351
|
Baldassari S, Licchetta L, Tinuper P, Bisulli F, Pippucci T. GATOR1 complex: the common genetic actor in focal epilepsies. J Med Genet 2016; 53:503-10. [PMID: 27208208 DOI: 10.1136/jmedgenet-2016-103883] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/28/2016] [Indexed: 01/15/2023]
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) signalling pathway has multiple roles in regulating physiology of the whole body and, particularly, the brain. Deregulation of mTOR signalling has been associated to various neurological conditions, including epilepsy. Mutations in genes encoding components of Gap Activity TOward Rags 1 (GATOR1) (DEPDC5, NPRL2 and NPRL3), a complex involved in the inhibition of the mTOR complex 1 (mTORC1), have been recently implicated in the pathogenesis of a wide spectrum of focal epilepsies (FEs), both lesional and non-lesional. The involvement of DEPDC5, NPRL2 and NRPL3 in about 10% of FEs is in contrast to the concept that specific seizure semiology points to the main involvement of a distinct brain area. The hypothesised pathogenic mechanism underlying epilepsy is the loss of the inhibitory function of GATOR1 towards mTORC1. The identification of the correct therapeutic strategy in patients with FE is challenging, especially in those with refractory epilepsy and/or malformations of cortical development (MCDs). In such cases, surgical excision of the epileptogenic zone is a curative option, although the long-term outcome is still undefined. The GATOR1/mTOR signalling represents a promising therapeutic target in FEs due to mutations in mTOR pathway genes, as in tuberous sclerosis complex, another MCD-associated epilepsy caused by mTOR signalling hyperactivation.
Collapse
Affiliation(s)
- Sara Baldassari
- Department of Medical and Surgical Sciences-Medical Genetics Unit, Università degli Studi di Bologna Azienda Ospedaliera Sant'Orsola-Malpighi, Bologna, Italy
| | - Laura Licchetta
- IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paolo Tinuper
- IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Francesca Bisulli
- IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Tommaso Pippucci
- Department of Medical and Surgical Sciences-Medical Genetics Unit, Università degli Studi di Bologna Azienda Ospedaliera Sant'Orsola-Malpighi, Bologna, Italy
| |
Collapse
|
352
|
Weckhuysen S, Marsan E, Lambrecq V, Marchal C, Morin-Brureau M, An-Gourfinkel I, Baulac M, Fohlen M, Kallay Zetchi C, Seeck M, de la Grange P, Dermaut B, Meurs A, Thomas P, Chassoux F, Leguern E, Picard F, Baulac S. Involvement of GATOR complex genes in familial focal epilepsies and focal cortical dysplasia. Epilepsia 2016; 57:994-1003. [DOI: 10.1111/epi.13391] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2016] [Indexed: 12/18/2022]
|
353
|
Citraro R, Leo A, Constanti A, Russo E, De Sarro G. mTOR pathway inhibition as a new therapeutic strategy in epilepsy and epileptogenesis. Pharmacol Res 2016; 107:333-343. [DOI: 10.1016/j.phrs.2016.03.039] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/23/2016] [Accepted: 03/31/2016] [Indexed: 12/24/2022]
|
354
|
Dlugos D, Worrell G, Davis K, Stacey W, Szaflarski J, Kanner A, Sunderam S, Rogawski M, Jackson-Ayotunde P, Loddenkemper T, Diehl B, Fureman B, Dingledine R. 2014 Epilepsy Benchmarks Area III: Improve Treatment Options for Controlling Seizures and Epilepsy-Related Conditions Without Side Effects. Epilepsy Curr 2016; 16:192-7. [PMID: 27330452 PMCID: PMC4913858 DOI: 10.5698/1535-7511-16.3.192] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Dennis Dlugos
- Professor of Neurology and Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Greg Worrell
- Associate Professor of Neurology, Mayo Systems Electrophysiology Laboratory, Departments of Neurology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Kathryn Davis
- Assistant Professor, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - William Stacey
- Assistant Professor of Neurology, Department of Neurology, Department of Biomedical Engineering, University of Michigan
| | - Jerzy Szaflarski
- Professor, Department of Neurology, University of Alabama at Birmingham Department of Neurology and UAB Epilepsy Center, Birmingham, AL
| | - Andres Kanner
- Profressor of Clinical Neurology, Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL
| | - Sridhar Sunderam
- Assistant Professor, Department of Biomedical Engineering, University of Kentucky, Lexington, KY
| | - Mike Rogawski
- Professor, Center for Neurotherapeutics Discovery and Development and Department of Neurology, UC Davis School of Medicine, Sacramento, CA
| | - Patrice Jackson-Ayotunde
- Associate Professor, Department of Pharmaceutical Sciences, University of Maryland Eastern Shore, Princess Anne, MD
