1
|
Liu AC, Shen Y, Serbinski CR, He H, Roman D, Endale M, Aschbacher-Smith L, King KA, Granadillo JL, López I, Krueger DA, Dye TJ, Smith DF, Hogenesch JB, Prada CE. Clinical and functional studies of MTOR variants in Smith-Kingsmore syndrome reveal deficits of circadian rhythm and sleep-wake behavior. HGG ADVANCES 2024; 5:100333. [PMID: 39030910 PMCID: PMC11342114 DOI: 10.1016/j.xhgg.2024.100333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 07/22/2024] Open
Abstract
Heterozygous de novo or inherited gain-of-function mutations in the MTOR gene cause Smith-Kingsmore syndrome (SKS). SKS is a rare autosomal dominant condition, and individuals with SKS display macrocephaly/megalencephaly, developmental delay, intellectual disability, and seizures. A few dozen individuals are reported in the literature. Here, we report a cohort of 28 individuals with SKS that represent nine MTOR pathogenic variants. We conducted a detailed natural history study and found pathophysiological deficits among individuals with SKS in addition to the common neurodevelopmental symptoms. These symptoms include sleep-wake disturbance, hyperphagia, and hyperactivity, indicative of homeostatic imbalance. To characterize these variants, we developed cell models and characterized their functional consequences. We showed that these SKS variants display a range of mechanistic target of rapamycin (mTOR) activities and respond to the mTOR inhibitor, rapamycin, differently. For example, the R1480_C1483del variant we identified here and the previously known C1483F are more active than wild-type controls and less responsive to rapamycin. Further, we showed that SKS mutations dampened circadian rhythms and low-dose rapamycin improved the rhythm amplitude, suggesting that optimal mTOR activity is required for normal circadian function. As SKS is caused by gain-of-function mutations in MTOR, rapamycin was used to treat several patients. While higher doses of rapamycin caused delayed sleep-wake phase disorder in a subset of patients, optimized lower doses improved sleep. Our study expands the clinical and molecular spectrum of SKS and supports further studies for mechanism-guided treatment options to improve sleep-wake behavior and overall health.
Collapse
Affiliation(s)
- Andrew C Liu
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| | - Yang Shen
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Carolyn R Serbinski
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Hongzhi He
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Destino Roman
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Mehari Endale
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Lindsey Aschbacher-Smith
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Katherine A King
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jorge L Granadillo
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Isabel López
- Pediatric Neurology Unit, Department of Neurology, Clínica Las Condes, Santiago, Chile
| | - Darcy A Krueger
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Thomas J Dye
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - David F Smith
- Divisions of Pediatric Otolaryngology and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; The Sleep Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; The Center for Circadian Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Otolaryngology Head and Neck Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - John B Hogenesch
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Carlos E Prada
- Divisions of Human Genetics, Neurology, Immunobiology, Pediatric Otolaryngology, and Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Genetics, Genomics & Metabolism, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Feinberg School of Medicine of Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
2
|
Ma Q, Chen G, Li Y, Guo Z, Zhang X. The molecular genetics of PI3K/PTEN/AKT/mTOR pathway in the malformations of cortical development. Genes Dis 2024; 11:101021. [PMID: 39006182 PMCID: PMC11245990 DOI: 10.1016/j.gendis.2023.04.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/07/2023] [Accepted: 04/30/2023] [Indexed: 07/16/2024] Open
Abstract
Malformations of cortical development (MCD) are a group of developmental disorders characterized by abnormal cortical structures caused by genetic or harmful environmental factors. Many kinds of MCD are caused by genetic variation. MCD is the common cause of intellectual disability and intractable epilepsy. With rapid advances in imaging and sequencing technologies, the diagnostic rate of MCD has been increasing, and many potential genes causing MCD have been successively identified. However, the high genetic heterogeneity of MCD makes it challenging to understand the molecular pathogenesis of MCD and to identify effective targeted drugs. Thus, in this review, we outline important events of cortical development. Then we illustrate the progress of molecular genetic studies about MCD focusing on the PI3K/PTEN/AKT/mTOR pathway. Finally, we briefly discuss the diagnostic methods, disease models, and therapeutic strategies for MCD. The information will facilitate further research on MCD. Understanding the role of the PI3K/PTEN/AKT/mTOR pathway in MCD could lead to a novel strategy for treating MCD-related diseases.
Collapse
Affiliation(s)
- Qing Ma
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Guang Chen
- Department of Urology, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Ying Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang 150000, China
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Zhenming Guo
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Xue Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang 150000, China
| |
Collapse
|
3
|
Pineau L, Buhler E, Tarhini S, Bauer S, Crepel V, Watrin F, Cardoso C, Represa A, Szepetowski P, Burnashev N. Pathogenic MTOR somatic variant causing focal cortical dysplasia drives hyperexcitability via overactivation of neuronal GluN2C N-methyl-D-aspartate receptors. Epilepsia 2024; 65:2111-2126. [PMID: 38717560 DOI: 10.1111/epi.18000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 07/17/2024]
Abstract
OBJECTIVE Genetic variations in proteins of the mechanistic target of rapamycin (mTOR) pathway cause a spectrum of neurodevelopmental disorders often associated with brain malformations and with intractable epilepsy. The mTORopathies are characterized by hyperactive mTOR pathway and comprise tuberous sclerosis complex (TSC) and focal cortical dysplasia (FCD) type II. How hyperactive mTOR translates into abnormal neuronal activity and hypersynchronous network remains to be better understood. Previously, the role of upregulated GluN2C-containing glutamate-gated N-methyl-D-aspartate receptors (NMDARs) has been demonstrated for germline defects in the TSC genes. Here, we questioned whether this mechanism would expand to other mTORopathies in the different context of a somatic genetic variation of the MTOR protein recurrently found in FCD type II. METHODS We used a rat model of FCD created by in utero electroporation of neural progenitors of dorsal telencephalon with expression vectors encoding either the wild-type or the pathogenic MTOR variant (p.S2215F). In this mosaic configuration, patch-clamp whole-cell recordings of the electroporated, spiny stellate neurons and extracellular recordings of the electroporated areas were performed in neocortical slices. Selective inhibitors were used to target mTOR activity and GluN2C-mediated currents. RESULTS Neurons expressing the mutant protein displayed an excessive activation of GluN2C NMDAR-mediated spontaneous excitatory postsynaptic currents. GluN2C-dependent increase in spontaneous spiking activity was detected in the area of electroporated neurons in the mutant condition and was restricted to a critical time window between postnatal days P9 and P20. SIGNIFICANCE Somatic MTOR pathogenic variant recurrently found in FCD type II resulted in overactivation of GluN2C-mediated neuronal NMDARs in neocortices of rat pups. The related and time-restricted local hyperexcitability was sensitive to subunit GluN2C-specific blockade. Our study suggests that GluN2C-related pathomechanisms might be shared in common by mTOR-related brain disorders.
Collapse
Affiliation(s)
- Louison Pineau
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille University, Marseille, France
| | - Emmanuelle Buhler
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille University, Marseille, France
| | - Sarah Tarhini
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille University, Marseille, France
| | - Sylvian Bauer
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille University, Marseille, France
| | - Valérie Crepel
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille University, Marseille, France
| | - Françoise Watrin
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille University, Marseille, France
| | - Carlos Cardoso
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille University, Marseille, France
| | - Alfonso Represa
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille University, Marseille, France
| | - Pierre Szepetowski
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille University, Marseille, France
| | - Nail Burnashev
- Institut de Neurobiologie de la Méditerranée, Institut National de la Santé et de la Recherche Médicale, Aix-Marseille University, Marseille, France
| |
Collapse
|
4
|
Carton RJ, Doyle MG, Kearney H, Steward CA, Lench NJ, Rogers A, Heinzen EL, McDonald S, Fay J, Lacey A, Beausang A, Cryan J, Brett F, El-Naggar H, Widdess-Walsh P, Costello D, Kilbride R, Doherty CP, Sweeney KJ, O'Brien DF, Henshall DC, Delanty N, Cavalleri GL, Benson KA. Somatic variants as a cause of drug-resistant epilepsy including mesial temporal lobe epilepsy with hippocampal sclerosis. Epilepsia 2024; 65:1451-1461. [PMID: 38491957 DOI: 10.1111/epi.17943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/18/2024]
Abstract
OBJECTIVE The contribution of somatic variants to epilepsy has recently been demonstrated, particularly in the etiology of malformations of cortical development. The aim of this study was to determine the diagnostic yield of somatic variants in genes that have been previously associated with a somatic or germline epilepsy model, ascertained from resected brain tissue from patients with multidrug-resistant focal epilepsy. METHODS Forty-two patients were recruited across three categories: (1) malformations of cortical development, (2) mesial temporal lobe epilepsy with hippocampal sclerosis, and (3) nonlesional focal epilepsy. Participants were subdivided based on histopathology of the resected brain. Paired blood- and brain-derived DNA samples were sequenced using high-coverage targeted next generation sequencing to high depth (585× and 1360×, respectively). Variants were identified using Genome Analysis ToolKit (GATK4) MuTect-2 and confirmed using high-coverage Amplicon-EZ sequencing. RESULTS Sequence data on 41 patients passed quality control. Four somatic variants were validated following amplicon sequencing: within CBL, ALG13, MTOR, and FLNA. The diagnostic yield across 41 patients was 10%, 9% in mesial temporal lobe epilepsy with hippocampal sclerosis and 20% in malformations of cortical development. SIGNIFICANCE This study provides novel insights into the etiology of mesial temporal lobe epilepsy with hippocampal sclerosis, highlighting a potential pathogenic role of somatic variants in CBL and ALG13. We also report candidate diagnostic somatic variants in FLNA in focal cortical dysplasia, while providing further insight into the importance of MTOR and related genes in focal cortical dysplasia. This work demonstrates the potential molecular diagnostic value of variants in both germline and somatic epilepsy genes.
Collapse
Affiliation(s)
- Robert J Carton
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Michael G Doyle
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
- Strategic Academic Recruitment Doctor of Medicine Programme, Royal College of Surgeons in Ireland in collaboration with Blackrock Clinic, Dublin, Ireland
| | - Hugh Kearney
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | | | | | - Anthony Rogers
- Congenica Limited, BioData Innovation Centre, Cambridge, UK
| | - Erin L Heinzen
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Seamus McDonald
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Joanna Fay
- Royal College of Surgeons in Ireland Biobanking Service, Dublin, Ireland
| | - Austin Lacey
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alan Beausang
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Jane Cryan
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Francesca Brett
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Hany El-Naggar
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Peter Widdess-Walsh
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Daniel Costello
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Neurology, Cork University Hospital, Cork, Ireland
| | - Ronan Kilbride
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Colin P Doherty
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Neurology, St. James's Hospital, Dublin, Ireland
| | - Kieron J Sweeney
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Donncha F O'Brien
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - David C Henshall
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Norman Delanty
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Epilepsy Programme, Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Gianpiero L Cavalleri
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Katherine A Benson
- FutureNeuro Science Foundation Ireland Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
5
|
Man A, Di Scipio M, Grewal S, Suk Y, Trinari E, Ejaz R, Whitney R. The Genetics of Tuberous Sclerosis Complex and Related mTORopathies: Current Understanding and Future Directions. Genes (Basel) 2024; 15:332. [PMID: 38540392 PMCID: PMC10970281 DOI: 10.3390/genes15030332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/02/2024] [Accepted: 03/02/2024] [Indexed: 06/14/2024] Open
Abstract
The mechanistic target of rapamycin (mTOR) pathway serves as a master regulator of cell growth, proliferation, and survival. Upregulation of the mTOR pathway has been shown to cause malformations of cortical development, medically refractory epilepsies, and neurodevelopmental disorders, collectively described as mTORopathies. Tuberous sclerosis complex (TSC) serves as the prototypical mTORopathy. Characterized by the development of benign tumors in multiple organs, pathogenic variants in TSC1 or TSC2 disrupt the TSC protein complex, a negative regulator of the mTOR pathway. Variants in critical domains of the TSC complex, especially in the catalytic TSC2 subunit, correlate with increased disease severity. Variants in less crucial exons and non-coding regions, as well as those undetectable with conventional testing, may lead to milder phenotypes. Despite the assumption of complete penetrance, expressivity varies within families, and certain variants delay disease onset with milder neurological effects. Understanding these genotype-phenotype correlations is crucial for effective clinical management. Notably, 15% of patients have no mutation identified by conventional genetic testing, with the majority of cases postulated to be caused by somatic TSC1/TSC2 variants which present complex diagnostic challenges. Advancements in genetic testing, prenatal screening, and precision medicine hold promise for changing the diagnostic and treatment paradigm for TSC and related mTORopathies. Herein, we explore the genetic and molecular mechanisms of TSC and other mTORopathies, emphasizing contemporary genetic methods in understanding and diagnosing the condition.
Collapse
Affiliation(s)
- Alice Man
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Matteo Di Scipio
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Shan Grewal
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Yujin Suk
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Elisabetta Trinari
- Division of Developmental Pediatrics, Department of Pediatrics, McMaster Children’s Hospital, Hamilton, ON L8N 3Z5, Canada
| | - Resham Ejaz
- Division of Genetics, Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Robyn Whitney
- Division of Neurology, Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
6
|
Nguyen LH, Xu Y, Nair M, Bordey A. The mTOR pathway genes MTOR, Rheb, Depdc5, Pten, and Tsc1 have convergent and divergent impacts on cortical neuron development and function. eLife 2024; 12:RP91010. [PMID: 38411613 PMCID: PMC10942629 DOI: 10.7554/elife.91010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024] Open
Abstract
Brain somatic mutations in various components of the mTOR complex 1 (mTORC1) pathway have emerged as major causes of focal malformations of cortical development and intractable epilepsy. While these distinct gene mutations converge on excessive mTORC1 signaling and lead to common clinical manifestations, it remains unclear whether they cause similar cellular and synaptic disruptions underlying cortical network hyperexcitability. Here, we show that in utero activation of the mTORC1 activator genes, Rheb or MTOR, or biallelic inactivation of the mTORC1 repressor genes, Depdc5, Tsc1, or Pten in the mouse medial prefrontal cortex leads to shared alterations in pyramidal neuron morphology, positioning, and membrane excitability but different changes in excitatory synaptic transmission. Our findings suggest that, despite converging on mTORC1 signaling, mutations in different mTORC1 pathway genes differentially impact cortical excitatory synaptic activity, which may confer gene-specific mechanisms of hyperexcitability and responses to therapeutic intervention.
Collapse
Affiliation(s)
- Lena H Nguyen
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at DallasRichardsonUnited States
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| | - Youfen Xu
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| | - Maanasi Nair
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| | - Angelique Bordey
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| |
Collapse
|
7
|
Nguyen LH, Xu Y, Nair M, Bordey A. The mTOR pathway genes mTOR, Rheb, Depdc5, Pten, and Tsc1 have convergent and divergent impacts on cortical neuron development and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.11.553034. [PMID: 37609221 PMCID: PMC10441381 DOI: 10.1101/2023.08.11.553034] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Brain somatic mutations in various components of the mTOR complex 1 (mTORC1) pathway have emerged as major causes of focal malformations of cortical development and intractable epilepsy. While these distinct gene mutations converge on excessive mTORC1 signaling and lead to common clinical manifestations, it remains unclear whether they cause similar cellular and synaptic disruptions underlying cortical network hyperexcitability. Here, we show that in utero activation of the mTORC1 activators, Rheb or mTOR, or biallelic inactivation of the mTORC1 repressors, Depdc5, Tsc1, or Pten in mouse medial prefrontal cortex leads to shared alterations in pyramidal neuron morphology, positioning, and membrane excitability but different changes in excitatory synaptic transmission. Our findings suggest that, despite converging on mTORC1 signaling, mutations in different mTORC1 pathway genes differentially impact cortical excitatory synaptic activity, which may confer gene-specific mechanisms of hyperexcitability and responses to therapeutic intervention.
