may be of use in treating epilepsy associated with mutations in mTOR pathway genes. Here, we review recent advances in the molecular and genetic understanding of mTOR signaling in epileptic disorders. In particular, we focus on the development of and limitations to therapies targeting the mTOR pathway to treat epileptic seizures. We also discuss future perspectives on mTOR inhibition therapies and special diagnostic methods for intractable epilepsies caused by brain somatic mutations.
Collapse
Affiliation(s)
- Jang Keun Kim
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Jeong Ho Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| |
Collapse
|
246
|
Nguyen LH, Mahadeo T, Bordey A. mTOR Hyperactivity Levels Influence the Severity of Epilepsy and Associated Neuropathology in an Experimental Model of Tuberous Sclerosis Complex and Focal Cortical Dysplasia. J Neurosci 2019; 39:2762-2773. [PMID: 30700531 PMCID: PMC6445990 DOI: 10.1523/jneurosci.2260-18.2019] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 01/28/2023] Open
Abstract
Tuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) are focal malformations of cortical development (FMCDs) that are highly associated with intractable epilepsy. TSC and FCD are mTORopathies caused by a spectrum of pathogenic variants in the mechanistic target of rapamycin (mTOR) pathway genes leading to differential activation of mTOR signaling. However, whether the degree of mTOR hyperactivity influences disease severity remains unclear. Here, we examined the effects of differential mTOR hyperactivity levels on epilepsy and associated neuropathology in a mouse model of TSC and FCD. Constitutively active Rheb (RhebCA), the canonical activator of mTOR complex 1 (mTORC1), was expressed in mouse embryos of either sex via in utero electroporation at low, intermediate, and high concentrations to induce different mTORC1 activity levels in developing cortical neurons. We found that RhebCA expression induced mTORC1 hyperactivation and increased neuronal soma size and misplacement in a dose-dependent manner. No seizures were detected in the low RhebCA mice, whereas the intermediate and high RhebCA mice displayed spontaneous, recurrent seizures that significantly increased with higher RhebCA concentrations. Seizures were associated with a global increase in microglial activation that was notably higher in the regions containing RhebCA-expressing neurons. These data demonstrate that neuronal mTOR hyperactivity levels influence the severity of epilepsy and associated neuropathology in experimental TSC and FCD. Overall, these findings highlight the importance of evaluating the outcome of individual variants on mTOR activity levels and support personalized medicine strategies based on patient variants and mTOR activity level for TSC, FCD, and potentially other mTORopathies.SIGNIFICANCE STATEMENT Tuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) are epileptogenic cortical malformations caused by pathogenic variants in mechanistic target of rapamycin (mTOR) pathway genes leading to differential mTOR hyperactivation. Here, we present novel findings that neuronal mTOR hyperactivity levels correlate with the severity of epilepsy and associated neuropathology in a mouse model of TSC and FCD. Our findings suggest the need to evaluate the outcome of individual variants on mTOR activity levels in clinical assessments and support personalized medicine strategies based on patient variants and mTOR activity level. Additionally, we present useful modifications to a previously described mouse model of TSC and FCD that allows for titration of seizure frequency and generation of a mild to severe epilepsy phenotype as applicable for preclinical drug testing and mechanistic studies.
Collapse
Affiliation(s)
| | | | - Angélique Bordey
- Department of Neurosurgery, and
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
247
|
Guarnieri FC, de Chevigny A, Falace A, Cardoso C. Disorders of neurogenesis and cortical development. DIALOGUES IN CLINICAL NEUROSCIENCE 2019. [PMID: 30936766 PMCID: PMC6436956 DOI: 10.31887/dcns.2018.20.4/ccardoso] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The development of the cerebral cortex requires complex sequential processes that have to be precisely orchestrated. The localization and timing of neuronal progenitor proliferation and of neuronal migration define the identity, laminar positioning, and specific connectivity of each single cortical neuron. Alterations at any step of this organized series of events—due to genetic mutations or environmental factors—lead to defined brain pathologies collectively known as malformations of cortical development (MCDs), which are now recognized as a leading cause of drug-resistant epilepsy and intellectual disability. In this heterogeneous group of disorders, macroscopic alterations of brain structure (eg, heterotopic nodules, small or absent gyri, double cortex) can be recognized and probably subtend a general reorganization of neuronal circuits. In this review, we provide an overview of the molecular mechanisms that are implicated in the generation of genetic MCDs associated with aberrations at various steps of neurogenesis and cortical development.