| | - Tobias Loddenkemper
- Associate Professor, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital & Harvard Medical School, Boston, MA
| | - Beate Diehl
- Clinical Neurophysiologist and Neurologist, Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, UK
| | - Brandy Fureman
- Program Director, Channels Synapses and Circuits Cluster, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Ray Dingledine
- Professor and Chair, Department of Pharmacology, Emory University, Atlanta, GA
| | - for the Epilepsy Benchmark Stewards
- Professor of Neurology and Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Associate Professor of Neurology, Mayo Systems Electrophysiology Laboratory, Departments of Neurology and Biomedical Engineering, Mayo Clinic, Rochester, MN
- Assistant Professor, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Assistant Professor of Neurology, Department of Neurology, Department of Biomedical Engineering, University of Michigan
- Professor, Department of Neurology, University of Alabama at Birmingham Department of Neurology and UAB Epilepsy Center, Birmingham, AL
- Profressor of Clinical Neurology, Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL
- Assistant Professor, Department of Biomedical Engineering, University of Kentucky, Lexington, KY
- Professor, Center for Neurotherapeutics Discovery and Development and Department of Neurology, UC Davis School of Medicine, Sacramento, CA
- Associate Professor, Department of Pharmaceutical Sciences, University of Maryland Eastern Shore, Princess Anne, MD
- Associate Professor, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital & Harvard Medical School, Boston, MA
- Clinical Neurophysiologist and Neurologist, Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, UK
- Program Director, Channels Synapses and Circuits Cluster, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
- Professor and Chair, Department of Pharmacology, Emory University, Atlanta, GA
| |
Collapse
|
355
|
Editorial. Curr Opin Neurol 2016; 29:148-50. [DOI: 10.1097/wco.0000000000000312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
356
|
Rapamycin prevents, but does not reverse, aberrant migration in Pten knockout neurons. Neurobiol Dis 2016; 93:12-20. [PMID: 26992888 DOI: 10.1016/j.nbd.2016.03.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 02/08/2016] [Accepted: 03/10/2016] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Phosphatase and tensin homolog (PTEN) is a major negative regulator of the Akt/mammalian target of rapamycin (MTOR) pathway. Mutations in PTEN have been found in a subset of individuals with autism and macrocephaly. Further, focal cortical dysplasia (FCD) has been observed in patients with PTEN mutations prompting us to examine the role of Pten in neuronal migration. The dentate gyrus of Pten(Flox/Flox) mice was injected with Cre- and non-Cre-expressing retroviral particles, which integrate into the dividing genome to birthdate cells. Control and Pten knockout (KO) cell position in the granule cell layer was quantified over time to reveal that Pten KO neurons exhibit an aberrant migratory phenotype beginning at 7.5days-post retroviral injection (DPI). We then assessed whether rapamycin, a mTor inhibitor, could prevent or reverse aberrant migration of granule cells. The preventative group received daily intraperitoneal (IP) injections of rapamycin from 3 to 14 DPI, before discrepancies in cell position have been established, while the reversal group received rapamycin afterward, from 14 to 24 DPI. We found that rapamycin prevented and reversed somal hypertrophy. However, rapamycin prevented, but did not reverse aberrant migration in Pten KO cells. We also find that altered migration occurs through mTorC1 and not mTorC2 activity. Together, these findings suggest a temporal window by which rapamycin can treat aberrant migration, and may have implications for the use of rapamycin to treat PTEN-mutation associated disorders. SIGNIFICANCE STATEMENT Mutations in phosphatase and tensin homolog (PTEN) have been linked to a subset of individuals with autism and macrocephaly, as well as Cowden Syndrome and focal cortical dysplasia. Pten loss leads to neuronal hypertrophy, but the role of Pten in neuronal migration is unclear. Here we have shown that loss of Pten leads to aberrant migration, which can be prevented but not reversed by treatment with rapamycin, a mTor inhibitor. These results are important to consider as clinical trials are developed to examine rapamycin as a therapeutic for autism with PTEN mutations. Our findings show that some abnormalities cannot be reversed, and suggest the potential need for genetic screening and preventative treatment.