Collapse
Affiliation(s)
- Lena H. Nguyen
- Department Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Youfen Xu
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Maanasi Nair
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Angelique Bordey
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
8
|
Liénard C, Pintart A, Bomont P. Neuronal Autophagy: Regulations and Implications in Health and Disease. Cells 2024; 13:103. [PMID: 38201307 PMCID: PMC10778363 DOI: 10.3390/cells13010103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/02/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Autophagy is a major degradative pathway that plays a key role in sustaining cell homeostasis, integrity, and physiological functions. Macroautophagy, which ensures the clearance of cytoplasmic components engulfed in a double-membrane autophagosome that fuses with lysosomes, is orchestrated by a complex cascade of events. Autophagy has a particularly strong impact on the nervous system, and mutations in core components cause numerous neurological diseases. We first review the regulation of autophagy, from autophagosome biogenesis to lysosomal degradation and associated neurodevelopmental/neurodegenerative disorders. We then describe how this process is specifically regulated in the axon and in the somatodendritic compartment and how it is altered in diseases. In particular, we present the neuronal specificities of autophagy, with the spatial control of autophagosome biogenesis, the close relationship of maturation with axonal transport, and the regulation by synaptic activity. Finally, we discuss the physiological functions of autophagy in the nervous system, during development and in adulthood.
Collapse
Affiliation(s)
- Caroline Liénard
- NeuroMyoGene Institute—PGNM, CNRS UMR 5261—INSERM U1315, University of Claude Bernard Lyon 1, 69008 Lyon, France; (C.L.); (A.P.)
- CHU Montpellier, University of Montpellier, 34295 Montpellier, France
| | - Alexandre Pintart
- NeuroMyoGene Institute—PGNM, CNRS UMR 5261—INSERM U1315, University of Claude Bernard Lyon 1, 69008 Lyon, France; (C.L.); (A.P.)
| | - Pascale Bomont
- NeuroMyoGene Institute—PGNM, CNRS UMR 5261—INSERM U1315, University of Claude Bernard Lyon 1, 69008 Lyon, France; (C.L.); (A.P.)
| |
Collapse
|
9
|
Krochmalnek E, Accogli A, St-Onge J, Addour-Boudrahem N, Prakash G, Kim SH, Brunette-Clement T, Alhajaj G, Mougharbel L, Bruneau E, Myers KA, Dubeau F, Karamchandani J, Farmer JP, Atkinson J, Hall J, Chantal Poulin C, Rosenblatt B, Lafond-Lapalme J, Weil A, Fallet-Bianco C, Albrecht S, Sonenberg N, Riviere JB, Dudley RW, Srour M. mTOR Pathway Somatic Pathogenic Variants in Focal Malformations of Cortical Development: Novel Variants, Topographic Mapping, and Clinical Outcomes. Neurol Genet 2023; 9:e200103. [PMID: 37900581 PMCID: PMC10602370 DOI: 10.1212/nxg.0000000000200103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/06/2023] [Indexed: 10/31/2023]
Abstract
Background and Objectives Somatic and germline pathogenic variants in genes of the mammalian target of rapamycin (mTOR) signaling pathway are a common mechanism underlying a subset of focal malformations of cortical development (FMCDs) referred to as mTORopathies, which include focal cortical dysplasia (FCD) type II, subtypes of polymicrogyria, and hemimegalencephaly. Our objective is to screen resected FMCD specimens with mTORopathy features on histology for causal somatic variants in mTOR pathway genes, describe novel pathogenic variants, and examine the variant distribution in relation to neuroimaging, histopathologic classification, and clinical outcomes. Methods We performed ultra-deep sequencing using a custom HaloPlexHS Target Enrichment kit in DNA from 21 resected fresh-frozen histologically confirmed FCD type II, tuberous sclerosis complex, or hemimegalencephaly specimens. We mapped the variant alternative allele frequency (AAF) across the resected brain using targeted ultra-deep sequencing in multiple formalin-fixed paraffin-embedded tissue blocks. We also functionally validated 2 candidate somatic MTOR variants and performed targeted RNA sequencing to validate a splicing defect associated with a novel DEPDC5 variant. Results We identified causal mTOR pathway gene variants in 66.7% (14/21) of patients, of which 13 were somatic with AAF ranging between 0.6% and 12.0%. Moreover, the AAF did not predict balloon cell presence. Favorable seizure outcomes were associated with genetically clear resection borders. Individuals in whom a causal somatic variant was undetected had excellent postsurgical outcomes. In addition, we demonstrate pathogenicity of the novel c.4373_4375dupATG and candidate c.7499T>A MTOR variants in vitro. We also identified a novel germline aberrant splice site variant in DEPDC5 (c.2802-1G>C). Discussion The AAF of somatic pathogenic variants correlated with the topographic distribution, histopathology, and postsurgical outcomes. Moreover, cortical regions with absent histologic FCD features had negligible or undetectable pathogenic variant loads. By contrast, specimens with frank histologic abnormalities had detectable pathogenic variant loads, which raises important questions as to whether there is a tolerable variant threshold and whether surgical margins should be clean, as performed in tumor resections. In addition, we describe 2 novel pathogenic variants, expanding the mTORopathy genetic spectrum. Although most pathogenic somatic variants are located at mutation hotspots, screening the full-coding gene sequence remains necessary in a subset of patients.
Collapse
Affiliation(s)
- Eric Krochmalnek
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Andrea Accogli
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Judith St-Onge
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Nassima Addour-Boudrahem
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Gyan Prakash
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Sung-Hoon Kim
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Tristan Brunette-Clement
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Ghadd Alhajaj
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Lina Mougharbel
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Elena Bruneau
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Kenneth A Myers
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Francois Dubeau
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Jason Karamchandani
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Jean-Pierre Farmer
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Jeffrey Atkinson
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Jeffrey Hall
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Chantal Chantal Poulin
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Bernard Rosenblatt
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Joel Lafond-Lapalme
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Alexander Weil
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Catherine Fallet-Bianco
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Steffen Albrecht
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Nahum Sonenberg
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Jean-Baptiste Riviere
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Roy W Dudley
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| | - Myriam Srour
- From the Research Institute of the McGill University Health Centre (E.K., J.S.-O., N.A.-B., L.M., E.B., K.A.M., J.L.-L., J.-B.R., R.W.D., M.S.); Integrated Program in Neuroscience (E.K.), McGill University; Department of Specialized Medicine (A.A.), McGill University Health Centre; Department of Human Genetics (A.A., J.-B.R.), Faculty of Medicine; Goodman Cancer Centre (G.P., S.-H.K., N.S.), Department of Biochemistry, McGill University; Department of Pediatric Neurosurgery (T.B.-C., A.W.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal; Division of Pediatric Neurology (G.A., K.A.M., C.C.P., M.S.), Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Department of Pediatrics (G.A.), Unaizah College of Medicine and Medical Sciences, Qassim University, Saudi Arabia; Department of Neurology and Neurosurgery (K.A.M., F.D., J.H., C.C.P., M.S.), McGill University Health Centre; Department of Pathology (J.K., S.A.), McGill University; Division of Neurosurgery (J.-P.F., J.A., R.W.D.), Department of Pediatric Surgery, McGill University Health Center; McGill University (B.R.); Department of Pathology (C.F.-B.), Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Quebec, Canada
| |
Collapse
|
10
|
Honke J, Hoffmann L, Coras R, Kobow K, Leu C, Pieper T, Hartlieb T, Bien CG, Woermann F, Cloppenborg T, Kalbhenn T, Gaballa A, Hamer H, Brandner S, Rössler K, Dörfler A, Rampp S, Lemke JR, Baldassari S, Baulac S, Lal D, Nürnberg P, Blümcke I. Deep histopathology genotype-phenotype analysis of focal cortical dysplasia type II differentiates between the GATOR1-altered autophagocytic subtype IIa and MTOR-altered migration deficient subtype IIb. Acta Neuropathol Commun 2023; 11:179. [PMID: 37946310 PMCID: PMC10633947 DOI: 10.1186/s40478-023-01675-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/21/2023] [Indexed: 11/12/2023] Open
Abstract
Focal cortical dysplasia type II (FCDII) is the most common cause of drug-resistant focal epilepsy in children. Herein, we performed a deep histopathology-based genotype-phenotype analysis to further elucidate the clinico-pathological and genetic presentation of FCDIIa compared to FCDIIb. Seventeen individuals with histopathologically confirmed diagnosis of FCD ILAE Type II and a pathogenic variant detected in brain derived DNA whole-exome sequencing or mTOR gene panel sequencing were included in this study. Clinical data were directly available from each contributing centre. Histopathological analyses were performed from formalin-fixed, paraffin-embedded tissue samples using haematoxylin-eosin and immunohistochemistry for NF-SMI32, NeuN, pS6, p62, and vimentin. Ten individuals carried loss-of-function variants in the GATOR1 complex encoding genes DEPDC5 (n = 7) and NPRL3 (n = 3), or gain-of-function variants in MTOR (n = 7). Whereas individuals with GATOR1 variants only presented with FCDIIa, i.e., lack of balloon cells, individuals with MTOR variants presented with both histopathology subtypes, FCDIIa and FCDIIb. Interestingly, 50% of GATOR1-positive cases showed a unique and predominantly vacuolizing phenotype with p62 immunofluorescent aggregates in autophagosomes. All cases with GATOR1 alterations had neurosurgery in the frontal lobe and the majority was confined to the cortical ribbon not affecting the white matter. This pattern was reflected by subtle or negative MRI findings in seven individuals with GATOR1 variants. Nonetheless, all individuals were seizure-free after surgery except four individuals carrying a DEPDC5 variant. We describe a yet underrecognized genotype-phenotype correlation of GATOR1 variants with FCDIIa in the frontal lobe. These lesions were histopathologically characterized by abnormally vacuolizing cells suggestive of an autophagy-altered phenotype. In contrast, individuals with FCDIIb and brain somatic MTOR variants showed larger lesions on MRI including the white matter, suggesting compromised neural cell migration.
Collapse
Affiliation(s)
- Jonas Honke
- Department of Neuropathology, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
- Partner of the European Reference Network (ERN) EpiCARE, Barcelona, Spain
| | - Lucas Hoffmann
- Department of Neuropathology, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
- Partner of the European Reference Network (ERN) EpiCARE, Barcelona, Spain
| | - Roland Coras
- Department of Neuropathology, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
- Partner of the European Reference Network (ERN) EpiCARE, Barcelona, Spain
| | - Katja Kobow
- Department of Neuropathology, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
- Partner of the European Reference Network (ERN) EpiCARE, Barcelona, Spain
| | - Costin Leu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, USA
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, UK
- Department of Neurology, McGovern Medical School, UTHealth Houston, University of Texas, Houston, USA
| | - Tom Pieper
- Center for Pediatric Neurology, Neurorehabilitation, and Epileptology, Schoen-Clinic, Vogtareuth, Germany
| | - Till Hartlieb
- Center for Pediatric Neurology, Neurorehabilitation, and Epileptology, Schoen-Clinic, Vogtareuth, Germany
- Research Institute for Rehabilitation, Transition, and Palliation, Paracelsus Medical University, Salzburg, Austria
| | - Christian G Bien
- Department of Epileptology (Krankenhaus Mara), Medical School, Bielefeld University, Bielefeld, Germany
| | - Friedrich Woermann
- Department of Epileptology (Krankenhaus Mara), Medical School, Bielefeld University, Bielefeld, Germany
| | - Thomas Cloppenborg
- Department of Epileptology (Krankenhaus Mara), Medical School, Bielefeld University, Bielefeld, Germany
| | - Thilo Kalbhenn
- Department of Epileptology (Krankenhaus Mara), Medical School, Bielefeld University, Bielefeld, Germany
- Department of Neurosurgery (Evangelisches Klinikum Bethel), Medical School, Bielefeld University, Bielefeld, Germany
| | - Ahmed Gaballa
- Department of Epileptology (Krankenhaus Mara), Medical School, Bielefeld University, Bielefeld, Germany
| | - Hajo Hamer
- Partner of the European Reference Network (ERN) EpiCARE, Barcelona, Spain
- Epilepsy Center, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Brandner
- Department of Neurosurgery, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Karl Rössler
- Department of Neurosurgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Arnd Dörfler
- Department of Neuroradiology, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Rampp
- Department of Neurosurgery, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
- Department of Neuroradiology, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
- Center for Rare Diseases, University of Leipzig Medical Center, Leipzig, Germany
| | - Sara Baldassari
- Inserm, CNRS, APHP, Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de La Pitié Salpêtrière, Sorbonne Université, Paris, France
| | - Stéphanie Baulac
- Inserm, CNRS, APHP, Institut du Cerveau - Paris Brain Institute - ICM, Hôpital de La Pitié Salpêtrière, Sorbonne Université, Paris, France
| | - Dennis Lal
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and M.I.T, Cambridge, MA, 02142, USA
- Cologne Center for Genomics (CCG), Medical Faculty of the University of Cologne, University Hospital of Cologne, 50931, Cologne, Germany
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, UK
- Department of Neurology, McGovern Medical School, UTHealth Houston, University of Texas, Houston, USA
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG), Medical Faculty of the University of Cologne, University Hospital of Cologne, 50931, Cologne, Germany
| | - Ingmar Blümcke
- Department of Neuropathology, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany.
- Partner of the European Reference Network (ERN) EpiCARE, Barcelona, Spain.
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and M.I.T, Cambridge, MA, 02142, USA.
| |
Collapse
|
11
|
Guo M, Zhang J, Wang J, Wang X, Gao Q, Tang C, Deng J, Xiong Z, Kong X, Guan Y, Zhou J, Boison D, Luan G, Li T. Aberrant adenosine signaling in patients with focal cortical dysplasia. Mol Neurobiol 2023; 60:4396-4417. [PMID: 37103687 PMCID: PMC10330374 DOI: 10.1007/s12035-023-03351-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 04/13/2023] [Indexed: 04/28/2023]
Abstract
Focal cortical dysplasia (FCD), a common malformation of cortical development, is frequently associated with pharmacoresistant epilepsy in both children and adults. Adenosine is an inhibitory modulator of brain activity and a prospective anti-seizure agent with potential for clinical translation. Our previous results demonstrated that the major adenosine-metabolizing enzyme adenosine kinase (ADK) was upregulated in balloon cells (BCs) within FCD type IIB lesions, suggesting that dysfunction of the adenosine system is implicated in the pathophysiology of FCD. In our current study, we therefore performed a comprehensive analysis of adenosine signaling in surgically resected cortical specimens from patients with FCD type I and type II via immunohistochemistry and immunoblot analysis. Adenosine enzyme signaling was assessed by quantifying the levels of the key enzymes of adenosine metabolism, i.e., ADK, adenosine deaminase (ADA), and ecto-5'-nucleotidase (CD73). Adenosine receptor signaling was assessed by quantifying the levels of adenosine A2A receptor (A2AR) and putative downstream mediators of adenosine, namely, glutamate transporter-1 (GLT-1) and mammalian target of rapamycin (mTOR). Within lesions in FCD specimens, we found that the adenosine-metabolizing enzymes ADK and ADA, as well as the adenosine-producing enzyme CD73, were upregulated. We also observed an increase in A2AR density, as well as a decrease in GLT-1 levels and an increase in mTOR levels, in FCD specimens compared with control tissue. These results suggest that dysregulation of the adenosine system is a common pathologic feature of both FCD type I and type II. The adenosine system might therefore be a therapeutic target for the treatment of epilepsy associated with FCD.
Collapse
Affiliation(s)
- Mengyi Guo
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jing Zhang
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jing Wang
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Xiongfei Wang
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurosurgery, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Qing Gao
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Chongyang Tang
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurosurgery, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jiahui Deng
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Zhonghua Xiong
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Xiangru Kong
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Yuguang Guan
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurosurgery, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Jian Zhou
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
- Department of Neurosurgery, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson & New Jersey Medical Schools, Rutgers University, Piscataway, NJ, 08854, USA
| | - Guoming Luan
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China.
- Department of Neurosurgery, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China.
| | - Tianfu Li
- Department of Brain Institute, Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing Key Laboratory of Epilepsy Research, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China.