Collapse
Affiliation(s)
| | | | - Antonio Falace
- Aix-Marseille University, INSERM U1249, INMED, Marseille 13009, France
| | - Carlos Cardoso
- Aix-Marseille University, INSERM U1249, INMED, Marseille 13009, France
| |
Collapse
|
248
|
Wei W, Keogh MJ, Aryaman J, Golder Z, Kullar PJ, Wilson I, Talbot K, Turner MR, McKenzie CA, Troakes C, Attems J, Smith C, Sarraj SA, Morris CM, Ansorge O, Jones NS, Ironside JW, Chinnery PF. Frequency and signature of somatic variants in 1461 human brain exomes. Genet Med 2019; 21:904-912. [PMID: 30214067 PMCID: PMC6544539 DOI: 10.1038/s41436-018-0274-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/06/2018] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To systematically study somatic variants arising during development in the human brain across a spectrum of neurodegenerative disorders. METHODS In this study we developed a pipeline to identify somatic variants from exome sequencing data in 1461 diseased and control human brains. Eighty-eight percent of the DNA samples were extracted from the cerebellum. Identified somatic variants were validated by targeted amplicon sequencing and/or PyroMark® Q24. RESULTS We observed somatic coding variants present in >10% of sampled cells in at least 1% of brains. The mutational signature of the detected variants showed a predominance of C>T variants most consistent with arising from DNA mismatch repair, occurred frequently in genes that are highly expressed within the central nervous system, and with a minimum somatic mutation rate of 4.25 × 10-10 per base pair per individual. CONCLUSION These findings provide proof-of-principle that deleterious somatic variants can affect sizeable brain regions in at least 1% of the population, and thus have the potential to contribute to the pathogenesis of common neurodegenerative diseases.
Collapse
Affiliation(s)
- Wei Wei
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Michael J Keogh
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Juvid Aryaman
- Department of Mathematics, Imperial College London, London, UK
| | - Zoe Golder
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Peter J Kullar
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Ian Wilson
- Institute of Genetic Medicine, Central Parkway, Newcastle University, Newcastle Upon Tyne, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Chris-Anne McKenzie
- National CJD Research & Surveillance Unit, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Claire Troakes
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Johannes Attems
- Institute of Neuroscience, Newcastle University, Campus for Aging and Vitality, Newcastle upon Tyne, UK
| | - Colin Smith
- National CJD Research & Surveillance Unit, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Safa Al Sarraj
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Chris M Morris
- Institute of Neuroscience, Newcastle University, Campus for Aging and Vitality, Newcastle upon Tyne, UK
| | - Olaf Ansorge
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Nick S Jones
- Department of Mathematics, Imperial College London, London, UK
| | - James W Ironside
- National CJD Research & Surveillance Unit, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Patrick F Chinnery
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
249
|
Ribierre T, Baulac S. Le mosaïcisme somatique en cause dans les épilepsies neurodéveloppementales. Med Sci (Paris) 2019; 35:289-291. [DOI: 10.1051/medsci/2019058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
250
|
Represa A. Why Malformations of Cortical Development Cause Epilepsy. Front Neurosci 2019; 13:250. [PMID: 30983952 PMCID: PMC6450262 DOI: 10.3389/fnins.2019.00250] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/04/2019] [Indexed: 12/13/2022] Open
Abstract
Malformations of cortical development (MCDs), a complex family of rare disorders, result from alterations of one or combined developmental steps, including progenitors proliferation, neuronal migration and differentiation. They are an important cause of childhood epilepsy and frequently associate cognitive deficits and behavioral alterations. Though the physiopathological mechanisms of epilepsy in MCD patients remain poorly elucidated, research during the past decade highlighted the contribution of some factors that will be reviewed in this paper and that include: (i) the genes that caused the malformation, that can be responsible for a significant reduction of inhibitory cells (e.g., ARX gene) or be inducing cell-autonomous epileptogenic changes in affected neurons (e.g., mutations on the mTOR pathway); (ii) the alteration of cortical networks development induced by the malformation that will also involve adjacent or distal cortical areas apparently sane so that the epileptogenic focus might be more extended that the malformation or even localized at distance from it; (iii) the normal developmental processes that would influence and determine the onset of epilepsy in MCD patients, particularly precocious in most of the cases.
Collapse
Affiliation(s)
- Alfonso Represa
- INSERM, Institut de Neurobiologie de la Méditerranée, Aix-Marseille University, Marseille, France
| |
Collapse
|