Collapse
|
357
|
Matsushita Y, Sakai Y, Shimmura M, Shigeto H, Nishio M, Akamine S, Sanefuji M, Ishizaki Y, Torisu H, Nakabeppu Y, Suzuki A, Takada H, Hara T. Hyperactive mTOR signals in the proopiomelanocortin-expressing hippocampal neurons cause age-dependent epilepsy and premature death in mice. Sci Rep 2016; 6:22991. [PMID: 26961412 PMCID: PMC4785342 DOI: 10.1038/srep22991] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/25/2016] [Indexed: 12/28/2022] Open
Abstract
Epilepsy is a frequent comorbidity in patients with focal cortical dysplasia (FCD). Recent studies utilizing massive sequencing data identified subsets of genes that are associated with epilepsy and FCD. AKT and mTOR-related signals have been recently implicated in the pathogenic processes of epilepsy and FCD. To clarify the functional roles of the AKT-mTOR pathway in the hippocampal neurons, we generated conditional knockout mice harboring the deletion of Pten (Pten-cKO) in Proopiomelanocortin-expressing neurons. The Pten-cKO mice developed normally until 8 weeks of age, then presented generalized seizures at 8–10 weeks of age. Video-monitored electroencephalograms detected paroxysmal discharges emerging from the cerebral cortex and hippocampus. These mice showed progressive hypertrophy of the dentate gyrus (DG) with increased expressions of excitatory synaptic markers (Psd95, Shank3 and Homer). In contrast, the expression of inhibitory neurons (Gad67) was decreased at 6–8 weeks of age. Immunofluorescence studies revealed the abnormal sprouting of mossy fibers in the DG of the Pten-cKO mice prior to the onset of seizures. The treatment of these mice with an mTOR inhibitor rapamycin successfully prevented the development of seizures and reversed these molecular phenotypes. These data indicate that the mTOR pathway regulates hippocampal excitability in the postnatal brain.
Collapse
Affiliation(s)
- Yuki Matsushita
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Mitsunori Shimmura
- Department of Neurology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hiroshi Shigeto
- Department of Neurology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Miki Nishio
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Satoshi Akamine
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Masafumi Sanefuji
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshito Ishizaki
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hiroyuki Torisu
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Akira Suzuki
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Hidetoshi Takada
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Toshiro Hara
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
358
|
Terrone G, Voisin N, Abdullah Alfaiz A, Cappuccio G, Vitiello G, Guex N, D'Amico A, James Barkovich A, Brunetti-Pierri N, Del Giudice E, Reymond A. De novo PIK3R2 variant causes polymicrogyria, corpus callosum hyperplasia and focal cortical dysplasia. Eur J Hum Genet 2016; 24:1359-62. [PMID: 26860062 DOI: 10.1038/ejhg.2016.7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/08/2015] [Accepted: 12/16/2015] [Indexed: 11/09/2022] Open
Abstract
We report an 8-year-old boy with a complex cerebral malformation, intellectual disability, and complex partial seizures. Whole-exome sequencing revealed a yet unreported de novo variant in the PIK3R2 gene that was recently associated with megalencephaly-polymicrogyria-polydactyly-hydrocephalus (MPPH) syndrome and bilateral perisylvian polymicrogyria (BPP). Our patient showed cerebral abnormalities (megalencephaly, perisylvian polymicrogyria, and mega corpus callosum) that were consistent with these conditions. Imaging also showed right temporal anomalies suggestive of cortical dysplasia. Until now, only three variants (c.1117G>A (p.(G373R)), c.1126A>G (p.(K376E)) and c.1202T>C (p.(L401P))) affecting the SH2 domain of the PIK3R2 protein have been reported in MPPH and BPP syndromes. In contrast to the variants reported so far, the patient described herein exhibits the c.1669G>C (p.(D557H)) variant that affects a highly conserved residue at the interface with the PI3K catalytic subunit α. The phenotypic spectrum associated with variants in this gene and its pathway are likely to continue to expand as more cases are identified.