- Department of Neurology, Center of Epilepsy, Beijing Institute for Brain Disorders, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China.
| |
Collapse
|
12
|
Lee HM, Hong SJ, Gill R, Caldairou B, Wang I, Zhang JG, Deleo F, Schrader D, Bartolomei F, Guye M, Cho KH, Barba C, Sisodiya S, Jackson G, Hogan RE, Wong-Kisiel L, Cascino GD, Schulze-Bonhage A, Lopes-Cendes I, Cendes F, Guerrini R, Bernhardt B, Bernasconi N, Bernasconi A. Multimodal mapping of regional brain vulnerability to focal cortical dysplasia. Brain 2023; 146:3404-3415. [PMID: 36852571 PMCID: PMC10393418 DOI: 10.1093/brain/awad060] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/17/2023] [Accepted: 02/02/2023] [Indexed: 03/01/2023] Open
Abstract
Focal cortical dysplasia (FCD) type II is a highly epileptogenic developmental malformation and a common cause of surgically treated drug-resistant epilepsy. While clinical observations suggest frequent occurrence in the frontal lobe, mechanisms for such propensity remain unexplored. Here, we hypothesized that cortex-wide spatial associations of FCD distribution with cortical cytoarchitecture, gene expression and organizational axes may offer complementary insights into processes that predispose given cortical regions to harbour FCD. We mapped the cortex-wide MRI distribution of FCDs in 337 patients collected from 13 sites worldwide. We then determined its associations with (i) cytoarchitectural features using histological atlases by Von Economo and Koskinas and BigBrain; (ii) whole-brain gene expression and spatiotemporal dynamics from prenatal to adulthood stages using the Allen Human Brain Atlas and PsychENCODE BrainSpan; and (iii) macroscale developmental axes of cortical organization. FCD lesions were preferentially located in the prefrontal and fronto-limbic cortices typified by low neuron density, large soma and thick grey matter. Transcriptomic associations with FCD distribution uncovered a prenatal component related to neuroglial proliferation and differentiation, likely accounting for the dysplastic makeup, and a postnatal component related to synaptogenesis and circuit organization, possibly contributing to circuit-level hyperexcitability. FCD distribution showed a strong association with the anterior region of the antero-posterior axis derived from heritability analysis of interregional structural covariance of cortical thickness, but not with structural and functional hierarchical axes. Reliability of all results was confirmed through resampling techniques. Multimodal associations with cytoarchitecture, gene expression and axes of cortical organization indicate that prenatal neurogenesis and postnatal synaptogenesis may be key points of developmental vulnerability of the frontal lobe to FCD. Concordant with a causal role of atypical neuroglial proliferation and growth, our results indicate that FCD-vulnerable cortices display properties indicative of earlier termination of neurogenesis and initiation of cell growth. They also suggest a potential contribution of aberrant postnatal synaptogenesis and circuit development to FCD epileptogenicity.
Collapse
Affiliation(s)
- Hyo M Lee
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Seok-Jun Hong
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
- Center for Neuroscience Imaging, Research Institute for Basic Science, Department of Global Biomedical Engineering, SungKyunKwan University, Suwon, KoreaSuwon, Korea
| | - Ravnoor Gill
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Benoit Caldairou
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Irene Wang
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jian-guo Zhang
- Department of Functional Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Francesco Deleo
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milano, Italy
| | - Dewi Schrader
- Department of Pediatrics, British Columbia Children’s Hospital, Vancouver, Canada
| | - Fabrice Bartolomei
- Aix Marseille Univ, INSERM, INS, Institut de Neurosciences des Systèmes, Marseille, 13005, France
| | - Maxime Guye
- Aix Marseille University, CNRS, CRMBM UMR 7339, Marseille, France
| | - Kyoo Ho Cho
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Carmen Barba
- Meyer Children's Hospital IRCCS, Florence, Italy
- University of Florence, 50121 Florence, Italy
| | - Sanjay Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Graeme Jackson
- The Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Victoria, Australia
| | - R Edward Hogan
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | - Iscia Lopes-Cendes
- Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP) and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas SP, Brazil
| | - Fernando Cendes
- Department of Neurology, School of Medical Sciences, University of Campinas (UNICAMP), and the Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas SP, Brazil
| | - Renzo Guerrini
- Meyer Children's Hospital IRCCS, Florence, Italy
- University of Florence, 50121 Florence, Italy
| | - Boris Bernhardt
- Multimodal Imaging and Connectome Analysis Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Neda Bernasconi
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Andrea Bernasconi
- Neuroimaging of Epilepsy Laboratory, Montreal Neurological Institute, McGill University, Montreal, Canada
| |
Collapse
|
13
|
Boßelmann CM, Leu C, Lal D. Technological and computational approaches to detect somatic mosaicism in epilepsy. Neurobiol Dis 2023:106208. [PMID: 37343892 DOI: 10.1016/j.nbd.2023.106208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/03/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023] Open
Abstract
Lesional epilepsy is a common and severe disease commonly associated with malformations of cortical development, including focal cortical dysplasia and hemimegalencephaly. Recent advances in sequencing and variant calling technologies have identified several genetic causes, including both short/single nucleotide and structural somatic variation. In this review, we aim to provide a comprehensive overview of the methodological advancements in this field while highlighting the unresolved technological and computational challenges that persist, including ultra-low variant allele fractions in bulk tissue, low availability of paired control samples, spatial variability of mutational burden within the lesion, and the issue of false-positive calls and validation procedures. Information from genetic testing in focal epilepsy may be integrated into clinical care to inform histopathological diagnosis, postoperative prognosis, and candidate precision therapies.
Collapse
Affiliation(s)
- Christian M Boßelmann
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Costin Leu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, UK.
| | - Dennis Lal
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and M.I.T., Cambridge, MA, USA; Cologne Center for Genomics (CCG), University of Cologne, Cologne, DE, USA
| |
Collapse
|
14
|
Gerasimenko A, Baldassari S, Baulac S. mTOR pathway: Insights into an established pathway for brain mosaicism in epilepsy. Neurobiol Dis 2023; 182:106144. [PMID: 37149062 DOI: 10.1016/j.nbd.2023.106144] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) signaling pathway is an essential regulator of numerous cellular activities such as metabolism, growth, proliferation, and survival. The mTOR cascade recently emerged as a critical player in the pathogenesis of focal epilepsies and cortical malformations. The 'mTORopathies' comprise a spectrum of cortical malformations that range from whole brain (megalencephaly) and hemispheric (hemimegalencephaly) abnormalities to focal abnormalities, such as focal cortical dysplasia type II (FCDII), which manifest with drug-resistant epilepsies. The spectrum of cortical dysplasia results from somatic brain mutations in the mTOR pathway activators AKT3, MTOR, PIK3CA, and RHEB and from germline and somatic mutations in mTOR pathway repressors, DEPDC5, NPRL2, NPRL3, TSC1 and TSC2. The mTORopathies are characterized by excessive mTOR pathway activation, leading to a broad range of structural and functional impairments. Here, we provide a comprehensive literature review of somatic mTOR-activating mutations linked to epilepsy and cortical malformations in 292 patients and discuss the perspectives of targeted therapeutics for personalized medicine.
Collapse
Affiliation(s)
- Anna Gerasimenko
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, 75013 Paris, France; APHP Sorbonne Université, GH Pitié Salpêtrière et Trousseau, Département de Génétique, Centre de référence "déficiences intellectuelles de causes rares", Paris, France
| | - Sara Baldassari
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, 75013 Paris, France
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, 75013 Paris, France.
| |
Collapse
|
15
|
Bychkova E, Dorofeeva M, Levov A, Kislyakov A, Karandasheva K, Strelnikov V, Anoshkin K. Specific Features of Focal Cortical Dysplasia in Tuberous Sclerosis Complex. Curr Issues Mol Biol 2023; 45:3977-3996. [PMID: 37232723 DOI: 10.3390/cimb45050254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Patients with tuberous sclerosis complex present with cognitive, behavioral, and psychiatric impairments, such as intellectual disabilities, autism spectrum disorders, and drug-resistant epilepsy. It has been shown that these disorders are associated with the presence of cortical tubers. Tuberous sclerosis complex results from inactivating mutations in the TSC1 or TSC2 genes, resulting in hyperactivation of the mTOR signaling pathway, which regulates cell growth, proliferation, survival, and autophagy. TSC1 and TSC2 are classified as tumor suppressor genes and function according to Knudson's two-hit hypothesis, which requires both alleles to be damaged for tumor formation. However, a second-hit mutation is a rare event in cortical tubers. This suggests that the molecular mechanism of cortical tuber formation may be more complicated and requires further research. This review highlights the issues of molecular genetics and genotype-phenotype correlations, considers histopathological characteristics and the mechanism of morphogenesis of cortical tubers, and also presents data on the relationship between these formations and the development of neurological manifestations, as well as treatment options.
Collapse
Affiliation(s)
- Ekaterina Bychkova
- Research Centre for Medical Genetics, Moskvorechye Street 1, 115522 Moscow, Russia
- Faculty of Biomedicine, Pirogov Russian National Research Medical University, Ostrovityanova Street 1, 117997 Moscow, Russia
| | - Marina Dorofeeva
- Veltischev Research and Clinical Institute for Pediatrics and Pediatric Surgery, Pirogov Russian National Research Medical University, Taldomskaya 2, 125412 Moscow, Russia
| | - Aleksandr Levov
- Morozov Children's City Clinical Hospital, 4th Dobryninsky Lane, 1/9, 119049 Moscow, Russia
| | - Alexey Kislyakov
- Morozov Children's City Clinical Hospital, 4th Dobryninsky Lane, 1/9, 119049 Moscow, Russia
| | | | - Vladimir Strelnikov
- Research Centre for Medical Genetics, Moskvorechye Street 1, 115522 Moscow, Russia
| | - Kirill Anoshkin
- Research Centre for Medical Genetics, Moskvorechye Street 1, 115522 Moscow, Russia
| |
Collapse
|
16
|
Shiraishi H, Teramoto T, Yokoshiki S, Tohyama J, Ueda Y, Egawa K, Sato N, Manabe A, Kato M. Efficacy of sirolimus for epileptic seizures in childhood associated with focal cortical dysplasia type II. Brain Dev 2023; 45:343-347. [PMID: 36870920 DOI: 10.1016/j.braindev.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023]
Abstract
OBJECTIVE The efficacy of the mechanistic target of rapamycin inhibitor, sirolimus, was recently reported for patients more than 6 years of age by Kato et al. We evaluated the efficacy and safety of sirolimus in a 2-year-old patient with recurrent focal seizures with impaired consciousness after focal cortical dysplasia (FCD) type IIa resection. METHODS The patient was a 2-year-old girl who had recurrent seizures after undergoing FCD resection at 4 months of age. The initial dose of sirolimus was 0.5 mg/day and was gradually increased using the trough blood concentration before oral administration as an index, and evaluation was performed at 92 weeks. RESULTS The trough blood level of sirolimus was increased to 6.1 ng/mL and maintenance therapy was started at 40 weeks. Focal seizures with impairment of consciousness with tonic extension of the limbs decreased. No critically serious adverse events occurred. CONCLUSION Sirolimus was effective against epileptic seizures from FCD type II even for a child under 5 years of age. There were no critically serious adverse events and administration could be continued.
Collapse
Affiliation(s)
- Hideaki Shiraishi
- Department of Pediatrics, Hokkaido University Hospital, Hokkaido 060-8648, Japan.
| | - Tsuyoshi Teramoto
- Hokkaido University Hospital Clinical Research and Medical Innovation Center, Research and Development Division, Hokkaido 060-8648, Japan; University of Toyama Hospital, Center for Clinical Research, Toyama 930-0194, Japan
| | - Saki Yokoshiki
- Hokkaido University Hospital Clinical Research and Medical Innovation Center, Research and Development Division, Hokkaido 060-8648, Japan
| | - Jun Tohyama
- Department of Child Neurology, National Hospital Organization Nishi-Niigata Chuo National Hospital, Niigata 950-2085, Japan
| | - Yuki Ueda
- Department of Pediatrics, Hokkaido University Hospital, Hokkaido 060-8648, Japan
| | - Kiyoshi Egawa
- Department of Pediatrics, Hokkaido University Hospital, Hokkaido 060-8648, Japan
| | - Norihiro Sato
- Hokkaido University Hospital Clinical Research and Medical Innovation Center, Research and Development Division, Hokkaido 060-8648, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University Hospital, Hokkaido 060-8648, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo 142-8555, Japan.
| |
Collapse
|
17
|
Fujita A, Kato M, Sugano H, Iimura Y, Suzuki H, Tohyama J, Fukuda M, Ito Y, Baba S, Okanishi T, Enoki H, Fujimoto A, Yamamoto A, Kawamura K, Kato S, Honda R, Ono T, Shiraishi H, Egawa K, Shirai K, Yamamoto S, Hayakawa I, Kawawaki H, Saida K, Tsuchida N, Uchiyama Y, Hamanaka K, Miyatake S, Mizuguchi T, Nakashima M, Saitsu H, Miyake N, Kakita A, Matsumoto N. An integrated genetic analysis of epileptogenic brain malformed lesions. Acta Neuropathol Commun 2023; 11:33. [PMID: 36864519 PMCID: PMC9983246 DOI: 10.1186/s40478-023-01532-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/17/2023] [Indexed: 03/04/2023] Open
Abstract
Focal cortical dysplasia is the most common malformation during cortical development, sometimes excised by epilepsy surgery and often caused by somatic variants of the mTOR pathway genes. In this study, we performed a genetic analysis of epileptogenic brain malformed lesions from 64 patients with focal cortical dysplasia, hemimegalencephy, brain tumors, or hippocampal sclerosis. Targeted sequencing, whole-exome sequencing, and single nucleotide polymorphism microarray detected four germline and 35 somatic variants, comprising three copy number variants and 36 single nucleotide variants and indels in 37 patients. One of the somatic variants in focal cortical dysplasia type IIB was an in-frame deletion in MTOR, in which only gain-of-function missense variants have been reported. In focal cortical dysplasia type I, somatic variants of MAP2K1 and PTPN11 involved in the RAS/MAPK pathway were detected. The in-frame deletions of MTOR and MAP2K1 in this study resulted in the activation of the mTOR pathway in transiently transfected cells. In addition, the PTPN11 missense variant tended to elongate activation of the mTOR or RAS/MAPK pathway, depending on culture conditions. We demonstrate that epileptogenic brain malformed lesions except for focal cortical dysplasia type II arose from somatic variants of diverse genes but were eventually linked to the mTOR pathway.
Collapse
Affiliation(s)
- Atsushi Fujita
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo, 142-8666, Japan
| | - Hidenori Sugano
- Department of Neurosurgery, Epilepsy Center, Juntendo University, Tokyo, 113-8421, Japan
| | - Yasushi Iimura
- Department of Neurosurgery, Epilepsy Center, Juntendo University, Tokyo, 113-8421, Japan
| | - Hiroharu Suzuki
- Department of Neurosurgery, Epilepsy Center, Juntendo University, Tokyo, 113-8421, Japan
| | - Jun Tohyama
- Department of Child Neurology, National Hospital Organization Nishiniigata Chuo Hospital, Niigata, 950-2085, Japan
| | - Masafumi Fukuda
- Department of Functional Neurosurgery, Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital, Niigata, 950-2085, Japan
| | - Yosuke Ito
- Department of Functional Neurosurgery, Epilepsy Center, National Hospital Organization Nishiniigata Chuo Hospital, Niigata, 950-2085, Japan
| | - Shimpei Baba
- Department of Child Neurology, Comprehensive Epilepsy Center, Seirei Hamamatsu General Hospital, Hamamatsu, 430-8558, Japan
| | - Tohru Okanishi
- Division of Child Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, 683-8503, Japan
| | - Hideo Enoki
- Department of Pediatrics, Kawasaki Medical School, Kurashiki, 701-0192, Japan
| | - Ayataka Fujimoto
- Comprehensive Epilepsy Center, Seirei Hamamatsu General Hospital, Hamamatsu, 430-8558, Japan
| | - Akiyo Yamamoto
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Kentaro Kawamura
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Shinsuke Kato
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Ryoko Honda
- Department of Pediatrics, National Hospital Organization Nagasaki Medical Center, Omura, 856-8562, Japan
| | - Tomonori Ono
- Epilepsy Center, National Hospital Organization Nagasaki Medical Center, Omura, 856-8562, Japan
| | - Hideaki Shiraishi
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
| | - Kiyoshi Egawa
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
| | - Kentaro Shirai
- Department of Pediatrics, Tsuchiura Kyodo General Hospital, Tsuchiura, 300-0028, Japan
| | - Shinji Yamamoto
- Department of Neurosurgery, Tsuchiura Kyodo General Hospital, Tsuchiura, 300-0028, Japan
| | - Itaru Hayakawa
- Division of Neurology, National Center for Child Health and Development, Tokyo, 157-8535, Japan
| | - Hisashi Kawawaki
- Department of Pediatric Neurology, Children's Medical Center, Osaka City General Hospital, Osaka, 534-0021, Japan
| | - Ken Saida
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Naomi Tsuchida
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan.,Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, 236-0004, Japan
| | - Yuri Uchiyama
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan.,Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, 236-0004, Japan
| | - Kohei Hamanaka
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan.,Department of Clinical Genetics, Yokohama City University Hospital, Yokohama, 236-0004, Japan
| | - Takeshi Mizuguchi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Mitsuko Nakashima
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan.,Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Hirotomo Saitsu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan.,Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan.,Department of Human Genetics, Research Institute, National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
18
|
Lee WS, Macdonald-Laurs E, Stephenson SEM, D'Arcy C, MacGregor D, Leventer RJ, Maixner W, Harvey AS, Lockhart PJ. Basal ganglia dysplasia and mTORopathy: A potential cause of postoperative seizures in focal cortical dysplasia. Epilepsia Open 2023; 8:205-210. [PMID: 36461712 PMCID: PMC9977751 DOI: 10.1002/epi4.12678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/01/2022] [Indexed: 12/05/2022] Open
Abstract
Pathogenic somatic MTOR variants in the cerebral cortex are a frequent cause of focal cortical dysplasia (FCD). We describe a child with drug and surgery-resistant focal epilepsy due to FCD type II who developed progressive enlargement and T2 signal hyperintensity in the ipsilateral caudate and lentiform nuclei. Histopathology of caudate nucleus biopsies showed dysmorphic neurons, similar to those in resected cortex. Genetic analysis of frontal and temporal cortex and caudate nucleus identified a pathogenic somatic MTOR variant [NM_004958.4:c.4375G > C (p.Ala1459Pro)] that was not present in blood-derived gDNA. The mean variant allele frequency ranged from 0.4% to 3.2% in cerebral cortex and up to 5.4% in the caudate nucleus. The basal ganglia abnormalities suggest more widespread, potentially hemispheric dysplasia in this patient, consistent with the pathogenic variant occurring in early cerebral development. This finding provides a potential explanation for persistent seizures in some patients with seemingly complete resection of FCD or disconnection of a dysplastic hemisphere.
Collapse
Affiliation(s)
- Wei Shern Lee
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Emma Macdonald-Laurs
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Department of Neurology, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Sarah E M Stephenson
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Colleen D'Arcy
- Department of Anatomical Pathology, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Duncan MacGregor
- Department of Anatomical Pathology, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Richard J Leventer
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Department of Neurology, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Wirginia Maixner
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Department of Neurosurgery, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - A Simon Harvey
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Department of Neurology, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Paul J Lockhart
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
19
|
Wang Y, Yu T, Blümcke I, Cai Y, Sun K, Gao R, Wang Y, Fu Y, Wang W, Wang Y, Zhang G, Piao Y. The clinico-pathological characterisation of focal cortical dysplasia type IIb genetically defined by MTOR mosaicism. Neuropathol Appl Neurobiol 2023; 49:e12874. [PMID: 36544434 DOI: 10.1111/nan.12874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 08/18/2022] [Accepted: 10/11/2022] [Indexed: 12/24/2022]
Abstract
AIMS Focal cortical dysplasia (FCD) is a major cause of drug-resistant paediatric epilepsy and is amenable to successful neurosurgical resection. FCD ILAE Type IIb is the most common FCD subtype, and brain somatic mutations affecting the mTOR pathway play a major pathogenic role. The aim of this study was to comprehensively describe the genotype-phenotype association of 20 patients with histopathologically confirmed FCDIIb using next generation sequencing (NGS) of paired blood-brain samples. METHODS Clinical and neuropathological data were retrospectively reviewed from the hospital archive. The NGS panel included 11 mTOR-pathway-related genes with maximum coverage of 2000×. The detected variants were validated by digital droplet PCR. RESULTS Pathogenic MTOR variants were identified in 10 patients (50%). Further comparison with MTOR-wildtype FCDIIb suggested a profound genotype-phenotype association characterised by (1) a non-temporal lobe lesion on MRI, (2) a larger lesion volume occupying grey and white matter (3.032 ± 1.859 cm3 vs 1.110 ± 0.856 cm3 , p = 0.014), (3) more balloon cells (50.20 ± 14.40 BC/mm2 vs 31.64 ± 30.56 BC/mm2 , p = 0.099) and dysmorphic neurons (48.72 ± 19.47DN/mm2 vs 15.28 ± 13.95DN/mm2 , p = 0.000) and (4) a positive correlation between VAF and the lesion volume (r = 0.802, p = 0.017). CONCLUSIONS Our study identified frequent MTOR mutations in the cell-rich FCDIIb phenotype, clinically characterised by a non-temporal location and large lesion volume. Comprehensive genotype-phenotype associations will help us further explore and define the broad spectrum of FCD lesions to make more targeted therapies available in the realm of epileptology.
Collapse
Affiliation(s)
- Yajie Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China
| | - Tao Yu
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China.,Beijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Yanning Cai
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China.,Department of Neurobiology and Clinical Biobank, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ke Sun
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China.,Beijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Runshi Gao
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China.,Beijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yujiao Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China
| | - Yongjuan Fu
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China
| | - Wei Wang
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China.,Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuping Wang
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China.,Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neuromodulation, Beijing, China.,Center of Epilepsy, Institute of Sleep and Consciousness Disorders, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Guojun Zhang
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China.,Beijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yueshan Piao
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, Beijing, China
| |
Collapse
|
20
|
Kamble VS, Pachpor TA, Khandagale SB, Wagh VV, Khare SP. Translation initiation and dysregulation of initiation factors in rare diseases. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
21
|
Focal cortical dysplasia as a cause of epilepsy: The current evidence of associated genes and future therapeutic treatments. INTERDISCIPLINARY NEUROSURGERY 2022. [DOI: 10.1016/j.inat.2022.101635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
22
|
Xu Y, Zhao R, Wang M, Wang X, Wang Y, Li H, Ma Y, Wu B, Zhou Y. Identification of genetic characteristics in pediatric epilepsy with focal cortical dysplasia type 2 using deep whole-exome sequencing. Mol Genet Genomic Med 2022; 10:e2086. [PMID: 36342087 PMCID: PMC9747558 DOI: 10.1002/mgg3.2086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/26/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Focal cortical dysplasia type 2 (FCD2) is a malformation of cortical development that constitutes a common cause of pediatric focal epilepsy. Germline or somatic variants in the mammalian target of rapamycin (mTOR) signaling pathway genes are the pathogenesis of FCD2. OBJECTIVE In this study, whole-exome deep sequencing was performed on dysplastic cortex from focal epilepsy in children to explore genetic characteristics in FCD2. METHODS Resected core lesions of FCD2 were confirmed by pathology, and peripheral blood was collected from 11 patients. Deep whole-exome sequencing (>500X) was performed on derived genomic DNA, germline, or somatic variants in brain-specific genes were analyzed and identified. RESULTS In 11 patients, a heterozygous likely pathogenic germline variant of DEPDC5 was identified in one case, while somatic variants were found in four brain samples. The frequencies of the somatic variant allele were 2.52%-5.12%. Somatic variants in AKT3, TSC2, and MTOR (mTOR signaling pathway genes) were found in three samples. Besides, one somatic variant was detected in MED12 which has not been reported to associate with FCD2. CONCLUSION Our study expanded the variant spectrum in the mTOR-GATOR pathway, and also detected a somatic variant in MED12 which was potentially associated with FCD 2.
Collapse
Affiliation(s)
- Yan Xu
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Rui Zhao
- Department of NeurosurgeryChildren's Hospital of Fudan UniversityShanghaiChina
| | - Min Wang
- Department of NeurosurgeryChildren's Hospital of Fudan UniversityShanghaiChina
| | - Xin‐hua Wang
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yi Wang
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Hao Li
- Department of NeurosurgeryChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yang‐yang Ma
- Department of PathologyChildren's Hospital of Fudan UniversityShanghaiChina
| | - Bing‐bing Wu
- Center for Molecular MedicinePediatrics Research Institute, Children's Hospital of Fudan UniversityShanghaiChina
| | - Yuan‐feng Zhou
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| |
Collapse
|
23
|
Lai D, Gade M, Yang E, Koh HY, Lu J, Walley NM, Buckley AF, Sands TT, Akman CI, Mikati MA, McKhann GM, Goldman JE, Canoll P, Alexander AL, Park KL, Von Allmen GK, Rodziyevska O, Bhattacharjee MB, Lidov HGW, Vogel H, Grant GA, Porter BE, Poduri AH, Crino PB, Heinzen EL. Somatic variants in diverse genes leads to a spectrum of focal cortical malformations. Brain 2022; 145:2704-2720. [PMID: 35441233 PMCID: PMC9612793 DOI: 10.1093/brain/awac117] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/19/2022] [Accepted: 03/13/2022] [Indexed: 11/14/2022] Open
Abstract
Post-zygotically acquired genetic variants, or somatic variants, that arise during cortical development have emerged as important causes of focal epilepsies, particularly those due to malformations of cortical development. Pathogenic somatic variants have been identified in many genes within the PI3K-AKT-mTOR-signalling pathway in individuals with hemimegalencephaly and focal cortical dysplasia (type II), and more recently in SLC35A2 in individuals with focal cortical dysplasia (type I) or non-dysplastic epileptic cortex. Given the expanding role of somatic variants across different brain malformations, we sought to delineate the landscape of somatic variants in a large cohort of patients who underwent epilepsy surgery with hemimegalencephaly or focal cortical dysplasia. We evaluated samples from 123 children with hemimegalencephaly (n = 16), focal cortical dysplasia type I and related phenotypes (n = 48), focal cortical dysplasia type II (n = 44), or focal cortical dysplasia type III (n = 15). We performed high-depth exome sequencing in brain tissue-derived DNA from each case and identified somatic single nucleotide, indel and large copy number variants. In 75% of individuals with hemimegalencephaly and 29% with focal cortical dysplasia type II, we identified pathogenic variants in PI3K-AKT-mTOR pathway genes. Four of 48 cases with focal cortical dysplasia type I (8%) had a likely pathogenic variant in SLC35A2. While no other gene had multiple disease-causing somatic variants across the focal cortical dysplasia type I cohort, four individuals in this group had a single pathogenic or likely pathogenic somatic variant in CASK, KRAS, NF1 and NIPBL, genes previously associated with neurodevelopmental disorders. No rare pathogenic or likely pathogenic somatic variants in any neurological disease genes like those identified in the focal cortical dysplasia type I cohort were found in 63 neurologically normal controls (P = 0.017), suggesting a role for these novel variants. We also identified a somatic loss-of-function variant in the known epilepsy gene, PCDH19, present in a small number of alleles in the dysplastic tissue from a female patient with focal cortical dysplasia IIIa with hippocampal sclerosis. In contrast to focal cortical dysplasia type II, neither focal cortical dysplasia type I nor III had somatic variants in genes that converge on a unifying biological pathway, suggesting greater genetic heterogeneity compared to type II. Importantly, we demonstrate that focal cortical dysplasia types I, II and III are associated with somatic gene variants across a broad range of genes, many associated with epilepsy in clinical syndromes caused by germline variants, as well as including some not previously associated with radiographically evident cortical brain malformations.
Collapse
Affiliation(s)
- Dulcie Lai
- Division of Pharmacology and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Meethila Gade
- Division of Pharmacology and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Edward Yang
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hyun Yong Koh
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA.,Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jinfeng Lu
- Division of Pharmacology and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nicole M Walley
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anne F Buckley
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Tristan T Sands
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY 10032, USA.,Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Cigdem I Akman
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | - Mohamad A Mikati
- Department of Neurobiology, Duke University, Durham, NC 27708, USA.,Division of Pediatric Neurology, Duke University Medical Center, Durham, NC 27710, USA
| | - Guy M McKhann
- Department of Neurosurgery, Columbia University, New York Presbyterian Hospital, New York, NY 10032, USA
| | - James E Goldman
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Allyson L Alexander
- Department of Neurosurgery, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kristen L Park
- Department of Pediatrics and Neurology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Gretchen K Von Allmen
- Department of Neurology, McGovern Medical School, Houston, TX 77030, USA.,Division of Child Neurology, Department of Pediatrics, McGovern Medical School, Houston, TX 77030, USA
| | - Olga Rodziyevska
- Division of Child Neurology, Department of Pediatrics, McGovern Medical School, Houston, TX 77030, USA
| | | | - Hart G W Lidov
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Gerald A Grant
- Department of Neurosurgery, Lucile Packard Children's Hospital at Stanford, School of Medicine, Stanford, CA 94305, USA
| | - Brenda E Porter
- Department of Neurology and Neurological Sciences, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Annapurna H Poduri
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA.,Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Erin L Heinzen
- Division of Pharmacology and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
24
|
Najm I, Lal D, Alonso Vanegas M, Cendes F, Lopes-Cendes I, Palmini A, Paglioli E, Sarnat HB, Walsh CA, Wiebe S, Aronica E, Baulac S, Coras R, Kobow K, Cross JH, Garbelli R, Holthausen H, Rössler K, Thom M, El-Osta A, Lee JH, Miyata H, Guerrini R, Piao YS, Zhou D, Blümcke I. The ILAE consensus classification of focal cortical dysplasia: An update proposed by an ad hoc task force of the ILAE diagnostic methods commission. Epilepsia 2022; 63:1899-1919. [PMID: 35706131 PMCID: PMC9545778 DOI: 10.1111/epi.17301] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/24/2022] [Accepted: 05/13/2022] [Indexed: 02/05/2023]
Abstract
Ongoing challenges in diagnosing focal cortical dysplasia (FCD) mandate continuous research and consensus agreement to improve disease definition and classification. An International League Against Epilepsy (ILAE) Task Force (TF) reviewed the FCD classification of 2011 to identify existing gaps and provide a timely update. The following methodology was applied to achieve this goal: a survey of published literature indexed with ((Focal Cortical Dysplasia) AND (epilepsy)) between 01/01/2012 and 06/30/2021 (n = 1349) in PubMed identified the knowledge gained since 2012 and new developments in the field. An online survey consulted the ILAE community about the current use of the FCD classification scheme with 367 people answering. The TF performed an iterative clinico-pathological and genetic agreement study to objectively measure the diagnostic gap in blood/brain samples from 22 patients suspicious for FCD and submitted to epilepsy surgery. The literature confirmed new molecular-genetic characterizations involving the mechanistic Target Of Rapamycin (mTOR) pathway in FCD type II (FCDII), and SLC35A2 in mild malformations of cortical development (mMCDs) with oligodendroglial hyperplasia (MOGHE). The electro-clinical-imaging phenotypes and surgical outcomes were better defined and validated for FCDII. Little new information was acquired on clinical, histopathological, or genetic characteristics of FCD type I (FCDI) and FCD type III (FCDIII). The survey identified mMCDs, FCDI, and genetic characterization as fields for improvement in an updated classification. Our iterative clinico-pathological and genetic agreement study confirmed the importance of immunohistochemical staining, neuroimaging, and genetic tests to improve the diagnostic yield. The TF proposes to include mMCDs, MOGHE, and "no definite FCD on histopathology" as new categories in the updated FCD classification. The histopathological classification can be further augmented by advanced neuroimaging and genetic studies to comprehensively diagnose FCD subtypes; these different levels should then be integrated into a multi-layered diagnostic scheme. This update may help to foster multidisciplinary efforts toward a better understanding of FCD and the development of novel targeted treatment options.