Collapse
Affiliation(s)
- Gaetano Terrone
- Department of Translational Medicine, Section of Pediatrics, Federico II University, Naples, Italy
| | - Norine Voisin
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Ali Abdullah Alfaiz
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland.,Bioinformatics Section, King Abdullah International Medical Research Center (KAIMRC), Riyadh, Kingdom of Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Gerarda Cappuccio
- Department of Translational Medicine, Section of Pediatrics, Federico II University, Naples, Italy
| | - Giuseppina Vitiello
- Department of Translational Medicine, Section of Pediatrics, Federico II University, Naples, Italy
| | - Nicolas Guex
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Alessandra D'Amico
- Department of Diagnostic Imaging, Neuroradiology Unit, Federico II University, Naples, Italy
| | - A James Barkovich
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Section of Pediatrics, Federico II University, Naples, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Ennio Del Giudice
- Department of Translational Medicine, Section of Pediatrics, Federico II University, Naples, Italy
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
359
|
Tee AR, Sampson JR, Pal DK, Bateman JM. The role of mTOR signalling in neurogenesis, insights from tuberous sclerosis complex. Semin Cell Dev Biol 2016; 52:12-20. [PMID: 26849906 DOI: 10.1016/j.semcdb.2016.01.040] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/05/2016] [Accepted: 01/25/2016] [Indexed: 11/30/2022]
Abstract
Understanding the development and function of the nervous system is one of the foremost aims of current biomedical research. The nervous system is generated during a relatively short period of intense neurogenesis that is orchestrated by a number of key molecular signalling pathways. Even subtle defects in the activity of these molecules can have serious repercussions resulting in neurological, neurodevelopmental and neurocognitive problems including epilepsy, intellectual disability and autism. Tuberous sclerosis complex (TSC) is a monogenic disease characterised by these problems and by the formation of benign tumours in multiple organs, including the brain. TSC is caused by mutations in the TSC1 or TSC2 gene leading to activation of the mechanistic target of rapamycin (mTOR) signalling pathway. A desire to understand the neurological manifestations of TSC has stimulated research into the role of the mTOR pathway in neurogenesis. In this review we describe TSC neurobiology and how the use of animal model systems has provided insights into the roles of mTOR signalling in neuronal differentiation and migration. Recent progress in this field has identified novel mTOR pathway components regulating neuronal differentiation. The roles of mTOR signalling and aberrant neurogenesis in epilepsy are also discussed. Continuing efforts to understand mTOR neurobiology will help to identify new therapeutic targets for TSC and other neurological diseases.
Collapse
Affiliation(s)
- Andrew R Tee
- Institute of Cancer & Genetics, Cardiff University School of Medicine, Institute of Medical Genetics Building, Heath Park, Cardiff CF14 4XN UK
| | - Julian R Sampson
- Institute of Cancer & Genetics, Cardiff University School of Medicine, Institute of Medical Genetics Building, Heath Park, Cardiff CF14 4XN UK
| | - Deb K Pal
- Department of Basic & Clinical Neurosciences, Institute of Psychiatry, Psychology & Neuroscience, King's College, London SE5 8RX UK
| | - Joseph M Bateman
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL UK.
| |
Collapse
|
360
|
Tuberous sclerosis--A model for tumour growth. Semin Cell Dev Biol 2016; 52:3-11. [PMID: 26816112 DOI: 10.1016/j.semcdb.2016.01.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/18/2015] [Accepted: 01/19/2016] [Indexed: 01/06/2023]
Abstract
Tuberous sclerosis complex (TSC) is a rare genetic disorder where patients develop benign tumours in several organ systems. Central to TSC pathology is hyper-activation of the mammalian target of rapamycin complex 1 (mTORC1) signalling pathway, which is a key controller of cell growth. As a result, TSC model systems are a valuable tool for examining mTORC1-driven cellular processes. The immunosuppressant, rapamycin, is a specific inhibitor of mTORC1 and has shown promise as a therapeutic agent in TSC as well as in malignancy. This review will focus on the cellular processes controlled by mTORC1 and how TSC-deficient cell lines and mouse models have broadened our understanding of the mTORC1 signalling network. It will also discuss how our knowledge of TSC signalling can help us understand sporadic conditions where mTORC1 activity is implicated in disease onset or progression, and the possibility of using rapamycin to treat sporadic disease.
Collapse
|
361
|
Poopal AC, Schroeder LM, Horn PS, Bassell GJ, Gross C. Increased expression of the PI3K catalytic subunit p110δ underlies elevated S6 phosphorylation and protein synthesis in an individual with autism from a multiplex family. Mol Autism 2016; 7:3. [PMID: 26770665 PMCID: PMC4712554 DOI: 10.1186/s13229-015-0066-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/28/2015] [Indexed: 12/26/2022] Open
Abstract
Background Dysfunctions in the PI3K/mTOR pathway have gained a lot of attention in autism research. This was initially based on the discovery of several monogenic autism spectrum disorders with mutations or defects in PI3K/mTOR signaling components. Recent genetic studies corroborate that defective PI3K/mTOR signaling might be a shared pathomechanism in autism disorders of so far unknown etiology, but functional molecular analyses in human cells are rare. The goals of this study were to perform a functional screen of cell lines from patients with idiopathic autism for defects in PI3K/mTOR signaling, to test if further functional analyses are suitable to detect underlying molecular mechanisms, and to evaluate this approach as a biomarker tool to identify therapeutic targets. Methods We performed phospho-S6- and S6-specific ELISA experiments on 21 lymphoblastoid cell lines from the AGRE collection and on 37 lymphoblastoid cell lines from the Simons Simplex Collection and their healthy siblings. Cell lines from one individual with increased S6 phosphorylation and his multiplex family were analyzed in further detail to identify upstream defects in PI3K signaling associated with autism diagnosis. Results We detected significantly increased S6 phosphorylation in 3 of the 21 lymphoblastoid cell lines from AGRE compared to a healthy control and in 1 of the 37 lymphoblastoid cell lines from the Simons Simplex Collection compared to the healthy sibling. Further analysis of cells from one individual with elevated S6 phosphorylation showed increased expression of the PI3K catalytic subunit p110δ, which was also observed in lymphoblastoid cells from other autistic siblings but not unaffected members in his multiplex family. The p110δ-selective inhibitor IC87114 reduced elevated S6 phosphorylation and protein synthesis in this cell line. Conclusions Our results suggest that functional analysis of PI3K/mTOR signaling is a biomarker tool to identify disease-associated molecular defects that could serve as therapeutic targets in autism. Using this approach, we discovered impaired signaling and protein synthesis through the PI3K catalytic subunit p110δ as an underlying molecular defect and potential treatment target in select autism spectrum disorders. Increased p110δ activity was recently associated with schizophrenia, and our results suggest that p110δ may also be implicated in autism. Electronic supplementary material The online version of this article (doi:10.1186/s13229-015-0066-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ashwini C Poopal
- Department of Cell Biology, Emory University Medical School, 615 Michael Street, Atlanta, GA 30322 USA
| | - Lindsay M Schroeder
- Division of Neurology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229 USA
| | - Paul S Horn
- Division of Neurology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229 USA ; Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229 USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University Medical School, 615 Michael Street, Atlanta, GA 30322 USA
| | - Christina Gross
- Department of Cell Biology, Emory University Medical School, 615 Michael Street, Atlanta, GA 30322 USA ; Division of Neurology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229 USA
| |
Collapse
|
362
|
Deletion of mTOR in Reactive Astrocytes Suppresses Chronic Seizures in a Mouse Model of Temporal Lobe Epilepsy. Mol Neurobiol 2016; 54:175-187. [PMID: 26732600 DOI: 10.1007/s12035-015-9590-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/29/2015] [Indexed: 01/28/2023]
Abstract
Germline and somatic mutations in key genes of the mammalian target of rapamycin (mTOR) pathway have been identified in seizure-associated disorders. mTOR mutations lead to aberrant activation of mTOR signaling, and, although affected neurons are critical for epileptogenesis, the role of mTOR activation in glial cells remains poorly understood. We previously reported a consistent activation of the mTOR pathway in astrocytes in the epileptic foci of temporal lobe epilepsy. In this study, it was demonstrated that mTOR deletion from reactive astrocytes prevents increases in seizure frequency over the disease course. By using a tamoxifen-inducible mTOR conditional knockout system and kainic acid, a model was developed that allowed astrocyte-specific mTOR gene deletion in mice with chronic epilepsy. Animals in which mTOR was deleted from 44 % of the astrocyte population exhibited a lower seizure frequency compared with controls. Down-regulation of mTOR significantly ameliorated astrogliosis in the sclerotic hippocampus but did not rescue mossy fiber sprouting. In cultured astrocytes, the mTOR pathway modulated the stability of the astroglial glutamate transporter 1 (Glt1) and influenced the ability of astrocytes to remove extracellular glutamate. Taken together, these data indicate that astrocytes with activated mTOR signaling may provide conditions that are favorable for spontaneous recurrent seizures.