Collapse
Affiliation(s)
- Imad Najm
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland, Ohio, USA
| | - Dennis Lal
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland, Ohio, USA.,Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Fernando Cendes
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas - UNICAMP, Campinas, Sao Paulo, Brazil.,Department of Neurology, University of Campinas - UNICAMP, Campinas, Sao Paulo, Brazil
| | - Iscia Lopes-Cendes
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas - UNICAMP, Campinas, Sao Paulo, Brazil.,Department of Translational Medicine, University of Campinas - UNICAMP, Campinas, Sao Paulo, Brazil
| | - Andre Palmini
- Department of Clinical Neurosciences, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Porto Alegre Epilepsy Surgery Program, Hospital São Lucas PUCRS, Porto Alegre, Brazil
| | - Eliseu Paglioli
- Department of Surgery, School of Medicine, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Harvey B Sarnat
- Department of Paediatrics, Department of Pathology (Neuropathology) and Department of Clinical Neurosciences, University of Calgary Faculty of Medicine, Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Christopher A Walsh
- Division of Genetics and Genomics and Howard Hughes Medical Institute, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA.,Departments of Pediatrics and Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel Wiebe
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Roland Coras
- Department of Neuropathology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katja Kobow
- Developmental Neurosciences Programme, UCL NIHR BRC Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - J Helen Cross
- Developmental Neurosciences Programme, UCL NIHR BRC Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Rita Garbelli
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Hans Holthausen
- Center for Pediatric Neurology, Neurorehabilitation and Epileptology, Schoen-Clinic, Vogtareuth, Germany
| | - Karl Rössler
- Department of Neurosurgery, Allgemeines Krankenhaus Wien, Vienna Medical University, Wien, Austria
| | - Maria Thom
- Department of Neuropathology, Institute of Neurology, University College London, UK
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Laboratory, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, KAIST and SoVarGen, Daejeon, South Korea
| | - Hajime Miyata
- Department of Neuropathology, Research Institute for Brain and Blood Vessels, Akita Cerebrospinal and Cardiovascular Center, Akita, Japan
| | - Renzo Guerrini
- Neuroscience Department, Children's Hospital Anna Meyer- University of Florence, Florence, Italy
| | - Yue-Shan Piao
- National Center for Neurological Disorders, Department of Pathology, Xuanwu Hospital, Capital Medical University, and Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Ingmar Blümcke
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland, Ohio, USA.,Department of Neuropathology, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
25
|
Billon V, Sanchez-Luque FJ, Rasmussen J, Bodea GO, Gerhardt DJ, Gerdes P, Cheetham SW, Schauer SN, Ajjikuttira P, Meyer TJ, Layman CE, Nevonen KA, Jansz N, Garcia-Perez JL, Richardson SR, Ewing AD, Carbone L, Faulkner GJ. Somatic retrotransposition in the developing rhesus macaque brain. Genome Res 2022; 32:1298-1314. [PMID: 35728967 PMCID: PMC9341517 DOI: 10.1101/gr.276451.121] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/14/2022] [Indexed: 12/03/2022]
Abstract
The retrotransposon LINE-1 (L1) is central to the recent evolutionary history of the human genome and continues to drive genetic diversity and germline pathogenesis. However, the spatiotemporal extent and biological significance of somatic L1 activity are poorly defined and are virtually unexplored in other primates. From a single L1 lineage active at the divergence of apes and Old World monkeys, successive L1 subfamilies have emerged in each descendant primate germline. As revealed by case studies, the presently active human L1 subfamily can also mobilize during embryonic and brain development in vivo. It is unknown whether nonhuman primate L1s can similarly generate somatic insertions in the brain. Here we applied approximately 40× single-cell whole-genome sequencing (scWGS), as well as retrotransposon capture sequencing (RC-seq), to 20 hippocampal neurons from two rhesus macaques (Macaca mulatta). In one animal, we detected and PCR-validated a somatic L1 insertion that generated target site duplications, carried a short 5' transduction, and was present in ∼7% of hippocampal neurons but absent from cerebellum and nonbrain tissues. The corresponding donor L1 allele was exceptionally mobile in vitro and was embedded in PRDM4, a gene expressed throughout development and in neural stem cells. Nanopore long-read methylome and RNA-seq transcriptome analyses indicated young retrotransposon subfamily activation in the early embryo, followed by repression in adult tissues. These data highlight endogenous macaque L1 retrotransposition potential, provide prototypical evidence of L1-mediated somatic mosaicism in a nonhuman primate, and allude to L1 mobility in the brain over the past 30 million years of human evolution.
Collapse
Affiliation(s)
- Victor Billon
- Queensland Brain Institute, University of Queensland, St. Lucia, Queensland 4067, Australia
- Biology Department, École Normale Supérieure Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Francisco J Sanchez-Luque
- GENYO. Pfizer-University of Granada-Andalusian Government Centre for Genomics and Oncological Research, PTS Granada 18016, Spain
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
- Institute of Parasitology and Biomedicine "Lopez-Neyra"-Spanish National Research Council, PTS Granada 18016, Spain
| | - Jay Rasmussen
- Queensland Brain Institute, University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Gabriela O Bodea
- Queensland Brain Institute, University of Queensland, St. Lucia, Queensland 4067, Australia
- Mater Research Institute-University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Daniel J Gerhardt
- Mater Research Institute-University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Patricia Gerdes
- Mater Research Institute-University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Seth W Cheetham
- Mater Research Institute-University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Stephanie N Schauer
- Mater Research Institute-University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Prabha Ajjikuttira
- Queensland Brain Institute, University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Thomas J Meyer
- Division of Genetics, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA
| | - Cora E Layman
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Kimberly A Nevonen
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Natasha Jansz
- Mater Research Institute-University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Jose L Garcia-Perez
- GENYO. Pfizer-University of Granada-Andalusian Government Centre for Genomics and Oncological Research, PTS Granada 18016, Spain
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| | - Sandra R Richardson
- Mater Research Institute-University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Adam D Ewing
- Mater Research Institute-University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Lucia Carbone
- Division of Genetics, Oregon National Primate Research Center, Beaverton, Oregon 97006, USA
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239, USA
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Geoffrey J Faulkner
- Queensland Brain Institute, University of Queensland, St. Lucia, Queensland 4067, Australia
- Mater Research Institute-University of Queensland, Woolloongabba, Queensland 4102, Australia
| |
Collapse
|
26
|
Xu H, Gao K, Liu Q, Wang T, Zhang Z, Cai L, Wu Y, Jiang Y. Brain Somatic Variant in Ras-Like Small GTPase RALA Causes Focal Cortical Dysplasia Type II. Front Behav Neurosci 2022; 16:919485. [PMID: 35846790 PMCID: PMC9280360 DOI: 10.3389/fnbeh.2022.919485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/03/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose In our group’s previous study, we performed deep whole-exome sequencing and targeted amplicon sequencing in the postoperative brain tissue of epilepsy patients with focal cortical dysplasia type II (FCD II). We identified the first somatic variant of RALA in the brain tissue of a child with FCD type IIb. RALA encodes a small GTPase of the Ras superfamily. To date, the role of RALA in brain development is not yet known. In this study, we reported that the RALA somatic variant led to FCD type II through activation of the mammalian target of rapamycin (mTOR) pathways. Materials and Methods HEK293T cells were transfected in vitro to analyze the expression of the RalA protein, as well as phosphorylated S6 (P-S6), one of the major markers of mTOR pathway activation, RalA GTPase activity, and the interaction between RalA and its downstream binding effectors. In vivo, wild-type, and mutant RALA plasmids were transfected into the local cortex of mice using in utero electroporation to evaluate the effect of RALA c.G482A on neuronal migration. Results The RALA c.G482A mutation increased RalA protein expression, the abnormal activation of the mTOR pathways, RalA GTPase activity, and binding to downstream effectors. RALA c.G482A local transfection in the embryonic brain in utero induced abnormal cortical neuron migration in mice. Conclusion This study demonstrated for the first time that the somatic gain-of-function variant of RALA activates the mTOR pathway and leads to neuronal migration disorders in the brain, facilitating the development of FCD II. Therefore, RALA brain somatic mutation may be one of the pathogenic mechanisms leading to FCD II, which is always related to drug-resistant epilepsy in children. However, more somatic variations of this gene are required to be confirmed in more FCD II patient brain samples.
Collapse
Affiliation(s)
- Han Xu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Qingzhu Liu
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Tianshuang Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Zhongbin Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Lixin Cai
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
- *Correspondence: Ye Wu,
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
- Institute for Brain Disorders, Beijing, China
- Yuwu Jiang, ,
| |
Collapse
|
27
|
Lee S, Lee JH. Brain somatic mutations as RNA therapeutic targets in neurological disorders. Ann N Y Acad Sci 2022; 1514:11-20. [PMID: 35527236 DOI: 10.1111/nyas.14786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Research into the genetic etiology of a neurological disorder can provide directions for genetic diagnosis and targeted therapy. In the past, germline mutations, which are transmitted from parents or newly arise from parental germ cells, were considered as major genetic causes of neurological disorders. However, recent evidence has shown that somatic mutations in the brain, which can arise from neural stem cells during development or over aging, account for a significant number of brain disorders, ranging from neurodevelopmental, neurodegenerative, and neuropsychiatric to neoplastic disease. Moreover, the identification of disease-causing somatic mutations or mutated genes has provided new insights into molecular pathogenesis and unveiled potential therapeutic targets for treating neurological disorders that have few, or no, therapeutic options. RNA therapeutics, including antisense oligonucleotide (ASO) and small interfering RNA (siRNA), are emerging as promising therapeutic tools for treating genetic neurological disorders. As the number of approved and investigational ASO and siRNA drugs for neurological disorders associated with germline mutations increases, they may also prove to be attractive modalities for treating neurologic disorders resulting from somatic mutations. In this perspective, we highlight several neurological diseases caused by brain somatic mutations and discuss the potential role of RNA therapeutics in these conditions.
Collapse
Affiliation(s)
- Sungyul Lee
- SoVarGen Co., Ltd., Daejeon, Republic of Korea
| | - Jeong Ho Lee
- SoVarGen Co., Ltd., Daejeon, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute Science and Technology (KAIST), KAIST BioMedical Research Center, Daejeon, Republic of Korea
| |
Collapse
|
28
|
Saida K, Chong PF, Yamaguchi A, Saito N, Ikehara H, Koshimizu E, Miyata R, Ishiko A, Nakamura K, Ohnishi H, Fujioka K, Sakakibara T, Asada H, Ogawa K, Kudo K, Ohashi E, Kawai M, Abe Y, Tsuchida N, Uchiyama Y, Hamanaka K, Fujita A, Mizuguchi T, Miyatake S, Miyake N, Kato M, Kira R, Matsumoto N. Monogenic causes of pigmentary mosaicism. Hum Genet 2022; 141:1771-1784. [PMID: 35503477 DOI: 10.1007/s00439-022-02437-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/28/2022] [Indexed: 12/30/2022]
Abstract
Pigmentary mosaicism of the Ito type, also known as hypomelanosis of Ito, is a neurocutaneous syndrome considered to be predominantly caused by somatic chromosomal mosaicism. However, a few monogenic causes of pigmentary mosaicism have been recently reported. Eleven unrelated individuals with pigmentary mosaicism (mostly hypopigmented skin) were recruited for this study. Skin punch biopsies of the probands and trio-based blood samples (from probands and both biological parents) were collected, and genomic DNA was extracted and analyzed by exome sequencing. In all patients, plausible monogenic causes were detected with somatic and germline variants identified in five and six patients, respectively. Among the somatic variants, four patients had MTOR variant (36%) and another had an RHOA variant. De novo germline variants in USP9X, TFE3, and KCNQ5 were detected in two, one, and one patients, respectively. A maternally inherited PHF6 variant was detected in one patient with hyperpigmented skin. Compound heterozygous GTF3C5 variants were highlighted as strong candidates in the remaining patient. Exome sequencing, using patients' blood and skin samples is highly recommended as the first choice for detecting causative genetic variants of pigmentary mosaicism.
Collapse
Affiliation(s)
- Ken Saida
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Pin Fee Chong
- Department of Pediatric Neurology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Asuka Yamaguchi
- Department of Pediatrics, Tokyo-Kita Medical Center, Tokyo, Japan
| | - Naka Saito
- Department of Pediatrics, Tsuruoka Municipal Shonai Hospital, Yamagata, Japan
| | - Hajime Ikehara
- Department of Pediatrics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Eriko Koshimizu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Rie Miyata
- Department of Pediatrics, Tokyo-Kita Medical Center, Tokyo, Japan
| | - Akira Ishiko
- Department of Dermatology, Toho University School of Medicine, Tokyo, Japan
| | - Kazuyuki Nakamura
- Department of Pediatrics, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Hidenori Ohnishi
- Department of Pediatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kei Fujioka
- Center of General Internal Medicine and Rheumatology, Gifu Municipal Hospital, Gifu, Japan
| | - Takafumi Sakakibara
- Department of Pediatrics, Nara Medical University School of Medicine, Nara, Japan
| | - Hideo Asada
- Department of Dermatology, Nara Medical University School of Medicine, Nara, Japan
| | - Kohei Ogawa
- Department of Dermatology, Nara Medical University School of Medicine, Nara, Japan
| | - Kyoko Kudo
- Department of Dermatology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Eri Ohashi
- Division of Neurology, National Center for Child Health and Development, Tokyo, Japan
| | - Michiko Kawai
- Division of Neurology, National Center for Child Health and Development, Tokyo, Japan
| | - Yuichi Abe
- Division of Neurology, National Center for Child Health and Development, Tokyo, Japan
| | - Naomi Tsuchida
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.,Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan
| | - Yuri Uchiyama
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.,Department of Rare Disease Genomics, Yokohama City University Hospital, Yokohama, Japan
| | - Kohei Hamanaka
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Atsushi Fujita
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Takeshi Mizuguchi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.,Clinical Genetics Department, Yokohama City University Hospital, Yokohama, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.,Department of Human Genetics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Ryutaro Kira
- Department of Pediatric Neurology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
29
|
Sakai K, Ishida C, Hayashi K, Tsuji N, Kannon T, Hosomichi K, Takei N, Kakita A, Tajima A, Yamada M. Familial idiopathic basal ganglia calcification with a heterozygous missense variant (c.902C>T/p.P307L) in SLC20A2 showing widespread cerebrovascular lesions. Neuropathology 2022; 42:126-133. [PMID: 35026865 DOI: 10.1111/neup.12781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 12/01/2022]
Abstract
We describe a postmortem case of familial idiopathic basal ganglia calcification (FIBGC) in a 72-year-old Japanese man. The patient showed progressive cognitive impairment with a seven-year clinical course and calcification of the basal ganglia, thalami, and cerebellar dentate nuclei. A novel heterozygous missense variant in SLC20A2 (c.920C>T/p.P307L), a type III sodium-dependent phosphate transporter (PiT-2), was subsequently identified, in addition to typical neuropathological findings of FIBGC, such as capillary calcification of the occipital gray matter, confluent calcification of the basal ganglia and cerebellar white matter, widespread occurrence of vasculopathic changes, cerebrovascular lesions, and vascular smooth muscle cell depletion. Immunohistochemistry for PiT-2 protein revealed no apparent staining in endothelial cells in the basal ganglia and insular cortex; however, the immunoreactivity in endothelial cells of the cerebellum was preserved. Moreover, Western blot analysis identified preserved PiT-2 immunoreactivity signals in the frontal cortex and cerebellum. The variant identified in the present patient could be associated with development of FIBGC and is known to be located at the large intracytoplasmic part of the PiT-2 protein, which has potential phosphorylation sites with importance in the regulation of inorganic phosphate transport activity. The present case is an important example to prove that FIGBC could stem from a missense variant in the large intracytoplasmic loop of the PiT-2 protein. Abnormal clearance of inorganic phosphate in the brain could be related to the development of vascular smooth muscle damage, the formation of cerebrovascular lesions, and subsequent brain calcification in patients with FIBGC with SLC20A2 variants.