Collapse
|
363
|
Baulac S. mTOR signaling pathway genes in focal epilepsies. PROGRESS IN BRAIN RESEARCH 2016; 226:61-79. [DOI: 10.1016/bs.pbr.2016.04.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
364
|
Myers C, Mefford H. Genetic investigations of the epileptic encephalopathies. PROGRESS IN BRAIN RESEARCH 2016; 226:35-60. [DOI: 10.1016/bs.pbr.2016.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
365
|
Ainslie GR, Gibson KM, Vogel KR. mTOR, Autophagy, Aminoacidopathies, and Human Genetic Disorders. MOLECULES TO MEDICINE WITH MTOR 2016:143-166. [DOI: 10.1016/b978-0-12-802733-2.00010-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
366
|
Sim JC, Scerri T, Fanjul-Fernández M, Riseley JR, Gillies G, Pope K, van Roozendaal H, Heng JI, Mandelstam SA, McGillivray G, MacGregor D, Kannan L, Maixner W, Harvey AS, Amor DJ, Delatycki MB, Crino PB, Bahlo M, Lockhart PJ, Leventer RJ. Familial cortical dysplasia caused by mutation in the mammalian target of rapamycin regulator NPRL3. Ann Neurol 2015; 79:132-7. [PMID: 26285051 DOI: 10.1002/ana.24502] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/04/2015] [Accepted: 08/04/2015] [Indexed: 02/03/2023]
Abstract
We describe first cousin sibling pairs with focal epilepsy, one of each pair having focal cortical dysplasia (FCD) IIa. Linkage analysis and whole-exome sequencing identified a heterozygous germline frameshift mutation in the gene encoding nitrogen permease regulator-like 3 (NPRL3). NPRL3 is a component of GAP Activity Towards Rags 1, a negative regulator of the mammalian target of rapamycin complex 1 signaling pathway. Immunostaining of resected brain tissue demonstrated mammalian target of rapamycin activation. Screening of 52 unrelated individuals with FCD identified 2 additional patients with FCDIIa and germline NPRL3 mutations. Similar to DEPDC5, NPRL3 mutations may be considered as causal variants in patients with FCD or magnetic resonance imaging-negative focal epilepsy.