Collapse
Affiliation(s)
- Kenji Sakai
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Chiho Ishida
- Department of Neurology, National Hospital Organization Iou National Hospital, Kanazawa, Japan
| | - Koji Hayashi
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
- Department of Neurology, National Hospital Organization Iou National Hospital, Kanazawa, Japan
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Naotaka Tsuji
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Takayuki Kannon
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kazuyoshi Hosomichi
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Nobuyuki Takei
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Masahito Yamada
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
30
|
Jha R, Kurup A, Kovilapu UB, Ranjan R, Sondhi V. Somatic mutations involving TSC 1 and TSC2 genes in two children with focal cortical dysplasia. Brain Dev 2022; 44:166-172. [PMID: 34756499 DOI: 10.1016/j.braindev.2021.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/21/2021] [Accepted: 10/07/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND The role of PI3K/AKT/mTOR pathway hyperactivation in localized brain overgrowth is evolving. We describe two patients with focal cortical dysplasia (FCD) who demonstrated somatic mutations in TSC1 and TSC2 genes in the dysplastic brain tissue but not peripheral blood. METHODS Paired whole-exome sequencing was performed on genomic DNA extracted from blood and excised brain tissue in two children with FCD who underwent excision of dysplastic tissue. RESULTS Patient 1, a 14-year boy, had drug-resistant focal epilepsy with onset at 20 months. His brain MRI showed abnormalities suggestive of FCD in the left superior and middle frontal lobes. Patient 2 presented at the age of 10 years with pharmaco-resistant focal epilepsy (onset at six years). His MRI suggested FCD in the left insular lobe. Both patients underwent surgical excision of FCD, and excised tissues were pathologically confirmed to have type IIb FCD. For patient 1, a missense mutation (c.64C > T; p.Arg22Trp) was detected in the TSC1 gene in DNA of dysplastic brain tissue but not peripheral blood lymphocytes. Similarly, for patient 2, a frameshift mutation (c.4258_4261delCAGT; p.Ser1420GlyfsTer55) in the TSC2 gene was identified in the brain tissue but not blood. Both gene variants are likely pathogenic and cause mTOR pathway activation. CONCLUSION Our report of TSC1/TSC2 somatic mutations in patients with non-syndromic FCD suggests that localized hyperactivation of the mTOR pathway can cause focal malformations during cortical development and presents pharmacological targets for precision therapy in FCD management.
Collapse
Affiliation(s)
- Ruchika Jha
- Department of Pediatrics, Armed Forces Medical College, Pune, India
| | - Arjun Kurup
- Department of Pediatrics, Armed Forces Medical College, Pune, India
| | - U B Kovilapu
- Department of Radiodiagnosis, Armed Forces Medical College, Pune, India
| | - Rakesh Ranjan
- Department of Neurosurgery, Aditya Birla Memorial Hospital, Pune, India
| | - Vishal Sondhi
- Department of Pediatrics, Armed Forces Medical College, Pune, India.
| |
Collapse
|
31
|
Ghosh C, Myers R, O'Connor C, Williams S, Liu X, Hossain M, Nemeth M, Najm IM. Cortical Dysplasia in Rats Provokes Neurovascular Alterations, GLUT1 Dysfunction, and Metabolic Disturbances That Are Sustained Post-Seizure Induction. Mol Neurobiol 2022; 59:2389-2406. [PMID: 35084654 PMCID: PMC9018620 DOI: 10.1007/s12035-021-02624-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/26/2021] [Indexed: 10/19/2022]
Abstract
Focal cortical dysplasia (FCD) is associated with blood-brain barrier (BBB) dysfunction in patients with difficult-to-treat epilepsy. However, the underlying cellular and molecular factors in cortical dysplasia (CD) associated with progressive neurovascular challenges during the pro-epileptic phase, post-seizure, and during epileptogenesis remain unclear. We studied the BBB function in a rat model of congenital (in utero radiation-induced, first hit) CD and longitudinally examined the cortical brain tissues at baseline and the progressive neurovascular alterations, glucose transporter-1 (GLUT1) expression, and glucose metabolic activity at 2, 15, and 30 days following a second hit using pentylenetetrazole-induced seizure. Our study revealed through immunoblotting, immunohistochemistry, and biochemical analysis that (1) altered vascular density and prolongation of BBB albumin leakages in CD rats continued through 30 days post-seizure; (2) CD brain tissues showed elevated matrix metalloproteinase-9 levels at 2 days post-seizure and microglial overactivation through 30 days post-seizure; (3) BBB tight junction protein and GLUT1 levels were decreased and neuronal monocarboxylate transporter-2 (MCT2) and mammalian target of rapamycin (mTOR) levels were increased in the CD rat brain: (4) ATPase activity is elevated and a low glucose/high lactate imbalance exists in CD rats; and (5) the mTOR pathway is activated and MCT2 levels are elevated in the presence of high lactate during glucose starvation in vitro. Together, this study suggests that BBB dysfunction, including decreased GLUT1 expression and metabolic disturbance, may contribute to epileptogenesis in this CD rat model through multiple mechanisms that could be translated to FCD therapy in medically refractory epilepsy.
Collapse
Affiliation(s)
- Chaitali Ghosh
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA. .,Department of Biomedical Engineering and Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
| | - Rosemary Myers
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Christina O'Connor
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sherice Williams
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Xuefeng Liu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mohammed Hossain
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Michael Nemeth
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Imad M Najm
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
32
|
Lee WS, Baldassari S, Stephenson SEM, Lockhart PJ, Baulac S, Leventer RJ. Cortical Dysplasia and the mTOR Pathway: How the Study of Human Brain Tissue Has Led to Insights into Epileptogenesis. Int J Mol Sci 2022; 23:1344. [PMID: 35163267 PMCID: PMC8835853 DOI: 10.3390/ijms23031344] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 02/01/2023] Open
Abstract
Type II focal cortical dysplasia (FCD) is a neuropathological entity characterised by cortical dyslamination with the presence of dysmorphic neurons only (FCDIIA) or the presence of both dysmorphic neurons and balloon cells (FCDIIB). The year 2021 marks the 50th anniversary of the recognition of FCD as a cause of drug resistant epilepsy, and it is now the most common reason for epilepsy surgery. The causes of FCD remained unknown until relatively recently. The study of resected human FCD tissue using novel genomic technologies has led to remarkable advances in understanding the genetic basis of FCD. Mechanistic parallels have emerged between these non-neoplastic lesions and neoplastic disorders of cell growth and differentiation, especially through perturbations of the mammalian target of rapamycin (mTOR) signalling pathway. This narrative review presents the advances through which the aetiology of FCDII has been elucidated in chronological order, from recognition of an association between FCD and the mTOR pathway to the identification of somatic mosaicism within FCD tissue. We discuss the role of a two-hit mechanism, highlight current challenges and future directions in detecting somatic mosaicism in brain and discuss how knowledge of FCD may inform novel precision treatments of these focal epileptogenic malformations of human cortical development.
Collapse
Affiliation(s)
- Wei Shern Lee
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Parkville 3052, Australia; (W.S.L.); (S.E.M.S.); (P.J.L.)
- Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - Sara Baldassari
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, F-75013 Paris, France;
| | - Sarah E. M. Stephenson
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Parkville 3052, Australia; (W.S.L.); (S.E.M.S.); (P.J.L.)
- Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - Paul J. Lockhart
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Parkville 3052, Australia; (W.S.L.); (S.E.M.S.); (P.J.L.)
- Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
| | - Stéphanie Baulac
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, F-75013 Paris, France;
| | - Richard J. Leventer
- Department of Paediatrics, The University of Melbourne, Parkville 3052, Australia
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- Department of Neurology, The Royal Children’s Hospital, Parkville 3052, Australia
| |
Collapse
|
33
|
Kato M, Kada A, Shiraishi H, Tohyama J, Nakagawa E, Takahashi Y, Akiyama T, Kakita A, Miyake N, Fujita A, Saito AM, Inoue Y. Sirolimus for epileptic seizures associated with focal cortical dysplasia type II. Ann Clin Transl Neurol 2022; 9:181-192. [PMID: 35040598 PMCID: PMC8862414 DOI: 10.1002/acn3.51505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/21/2021] [Accepted: 12/28/2021] [Indexed: 01/16/2023] Open
Abstract
Objective To determine whether sirolimus, a mechanistic target of rapamycin (mTOR) inhibitor, reduces epileptic seizures associated with focal cortical dysplasia (FCD) type II. Methods Sixteen patients (aged 6–57 years) with FCD type II received sirolimus at an initial dose of 1 or 2 mg/day based on body weight (FCDS‐01). In 15 patients, the dose was adjusted to achieve target trough ranges of 5–15 ng/mL, followed by a 12‐week maintenance therapy period. The primary endpoint was a lower focal seizure frequency during the maintenance therapy period. Further, we also conducted a prospective cohort study (RES‐FCD) in which 60 patients with FCD type II were included as an external control group. Results The focal seizure frequency reduced by 25% in all patients during the maintenance therapy period and by a median value of 17%, 28%, and 23% during the 1–4‐, 5–8‐, and 9–12‐week periods. The response rate was 33%. The focal seizure frequency in the external control group reduced by 0.5%. However, the background characteristics of external and sirolimus‐treated groups differed. Adverse events were consistent with those of mTOR inhibitors reported previously. The blood KL‐6 level was elevated over time. Interpretation The reduction of focal seizures did not meet the predetermined level of statistical significance. The safety profile of the drug was tolerable. The potential for a reduction of focal seizures over time merit further investigations.
Collapse
Affiliation(s)
- Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Akiko Kada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Hideaki Shiraishi
- Department of Pediatrics, Hokkaido University Hospital, Sapporo, Japan
| | - Jun Tohyama
- Department of Child Neurology, National Hospital Organization Nishiniigata Chuo Hospital, Niigata, Japan
| | - Eiji Nakagawa
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yukitoshi Takahashi
- National Hospital Organization, Shizuoka Institute of Epilepsy and Neurological Disorders, Shizuoka, Japan
| | - Tomoyuki Akiyama
- Department of Child Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Department of Human Genetics, Research Institute National Center for Global Health and Medicine, Tokyo, Japan
| | - Atsushi Fujita
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akiko M Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Yushi Inoue
- National Hospital Organization, Shizuoka Institute of Epilepsy and Neurological Disorders, Shizuoka, Japan
| |
Collapse
|
34
|
Coleman N, Subbiah V, Pant S, Patel K, Roy-Chowdhuri S, Yedururi S, Johnson A, Yap TA, Rodon J, Shaw K, Meric-Bernstam F. Emergence of mTOR mutation as an acquired resistance mechanism to AKT inhibition, and subsequent response to mTORC1/2 inhibition. NPJ Precis Oncol 2021; 5:99. [PMID: 34853384 PMCID: PMC8636467 DOI: 10.1038/s41698-021-00240-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/09/2021] [Indexed: 01/14/2023] Open
Abstract
Acquired resistance to molecular targeted therapy is a significant challenge of the precision medicine era. The ability to understand these mechanisms of resistance may improve patient selection and allow for the development of rationally designed next-line or combination treatment strategies and improved patient outcomes. AKT is a critical effector of the phosphoinositide 3-kinase signaling cascade, one of the most commonly activated pathways in human cancer. Deregulation of signaling pathways, such as RAF/MEK/ERK are previously described mechanisms of resistance to AKT/PI3K inhibitors. Mutations in the mTOR gene, however, are exceedingly rare. We present a case of acquired mTOR resistance, following targeted AKT inhibition, and subsequent response to mTOR1/2 inhibitor in a patient with metastatic endometrial cancer, the first documented response to ATP-competitive mTOR inhibition in this setting. This case supports mTOR mutation as a mechanism of resistance, and underscores the importance of tumor molecular profiling, exemplifying precision medicine in action.
Collapse
Affiliation(s)
- Niamh Coleman
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Shubham Pant
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Keyur Patel
- Khalifa Institute for Personalized Cancer Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Sireesha Yedururi
- Abdominal Imaging Department, MD Anderson Cancer Center, Houston, TX, USA
| | - Amber Johnson
- Khalifa Institute for Personalized Cancer Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
- Khalifa Institute for Personalized Cancer Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Kenna Shaw
- Khalifa Institute for Personalized Cancer Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
- Khalifa Institute for Personalized Cancer Therapy, MD Anderson Cancer Center, Houston, TX, USA.
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
35
|
Neocortical development and epilepsy: insights from focal cortical dysplasia and brain tumours. Lancet Neurol 2021; 20:943-955. [PMID: 34687638 DOI: 10.1016/s1474-4422(21)00265-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/14/2021] [Accepted: 08/05/2021] [Indexed: 01/16/2023]
Abstract
During the past decade, there have been considerable advances in understanding of the genetic and morphogenic processes underlying cortical malformations and developmental brain tumours. Focal malformations are caused by somatic (postzygotic) variants in genes related to cell growth (ie, in the mTOR pathway in focal cortical dysplasia type 2), which are acquired in neuronal progenitors during neurodevelopment. In comparison, developmental brain tumours result from somatic variants in genes related to cell proliferation (eg, in the MAP-kinase pathway in ganglioglioma), which affect proliferating glioneuronal precursors. The timing of the genetic event and the specific gene involved during neurodevelopment will drive the nature and size of the lesion, whether it is a developmental malformation or a brain tumour. There is also emerging evidence that epigenetic processes underlie a molecular memory in epileptogenesis. This knowledge will together facilitate understanding of why and how patients with these lesions have epilepsy, and could form a basis for a move towards precision medicine for this challenging cohort of patients.
Collapse
|
36
|
Marashly A, Karia S, Zonjy B. Epilepsy Surgery: Special Circumstances. Semin Pediatr Neurol 2021; 39:100921. [PMID: 34620459 DOI: 10.1016/j.spen.2021.100921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 11/25/2022]
Abstract
Epilepsy surgery has proven to be very effective in treating refractory focal epilepsies in children, producing seizure freedom or partial seizure control well beyond any other medical or dietary therapies. While surgery is mostly utilized in certain clinical phenotypes, either based on the location such as temporal lobe epilepsy, or based on the presence of known epileptogenic lesions such as focal cortical dysplasia, tumors or hemimegalencephaly, there is a growing body of evidence to support the role of surgery in other patients' cohorts that were classically not thought of as surgical candidates. These include patients with rare genetic disorders, electrical status epilepticus in sleep, status epilepticus and the very young patients. Furthermore, epilepsy surgery is not considered as a "last resort" as seizure and cognitive outcomes of surgery are considerably better when done earlier rather than later in relation to the time of onset of epilepsy and age of surgery especially in the context of known focal cortical dysplasia. This article examines the accumulating evidence of the utility of epilepsy surgery in these special circumstances.
Collapse
Affiliation(s)
- Ahmad Marashly
- Assistant Professor, University of Washington/Seattle Children's Hospital, Seattle, WA.
| | - Samir Karia
- Associate Professor, Univeristy of Louisville, Luisiville, KY
| | - Bilal Zonjy
- Assistant Professor, University of Washington/Seattle Children's Hospital, Seattle, WA
| |
Collapse
|
37
|
Dimartino P, Mariani V, Marconi C, Minardi R, Bramerio M, Licchetta L, Menghi V, Morandi L, Magini P, Mongelli P, Cardinale F, Seri M, Tinuper P, Tassi L, Pippucci T, Bisulli F. Accurate Detection of Hot-Spot MTOR Somatic Mutations in Archival Surgical Specimens of Focal Cortical Dysplasia by Molecular Inversion Probes. Mol Diagn Ther 2021; 24:571-577. [PMID: 32772316 DOI: 10.1007/s40291-020-00488-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Formalin-fixed, paraffin-embedded brain specimens are a potentially rich resource to identify somatic variants, but their DNA is characterised by low yield and extensive degradation, and matched peripheral samples are usually unavailable for analysis. METHODS We designed single-molecule molecular inversion probes to target 18 MTOR somatic mutational hot-spots in unmatched, histologically proven focal cortical dysplasias from formalin-fixed, paraffin-embedded tissues of 50 patients. RESULTS We achieved adequate DNA and sequencing quality in 28 focal cortical dysplasias, mostly extracted within 2 years from fixation, showing a statistically significant effect of time from fixation as a major determinant for successful genetic analysis. We identified and validated seven encompassing hot-spot residues (found in 14% of all patients and in 25% of those sequenced and analysed). The allele fraction had a range of 2-5% and variants were absent in available neighbouring non-focal cortical dysplasia specimens. We computed an alternate allele threshold for calling true variants, based on an experiment-wise mismatch count distribution, well predicting call reliability. CONCLUSIONS Single-molecule molecular inversion probes are experimentally simple, cost effective and scalable, accurately detecting clinically relevant somatic variants in challenging brain formalin-fixed, paraffin-embedded tissues.
Collapse
Affiliation(s)
- Paola Dimartino
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Valeria Mariani
- "Claudio Munari" Epilepsy Surgery Center, Niguarda Hospital, Milan, Italy
| | - Caterina Marconi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Raffaella Minardi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Epilepsy Center (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy
| | | | - Laura Licchetta
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Epilepsy Center (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Veronica Menghi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Epilepsy Center (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Luca Morandi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Epilepsy Center (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy.,Functional MR Unit, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Pamela Magini
- Unità Operativa di Genetica Medica, Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138, Bologna, Italy
| | - Patrizia Mongelli
- Unità Operativa di Genetica Medica, Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138, Bologna, Italy
| | | | - Marco Seri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.,Unità Operativa di Genetica Medica, Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138, Bologna, Italy
| | - Paolo Tinuper
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Epilepsy Center (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Laura Tassi
- "Claudio Munari" Epilepsy Surgery Center, Niguarda Hospital, Milan, Italy
| | - Tommaso Pippucci
- Unità Operativa di Genetica Medica, Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138, Bologna, Italy.
| | - Francesca Bisulli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Epilepsy Center (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| |
Collapse
|
38
|
Szczałuba K, Rydzanicz M, Walczak A, Kosińska J, Koppolu A, Biernacka A, Iwanicka-Pronicka K, Grajkowska W, Jurkiewicz E, Kowalczyk P, Płoski R. Brain Tissue Low-Level Mosaicism for MTOR Mutation Causes Smith-Kingsmore Phenotype with Recurrent Hypoglycemia-A Novel Phenotype and a Further Proof for Testing of an Affected Tissue. Diagnostics (Basel) 2021; 11:diagnostics11071269. [PMID: 34359351 PMCID: PMC8303645 DOI: 10.3390/diagnostics11071269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 11/16/2022] Open
Abstract
De novo somatic variants in genes encoding components of the PI3K-AKT3-mTOR pathway, including MTOR, have been linked to hemimegalencephaly or focal cortical dysplasia. Similarly to other malformations of cortical development, this condition presents with developmental delay and intractable epilepsy, often necessitating surgical treatment. We describe a first patient with the Smith-Kingsmore syndrome phenotype with recurrent hypoglycemia caused by low-level mosaic MTOR mutation restricted to the brain. We provide discussion on different aspects of somatic mosaicism. Deep exome sequencing combined with a variant search in multiple tissues and careful phenotyping may constitute a key to the diagnosis of the causes of rare brain anomalies.
Collapse
Affiliation(s)
- Krzysztof Szczałuba
- Department of Medical Genetics, Medical University of Warsaw, Pawinskiego 3c Str., 02-106 Warsaw, Poland; (M.R.); (A.W.); (J.K.); (A.K.); (A.B.)
- Correspondence: (K.S.); (R.P.); Tel.: +48-22-5720-695 (K.S. & R.P.); Fax: +48-22-5720-696 (K.S. & R.P.)
| | - Małgorzata Rydzanicz
- Department of Medical Genetics, Medical University of Warsaw, Pawinskiego 3c Str., 02-106 Warsaw, Poland; (M.R.); (A.W.); (J.K.); (A.K.); (A.B.)
| | - Anna Walczak
- Department of Medical Genetics, Medical University of Warsaw, Pawinskiego 3c Str., 02-106 Warsaw, Poland; (M.R.); (A.W.); (J.K.); (A.K.); (A.B.)
| | - Joanna Kosińska
- Department of Medical Genetics, Medical University of Warsaw, Pawinskiego 3c Str., 02-106 Warsaw, Poland; (M.R.); (A.W.); (J.K.); (A.K.); (A.B.)
| | - Agnieszka Koppolu
- Department of Medical Genetics, Medical University of Warsaw, Pawinskiego 3c Str., 02-106 Warsaw, Poland; (M.R.); (A.W.); (J.K.); (A.K.); (A.B.)
| | - Anna Biernacka
- Department of Medical Genetics, Medical University of Warsaw, Pawinskiego 3c Str., 02-106 Warsaw, Poland; (M.R.); (A.W.); (J.K.); (A.K.); (A.B.)
| | | | - Wiesława Grajkowska
- Department of Pathology, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
| | - Elżbieta Jurkiewicz
- Department of Diagnostic Imaging, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
| | - Paweł Kowalczyk
- Department of Neurosurgery, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
| | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, Pawinskiego 3c Str., 02-106 Warsaw, Poland; (M.R.); (A.W.); (J.K.); (A.K.); (A.B.)
- Correspondence: (K.S.); (R.P.); Tel.: +48-22-5720-695 (K.S. & R.P.); Fax: +48-22-5720-696 (K.S. & R.P.)
| |
Collapse
|
39
|
Functional and structural analyses of novel Smith-Kingsmore Syndrome-Associated MTOR variants reveal potential new mechanisms and predictors of pathogenicity. PLoS Genet 2021; 17:e1009651. [PMID: 34197453 PMCID: PMC8279410 DOI: 10.1371/journal.pgen.1009651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/14/2021] [Accepted: 06/08/2021] [Indexed: 12/31/2022] Open
Abstract
Smith-Kingsmore syndrome (SKS) is a rare neurodevelopmental disorder characterized by macrocephaly/megalencephaly, developmental delay, intellectual disability, hypotonia, and seizures. It is caused by dominant missense mutations in MTOR. The pathogenicity of novel variants in MTOR in patients with neurodevelopmental disorders can be difficult to determine and the mechanism by which variants cause disease remains poorly understood. We report 7 patients with SKS with 4 novel MTOR variants and describe their phenotypes. We perform in vitro functional analyses to confirm MTOR activation and interrogate disease mechanisms. We complete structural analyses to understand the 3D properties of pathogenic variants. We examine the accuracy of relative accessible surface area, a quantitative measure of amino acid side-chain accessibility, as a predictor of MTOR variant pathogenicity. We describe novel clinical features of patients with SKS. We confirm MTOR Complex 1 activation and identify MTOR Complex 2 activation as a new potential mechanism of disease in SKS. We find that pathogenic MTOR variants disproportionately cluster in hotspots in the core of the protein, where they disrupt alpha helix packing due to the insertion of bulky amino acid side chains. We find that relative accessible surface area is significantly lower for SKS-associated variants compared to benign variants. We expand the phenotype of SKS and demonstrate that additional pathways of activation may contribute to disease. Incorporating 3D properties of MTOR variants may help in pathogenicity classification. We hope these findings may contribute to improving the precision of care and therapeutic development for individuals with SKS. Smith-Kingsmore Syndrome is a rare disease caused by damage in a gene named MTOR that is associated with excessive growth of the head and brain, delays in development and deficits in intellectual functioning. We report 7 patients who have changes in MTOR that have never been reported before. We describe new medical findings in these patients that may be common in Smith-Kingsmore Syndrome more broadly. We then identify how these new gene changes impact the function of the MTOR protein and thus cell function downstream. Lastly, we show that changes in the gene that lie deep inside the 3D structure of the MTOR protein are more likely to cause disease than those changes that lie on the surface of the protein. We may be able to use the 3D properties of MTOR gene changes to predict if future changes we see are likely to cause disease or not.
Collapse
|
40
|
Khoshkhoo S, Lal D, Walsh CA. Application of single cell genomics to focal epilepsies: A call to action. Brain Pathol 2021; 31:e12958. [PMID: 34196990 PMCID: PMC8412079 DOI: 10.1111/bpa.12958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/24/2022] Open
Abstract
Focal epilepsies are the largest epilepsy subtype and associated with significant morbidity. Somatic variation is a newly recognized genetic mechanism underlying a subset of focal epilepsies, but little is known about the processes through which somatic mosaicism causes seizures, the cell types carrying the pathogenic variants, or their developmental origin. Meanwhile, the inception of single cell biology has completely revolutionized the study of neurological diseases and has the potential to answer some of these key questions. Focusing on single cell genomics, transcriptomics, and epigenomics in focal epilepsy research, circumvents the averaging artifact associated with studying bulk brain tissue and offers the kind of granularity that is needed for investigating the consequences of somatic mosaicism. Here we have provided a brief overview of some of the most developed single cell techniques and the major considerations around applying them to focal epilepsy research.
Collapse
Affiliation(s)
- Sattar Khoshkhoo
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA.,Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.,Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.,Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dennis Lal
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Cologne Center for Genomics, University of Cologne, Cologne, Germany.,Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.,Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.,Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
41
|
The Role of KRAS Mutations in Cortical Malformation and Epilepsy Surgery: A Novel Report of Nevus Sebaceous Syndrome and Review of the Literature. Brain Sci 2021; 11:brainsci11060793. [PMID: 34208656 PMCID: PMC8234150 DOI: 10.3390/brainsci11060793] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/27/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
The rare nevus sebaceous (NS) syndrome (NSS) includes cortical malformations and drug-resistant epilepsy. Somatic RAS-pathway genetic variants are pathogenetic in NS, but not yet described within the brain of patients with NSS. We report on a 5-year-old boy with mild psychomotor delay. A brown-yellow linear skin lesion suggestive of NS in the left temporo-occipital area was evident at birth. Epileptic spasms presented at aged six months. EEG showed continuous left temporo-occipital epileptiform abnormalities. Brain MRI revealed a similarly located diffuse cortical malformation with temporal pole volume reduction and a small hippocampus. We performed a left temporo-occipital resection with histopathological diagnosis of focal cortical dysplasia type Ia in the occipital region and hippocampal sclerosis type 1. Three years after surgery, he is seizure-and drug-free (Engel class Ia) and showed cognitive improvement. Genetic examination of brain and skin specimens revealed the c.35G > T (p.Gly12Val) KRAS somatic missense mutation. Literature review suggests epilepsy surgery in patients with NSS is highly efficacious, with 73% probability of seizure freedom. The few histological analyses reported evidenced disorganized cortex, occasionally with cytomegalic neurons. This is the first reported association of a KRAS genetic variant with cortical malformations associated with epilepsy, and suggests a possible genetic substrate for hippocampal sclerosis.
Collapse
|
42
|
Kada A, Tohyama J, Shiraishi H, Takahashi Y, Nakagawa E, Akiyama T, Saito AM, Inoue Y, Kato M. A Single-Arm Open-Label Clinical Trial on the Efficacy and Safety of Sirolimus for Epileptic Seizures Associated with Focal Cortical Dysplasia Type II: A Study Protocol. Kurume Med J 2021; 66:115-120. [PMID: 34135202 DOI: 10.2739/kurumemedj.ms662007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Epileptic seizures are core symptoms in focal cortical dysplasia (FCD), a disease that often develops in infancy. Such seizures are refractory to conventional antiepileptic drugs (AED) and temporarily disappear in response to AED in only 17% of patients. Currently, surgical resection is an important option for the treatment of epileptic seizures in FCD. In 2015, Korean and Japanese groups independently reported that FCD is caused by somatic mosaic mutation of the MTOR gene in the brain tissue. Based on these results we decided to test a novel treatment using sirolimus, an mTOR inhibitor, for epileptic seizures in patients with FCD type II. A single arm open-label clinical trial for FCD type II patients is being conducted in order to evaluate the efficacy and safety of sirolimus. The dose of sirolimus is fixed for the first 4 weeks and dose adjustment is achieved to maintain a blood level of 5 to 15 ng/mL during 8 to 24 weeks after initiation of administration, and it is kept within this level during a maintenance therapy period of 12 weeks. Primary endpoint is a reduction in the rate of incidence of focal seizures (including focal to bilateral tonic-clonic seizures) per 28 days during the maintenance therapy period from the observation period. To evaluate the frequency of epileptic seizures, registry data will be used as an external control group. We hope that the results of this trial will lead to future innovative treatments for FCD type II patients.
Collapse
Affiliation(s)
- Akiko Kada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center
| | - Jun Tohyama
- Department of Child Neurology, National Hospital Organization Nishi-Niigata Chuo National Hospital
| | | | - Yukitoshi Takahashi
- Department of Pediatrics, National Epilepsy Center, National Hospital Organization Shizuoka Institute of Epilepsy and Neurological Disorders
| | - Eiji Nakagawa
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry
| | - Tomoyuki Akiyama
- Department of Child Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Akiko M Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center
| | - Yushi Inoue
- National Epilepsy Center, National Hospital Organization Shizuoka Institute of Epilepsy and Neurological Disorders
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine
| |
Collapse
|
43
|
Nguyen LH, Bordey A. Convergent and Divergent Mechanisms of Epileptogenesis in mTORopathies. Front Neuroanat 2021; 15:664695. [PMID: 33897381 PMCID: PMC8064518 DOI: 10.3389/fnana.2021.664695] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/17/2021] [Indexed: 12/30/2022] Open
Abstract
Hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1) due to mutations in genes along the PI3K-mTOR pathway and the GATOR1 complex causes a spectrum of neurodevelopmental disorders (termed mTORopathies) associated with malformation of cortical development and intractable epilepsy. Despite these gene variants’ converging impact on mTORC1 activity, emerging findings suggest that these variants contribute to epilepsy through both mTORC1-dependent and -independent mechanisms. Here, we review the literature on in utero electroporation-based animal models of mTORopathies, which recapitulate the brain mosaic pattern of mTORC1 hyperactivity, and compare the effects of distinct PI3K-mTOR pathway and GATOR1 complex gene variants on cortical development and epilepsy. We report the outcomes on cortical pyramidal neuronal placement, morphology, and electrophysiological phenotypes, and discuss some of the converging and diverging mechanisms responsible for these alterations and their contribution to epileptogenesis. We also discuss potential therapeutic strategies for epilepsy, beyond mTORC1 inhibition with rapamycin or everolimus, that could offer personalized medicine based on the gene variant.
Collapse
Affiliation(s)
- Lena H Nguyen
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, United States.,Department of Cellular & Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Angélique Bordey
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, United States.,Department of Cellular & Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
44
|
4E-BP2-dependent translation in parvalbumin neurons controls epileptic seizure threshold. Proc Natl Acad Sci U S A 2021; 118:2025522118. [PMID: 33876772 DOI: 10.1073/pnas.2025522118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The mechanistic/mammalian target of rapamycin complex 1 (mTORC1) integrates multiple signals to regulate critical cellular processes such as mRNA translation, lipid biogenesis, and autophagy. Germline and somatic mutations in mTOR and genes upstream of mTORC1, such as PTEN, TSC1/2, AKT3, PIK3CA, and components of GATOR1 and KICSTOR complexes, are associated with various epileptic disorders. Increased mTORC1 activity is linked to the pathophysiology of epilepsy in both humans and animal models, and mTORC1 inhibition suppresses epileptogenesis in humans with tuberous sclerosis and animal models with elevated mTORC1 activity. However, the role of mTORC1-dependent translation and the neuronal cell types mediating the effect of enhanced mTORC1 activity in seizures remain unknown. The eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) and 2 (4E-BP2) are translational repressors downstream of mTORC1. Here we show that the ablation of 4E-BP2, but not 4E-BP1, in mice increases the sensitivity to pentylenetetrazole (PTZ)- and kainic acid (KA)-induced seizures. We demonstrate that the deletion of 4E-BP2 in inhibitory, but not excitatory neurons, causes an increase in the susceptibility to PTZ-induced seizures. Moreover, mice lacking 4E-BP2 in parvalbumin, but not somatostatin or VIP inhibitory neurons exhibit a lowered threshold for seizure induction and reduced number of parvalbumin neurons. A mouse model harboring a human PIK3CA mutation that enhances the activity of the PI3K-AKT pathway (Pik3ca H1047R-Pvalb ) selectively in parvalbumin neurons shows susceptibility to PTZ-induced seizures. Our data identify 4E-BP2 as a regulator of epileptogenesis and highlight the central role of increased mTORC1-dependent translation in parvalbumin neurons in the pathophysiology of epilepsy.