Collapse
Affiliation(s)
- Joe C Sim
- Bruce Lefroy Center for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia
| | - Thomas Scerri
- Bioinformatics and Population Health and Immunity Divisions, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Miriam Fanjul-Fernández
- Bruce Lefroy Center for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia
| | - Jessica R Riseley
- Bruce Lefroy Center for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia
| | - Greta Gillies
- Bruce Lefroy Center for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia
| | - Kate Pope
- Bruce Lefroy Center for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia
| | | | - Julian I Heng
- The Harry Perkins Institute of Medical Research, The Center for Medical Research, University of Western Australia, Perth, Australia
| | - Simone A Mandelstam
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia.,University of Melbourne, Department of Radiology, Melbourne, Australia.,University of Melbourne, Department of Pediatrics, Melbourne, Australia
| | - George McGillivray
- Bruce Lefroy Center for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia
| | - Duncan MacGregor
- Department of Anatomical Pathology, Royal Children's Hospital, Melbourne, Australia
| | | | - Wirginia Maixner
- Neuroscience Research Group, Murdoch Childrens Research Institute, Melbourne, Australia.,Department of Neurosurgery, Royal Children's Hospital, Melbourne, Australia
| | - A Simon Harvey
- Department of Neurology, Royal Children's Hospital, Melbourne, Australia.,Neuroscience Research Group, Murdoch Childrens Research Institute, Melbourne, Australia.,University of Melbourne, Department of Pediatrics, Melbourne, Australia
| | - David J Amor
- Bruce Lefroy Center for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia.,University of Melbourne, Department of Pediatrics, Melbourne, Australia
| | - Martin B Delatycki
- Bruce Lefroy Center for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia.,University of Melbourne, Department of Pediatrics, Melbourne, Australia.,Clinical Genetics, Austin Health, Melbourne, Australia
| | - Peter B Crino
- Shriners Hospital Pediatric Research Center, Temple University, Philadelphia, PA
| | - Melanie Bahlo
- Bioinformatics and Population Health and Immunity Divisions, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Paul J Lockhart
- Bruce Lefroy Center for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia.,University of Melbourne, Department of Pediatrics, Melbourne, Australia
| | - Richard J Leventer
- Department of Neurology, Royal Children's Hospital, Melbourne, Australia.,Neuroscience Research Group, Murdoch Childrens Research Institute, Melbourne, Australia.,University of Melbourne, Department of Pediatrics, Melbourne, Australia
| |
Collapse
|
367
|
Roy A, Skibo J, Kalume F, Ni J, Rankin S, Lu Y, Dobyns WB, Mills GB, Zhao JJ, Baker SJ, Millen KJ. Mouse models of human PIK3CA-related brain overgrowth have acutely treatable epilepsy. eLife 2015; 4. [PMID: 26633882 PMCID: PMC4744197 DOI: 10.7554/elife.12703] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 11/26/2015] [Indexed: 12/12/2022] Open
Abstract
Mutations in the catalytic subunit of phosphoinositide 3-kinase (PIK3CA) and other PI3K-AKT pathway components have been associated with cancer and a wide spectrum of brain and body overgrowth. In the brain, the phenotypic spectrum of PIK3CA-related segmental overgrowth includes bilateral dysplastic megalencephaly, hemimegalencephaly and focal cortical dysplasia, the most common cause of intractable pediatric epilepsy. We generated mouse models expressing the most common activating Pik3ca mutations (H1047R and E545K) in developing neural progenitors. These accurately recapitulate all the key human pathological features including brain enlargement, cortical malformation, hydrocephalus and epilepsy, with phenotypic severity dependent on the mutant allele and its time of activation. Underlying mechanisms include increased proliferation, cell size and altered white matter. Notably, we demonstrate that acute 1 hr-suppression of PI3K signaling despite the ongoing presence of dysplasia has dramatic anti-epileptic benefit. Thus PI3K inhibitors offer a promising new avenue for effective anti-epileptic therapy for intractable pediatric epilepsy patients. DOI:http://dx.doi.org/10.7554/eLife.12703.001 An enzyme called PI3K is involved in a major signaling pathway that controls cell growth. Mutations in this pathway have devastating consequences. When such mutations happen in adults, they can lead to cancer. Mutations that occur in embryos can cause major developmental birth defects, including abnormally large brains. After birth, these developmental problems can cause intellectual disabilities, autism and epilepsy. Children with this kind of epilepsy often do not respond to currently available seizure medications. There are several outstanding questions that if answered could help efforts to develop treatments for children with brain growth disorders. Firstly, how do the developmental abnormalities happen? Do the abnormalities themselves cause epilepsy? And can drugs that target this pathway, and are already in clinical trials for cancer, control seizures? Now, Roy et al. have made mouse models of these human developmental brain disorders and used them to answer these questions. The mice