Collapse
|
45
|
Specchio N, Pepi C, De Palma L, Trivisano M, Vigevano F, Curatolo P. Neuroimaging and genetic characteristics of malformation of cortical development due to mTOR pathway dysregulation: clues for the epileptogenic lesions and indications for epilepsy surgery. Expert Rev Neurother 2021; 21:1333-1345. [PMID: 33754929 DOI: 10.1080/14737175.2021.1906651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Malformation of cortical development (MCD) is strongly associated with drug-resistant epilepsies for which surgery to remove epileptogenic lesions is common. Two notable technological advances in this field are identification of the underlying genetic cause and techniques in neuroimaging. These now question how presurgical evaluation ought to be approached for 'mTORpathies.'Area covered: From review of published primary and secondary articles, the authors summarize evidence to consider focal cortical dysplasia (FCD), tuber sclerosis complex (TSC), and hemimegalencephaly (HME) collectively as MCD mTORpathies. The authors also consider the unique features of these related conditions with particular focus on the practicalities of using neuroimaging techniques currently available to define surgical targets and predict post-surgical outcome. Ultimately, the authors consider the surgical dilemmas faced for each condition.Expert opinion: Considering FCD, TSC, and HME collectively as mTORpathies has some merit; however, a unified approach to presurgical evaluation would seem unachievable. Nevertheless, the authors believe combining genetic-centered classification and morphologic findings using advanced imaging techniques will eventually form the basis of a paradigm when considering candidacy for early surgery.
Collapse
Affiliation(s)
- Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Chiara Pepi
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Luca De Palma
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Marina Trivisano
- Rare and Complex Epilepsy Unit, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Federico Vigevano
- Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy
| |
Collapse
|
46
|
Goldstein HE, Hauptman JS. The Putative Role of mTOR Inhibitors in Non-tuberous Sclerosis Complex-Related Epilepsy. Front Neurol 2021; 12:639319. [PMID: 33643212 PMCID: PMC7907183 DOI: 10.3389/fneur.2021.639319] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/25/2021] [Indexed: 11/13/2022] Open
Abstract
Epilepsy affects ~5 out of every 10,000 children per year. Up to one-third of these children have medically refractory epilepsy, with limited to no options for improved seizure control. mTOR, a ubiquitous 289 kDa serine/threonine kinase in the phosphatidylinositol 3-kinase (PI3K)-related kinases (PIKK) family, is dysregulated in a number of human diseases, including tuberous sclerosis complex (TSC) and epilepsy. In cell models of epilepsy and TSC, rapamycin, an mTOR inhibitor, has been shown to decrease seizure frequency and duration, and positively affect cell growth and morphology. Rapamycin has also been shown to prevent or improve epilepsy and prolong survival in animal models of TSC. To date, clinical studies looking at the effects of mTOR inhibitors on the reduction of seizures have mainly focused on patients with TSC. Everolimus (Novartis Pharmaceuticals), a chemically modified rapamycin derivative, has been shown to reduce seizure frequency with reasonable safety and tolerability. Mutations in mTOR or the mTOR pathway have been found in hemimegalencephaly (HME) and focal cortical dysplasias (FCDs), both of which are highly correlated with medically refractory epilepsy. Given the evidence to date, a logical next step is to investigate the role of mTOR inhibitors in the treatment of children with medically refractory non-TSC epilepsy, particularly those children who have also failed resective surgery.
Collapse
Affiliation(s)
- Hannah E Goldstein
- Department of Neurological Surgery, University of Washington, Seattle, WA, United States.,Department of Neurosurgery, Seattle Children's Hospital, Seattle, WA, United States
| | - Jason S Hauptman
- Department of Neurological Surgery, University of Washington, Seattle, WA, United States.,Department of Neurosurgery, Seattle Children's Hospital, Seattle, WA, United States
| |
Collapse
|
47
|
Rodin RE, Dou Y, Kwon M, Sherman MA, D'Gama AM, Doan RN, Rento LM, Girskis KM, Bohrson CL, Kim SN, Nadig A, Luquette LJ, Gulhan DC, Park PJ, Walsh CA. The landscape of somatic mutation in cerebral cortex of autistic and neurotypical individuals revealed by ultra-deep whole-genome sequencing. Nat Neurosci 2021; 24:176-185. [PMID: 33432195 PMCID: PMC7983596 DOI: 10.1038/s41593-020-00765-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 11/21/2020] [Indexed: 01/29/2023]
Abstract
We characterize the landscape of somatic mutations-mutations occurring after fertilization-in the human brain using ultra-deep (~250×) whole-genome sequencing of prefrontal cortex from 59 donors with autism spectrum disorder (ASD) and 15 control donors. We observe a mean of 26 somatic single-nucleotide variants per brain present in ≥4% of cells, with enrichment of mutations in coding and putative regulatory regions. Our analysis reveals that the first cell division after fertilization produces ~3.4 mutations, followed by 2-3 mutations in subsequent generations. This suggests that a typical individual possesses ~80 somatic single-nucleotide variants present in ≥2% of cells-comparable to the number of de novo germline mutations per generation-with about half of individuals having at least one potentially function-altering somatic mutation somewhere in the cortex. ASD brains show an excess of somatic mutations in neural enhancer sequences compared with controls, suggesting that mosaic enhancer mutations may contribute to ASD risk.
Collapse
Affiliation(s)
- Rachel E Rodin
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Yanmei Dou
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Minseok Kwon
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Maxwell A Sherman
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alissa M D'Gama
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Ryan N Doan
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
| | - Lariza M Rento
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Kelly M Girskis
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Craig L Bohrson
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Sonia N Kim
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Ajay Nadig
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA
| | - Lovelace J Luquette
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Doga C Gulhan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA.
| | - Christopher A Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
- Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
48
|
Jesus-Ribeiro J, Pires LM, Melo JD, Ribeiro IP, Rebelo O, Sales F, Freire A, Melo JB. Genomic and Epigenetic Advances in Focal Cortical Dysplasia Types I and II: A Scoping Review. Front Neurosci 2021; 14:580357. [PMID: 33551717 PMCID: PMC7862327 DOI: 10.3389/fnins.2020.580357] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction: Focal cortical dysplasias (FCDs) are a group of malformations of cortical development that constitute a common cause of drug-resistant epilepsy, often subjected to neurosurgery, with a suboptimal long-term outcome. The past few years have witnessed a dramatic leap in our understanding of the molecular basis of FCD. This study aimed to provide an updated review on the genomic and epigenetic advances underlying FCD etiology, to understand a genotype-phenotype correlation and identify priorities to lead future translational research. Methods: A scoping review of the literature was conducted, according to previously described methods. A comprehensive search strategy was applied in PubMed, Embase, and Web of Science from inception to 07 May 2020. References were screened based on title and abstract, and posteriorly full-text articles were assessed for inclusion according to eligibility criteria. Studies with novel gene variants or epigenetic regulatory mechanisms in patients that underwent epilepsy surgery, with histopathological diagnosis of FCD type I or II according to Palmini's or the ILAE classification system, were included. Data were extracted and summarized for an overview of evidence. Results: Of 1,156 candidate papers, 39 met the study criteria and were included in this review. The advent of next-generation sequencing enabled the detection in resected FCD tissue of low-level brain somatic mutations that occurred during embryonic corticogenesis. The mammalian target of rapamycin (mTOR) signaling pathway, involved in neuronal growth and migration, is the key player in the pathogenesis of FCD II. Somatic gain-of-function variants in MTOR and its activators as well as germline, somatic, and second-hit mosaic loss-of-function variants in its related repressors have been reported. However, the genetic background of FCD type I remains elusive, with a pleomorphic repertoire of genes affected. DNA methylation and microRNAs were the two epigenetic mechanisms that proved to have a functional role in FCD and may represent molecular biomarkers. Conclusion: Further research into the possible pathogenic causes of both FCD subtypes is required, incorporating single-cell DNA/RNA sequencing as well as methylome and proteomic analysis. The collected data call for an integrated clinicopathologic and molecular genetic diagnosis in current practice not only to improve diagnostic accuracy but also to guide the development of future targeted treatments.
Collapse
Affiliation(s)
- Joana Jesus-Ribeiro
- Epilepsy and Sleep Monitoring Unit, Neurology Department, Coimbra University Hospital Center, Coimbra, Portugal.,iCBR/CIMAGO, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Luís Miguel Pires
- iCBR/CIMAGO, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Laboratory of Cytogenetics and Genomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | - Ilda Patrícia Ribeiro
- iCBR/CIMAGO, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Laboratory of Cytogenetics and Genomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Olinda Rebelo
- Neuropathology Laboratory, Neurology Department, Coimbra University Hospital Center, Coimbra, Portugal
| | - Francisco Sales
- Epilepsy and Sleep Monitoring Unit, Neurology Department, Coimbra University Hospital Center, Coimbra, Portugal
| | - António Freire
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Joana Barbosa Melo
- iCBR/CIMAGO, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Laboratory of Cytogenetics and Genomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
49
|
Guerrini R, Cavallin M, Pippucci T, Rosati A, Bisulli F, Dimartino P, Barba C, Garbelli R, Buccoliero AM, Tassi L, Conti V. Is Focal Cortical Dysplasia/Epilepsy Caused by Somatic MTOR Mutations Always a Unilateral Disorder? NEUROLOGY-GENETICS 2020; 7:e540. [PMID: 33542949 PMCID: PMC7735020 DOI: 10.1212/nxg.0000000000000540] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 10/21/2020] [Indexed: 11/15/2022]
Abstract
Objective To alert about the wide margin of unpredictability that distribution of somatic MTOR mosaicism may have in the brain and the risk for independent epileptogenesis arising from the seemingly healthy contralateral hemisphere after complete removal of epileptogenic focal cortical dysplasia (FCD). Methods Clinical, EEG, MRI, histopathology, and molecular genetics in 2 patients (1 and 2) treated with focal resections and subsequent complete hemispherectomy for epileptogenic FCD due to somatic MTOR mutations. Autoptic brain study of bilateral asymmetric hemispheric dysplasia and identification of alternative allele fraction (AAF) rates for AKT1 (patient 3). Results The strongly hyperactivating p.Ser2215Phe (patient 1) and p.Leu1460Pro (patient 2) MTOR mutations were at low-level AAF in the dysplastic tissue. After repeated resections and eventual complete hemispherectomy, both patients manifested intractable seizures arising from the contralateral, seemingly healthy hemisphere. In patient 3, the p.Glu17Lys AKT1 mutation exhibited random distribution and AAF rates in different tissues with double levels in the more severely dysplastic cerebral hemisphere. Conclusions Our understanding of the distribution of somatic mutations in the brain in relation to the type of malformation and its hypothesized time of origin may be faulty. Large studies may reveal that the risk of a first surgery being disappointing might be related more to the specific somatic mammalian target of rapamycin mutation identified than to completeness of resection and that the advantages of repeated resections after a first unsuccessful operation should be weighed against the risk of the contralateral hemisphere becoming in turn epileptogenic.
Collapse
Affiliation(s)
- Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| | - Mara Cavallin
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| | - Tommaso Pippucci
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| | - Anna Rosati
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| | - Francesca Bisulli
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| | - Paola Dimartino
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| | - Carmen Barba
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| | - Rita Garbelli
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| | - Anna Maria Buccoliero
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| | - Laura Tassi
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| | - Valerio Conti
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories (R. Guerrini, M.C., A.R, C.B., V.C.), Children's Hospital A. Meyer, University of Florence; Medical Genetics Unit (T.P.), Sant'Orsola-Malpighi University Hospital, Bologna; IRCCS Bologna Institute for Neurological Sciences (F.B.), Bologna. Member of ERN EpiCARE; Department of Medical and Surgical Sciences (P.D.), University of Bologna; Clinical Epileptology and Experimental Neurophysiology Unit (R. Garbelli), IRCCS Istituto Neurologico C. Besta, Milan; Pathology Unit (A.M.B.), Children's Hospital A. Meyer-University of Florence; and "C. Munari" Epilepsy Surgery Center (L.T.), Niguarda Hospital, Milan, Italy
| |
Collapse
|
50
|
Kobow K, Jabari S, Pieper T, Kudernatsch M, Polster T, Woermann FG, Kalbhenn T, Hamer H, Rössler K, Mühlebner A, Spliet WGM, Feucht M, Hou Y, Stichel D, Korshunov A, Sahm F, Coras R, Blümcke I, von Deimling A. Mosaic trisomy of chromosome 1q in human brain tissue associates with unilateral polymicrogyria, very early-onset focal epilepsy, and severe developmental delay. Acta Neuropathol 2020; 140:881-891. [PMID: 32979071 PMCID: PMC7666281 DOI: 10.1007/s00401-020-02228-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/16/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
Polymicrogyria (PMG) is a developmental cortical malformation characterized by an excess of small and frustrane gyration and abnormal cortical lamination. PMG frequently associates with seizures. The molecular pathomechanisms underlying PMG development are not yet understood. About 40 genes have been associated with PMG, and small copy number variations have also been described in selected patients. We recently provided evidence that epilepsy-associated structural brain lesions can be classified based on genomic DNA methylation patterns. Here, we analyzed 26 PMG patients employing array-based DNA methylation profiling on formalin-fixed paraffin-embedded material. A series of 62 well-characterized non-PMG cortical malformations (focal cortical dysplasia type 2a/b and hemimegalencephaly), temporal lobe epilepsy, and non-epilepsy autopsy controls was used as reference cohort. Unsupervised dimensionality reduction and hierarchical cluster analysis of DNA methylation profiles showed that PMG formed a distinct DNA methylation class. Copy number profiling from DNA methylation data identified a uniform duplication spanning the entire long arm of chromosome 1 in 7 out of 26 PMG patients, which was verified by additional fluorescence in situ hybridization analysis. In respective cases, about 50% of nuclei in the center of the PMG lesion were 1q triploid. No chromosomal imbalance was seen in adjacent, architecturally normal-appearing tissue indicating mosaicism. Clinically, PMG 1q patients presented with a unilateral frontal or hemispheric PMG without hemimegalencephaly, a severe form of intractable epilepsy with seizure onset in the first months of life, and severe developmental delay. Our results show that PMG can be classified among other structural brain lesions according to their DNA methylation profile. One subset of PMG with distinct clinical features exhibits a duplication of chromosomal arm 1q.
Collapse
Affiliation(s)
- Katja Kobow
- Department of Neuropathology, Institute of Neuropathology, Affiliated Partner of the ERN EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054, Erlangen, Germany.
| | - Samir Jabari
- Department of Neuropathology, Institute of Neuropathology, Affiliated Partner of the ERN EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054, Erlangen, Germany
| | - Tom Pieper
- Department of Neurology, Schön Klinik Vogtareuth, Vogtareuth, Germany
| | - Manfred Kudernatsch
- Department of Neurosurgery and Epilepsy Surgery, Schön Klinik Vogtareuth, Vogtareuth, Germany
- Research Institute "Rehabilitation, Transition, Palliation", PMU Salzburg, Salzburg, Austria
| | - Tilman Polster
- Epilepsy Center Bethel, Krankenhaus Mara, Bielefeld, Germany
| | | | - Thilo Kalbhenn
- Department of Neurosurgery, Evangelisches Klinikum Bethel, Bielefeld, Germany
| | - Hajo Hamer
- Department of Neurology, Epilepsy Center, Universitätsklinikum Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Karl Rössler
- Department of Neurosurgery, Universitätsklinikum Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Angelika Mühlebner
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Wim G M Spliet
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martha Feucht
- Department of Pediatrics and Adolescent Medicine, Affiliated Partner of the ERN EpiCARE, Medical University Vienna, Vienna, Austria
| | - Yanghao Hou
- Department of Neuropathology, Universitätsklinikum Heidelberg, and, CCU Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Damian Stichel
- Department of Neuropathology, Universitätsklinikum Heidelberg, and, CCU Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrey Korshunov
- Department of Neuropathology, Universitätsklinikum Heidelberg, and, CCU Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Universitätsklinikum Heidelberg, and, CCU Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roland Coras
- Department of Neuropathology, Institute of Neuropathology, Affiliated Partner of the ERN EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054, Erlangen, Germany
| | - Ingmar Blümcke
- Department of Neuropathology, Institute of Neuropathology, Affiliated Partner of the ERN EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054, Erlangen, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Universitätsklinikum Heidelberg, and, CCU Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|