were genetically engineered to have various mutations in the gene that encodes the catalytic subunit of the PI3K enzyme. The mutations were the same as those found in people with brain overgrowth disorders, and were activated only in the developing brain of the mice. These mutations caused enlarged brain size, fluid accumulation in the brain, brain malformations and epilepsy in developing mice – thus mimicking the human birth defects. The severity of these symptoms depended on the specific mutation and when the mutant genes were turned on during development. Next, Roy et al. studied these mice to see if the seizures could be treated using a drug, that has already been developed for brain cancer. This drug specifically targets and reduces the activity of PI3K. Adult mutant mice with brain malformations were treated for just one hour; this dramatically reduced their seizures. These experiments prove that seizures associated with this kind of brain overgrowth disorder are driven by ongoing abnormal PI3K activity and can be treated even when underlying brain abnormalities persist. Roy et al. suggest that drugs targeting PI3K might help treat seizures in children with these brain overgrowth disorders. DOI:http://dx.doi.org/10.7554/eLife.12703.002
Collapse
Affiliation(s)
- Achira Roy
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States
| | - Jonathan Skibo
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States
| | - Franck Kalume
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States
| | - Jing Ni
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, United States
| | - Sherri Rankin
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, United States
| | - Yiling Lu
- The University of Texas MD Anderson Cancer Center, Houston, United States
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States
| | - Gordon B Mills
- The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Jean J Zhao
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, United States
| | - Suzanne J Baker
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, United States
| | - Kathleen J Millen
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States
| |
Collapse
|
368
|
LRP12 silencing during brain development results in cortical dyslamination and seizure sensitization. Neurobiol Dis 2015; 86:170-6. [PMID: 26639854 DOI: 10.1016/j.nbd.2015.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 10/20/2015] [Accepted: 11/26/2015] [Indexed: 02/04/2023] Open
Abstract
Correct positioning and differentiation of neurons during brain development is a key precondition for proper function. Focal cortical dysplasias (FCDs) are increasingly recognized as causes of therapy refractory epilepsies. Neuropathological analyses of respective surgical specimens from neurosurgery for seizure control often reveal aberrant cortical architecture and/or aberrantly shaped neurons in FCDs. However, the molecular pathogenesis particularly of FCDs with aberrant lamination (so-called FCD type I) is largely unresolved. Lipoproteins and particularly low-density lipoprotein receptor-related protein 12 (LRP12) are involved in brain development. Here, we have examined a potential role of LRP12 in the pathogenesis of FCDs. In vitro knockdown of LRP12 in primary neurons results in impaired neuronal arborization. In vivo ablation of LRP12 by intraventricularly in utero electroporated shRNAs elicits cortical maldevelopment, i.e. aberrant lamination by malpositioning of upper cortical layer neurons. Subsequent epilepsy phenotyping revealed pentylenetetrazol (PTZ)-induced seizures to be aggravated in cortical LRP12-silenced mice. Our data demonstrates IUE mediated cortical gene silencing as an excellent approach to study the role of distinct molecules for epilepsy associated focal brain lesions and suggests LRP12 and lipoprotein homeostasis as potential molecular target structures for the emergence of epilepsy-associated FCDs.
Collapse
|
369
|
Baek ST, Copeland B, Yun EJ, Kwon SK, Guemez-Gamboa A, Schaffer AE, Kim S, Kang HC, Song S, Mathern GW, Gleeson JG. An AKT3-FOXG1-reelin network underlies defective migration in human focal malformations of cortical development. Nat Med 2015; 21:1445-54. [PMID: 26523971 PMCID: PMC4955611 DOI: 10.1038/nm.3982] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 10/01/2015] [Indexed: 02/07/2023]
Abstract
Focal malformations of cortical development (FMCDs) account for the majority of drug-resistant pediatric epilepsy. Postzygotic somatic mutations activating the phosphatidylinositol-4,5-bisphosphate-3-kinase (PI3K)-protein kinase B (AKT)-mammalian target of rapamycin (mTOR) pathway are found in a wide range of brain diseases, including FMCDs. It remains unclear how a mutation in a small fraction of cells disrupts the architecture of the entire hemisphere. Within human FMCD-affected brain, we found that cells showing activation of the PI3K-AKT-mTOR pathway were enriched for the AKT3(E17K) mutation. Introducing the FMCD-causing mutation into mouse brain resulted in electrographic seizures and impaired hemispheric architecture. Mutation-expressing neural progenitors showed misexpression of reelin, which led to a non-cell autonomous migration defect in neighboring cells, due at least in part to derepression of reelin transcription in a manner dependent on the forkhead box (FOX) transcription factor FOXG1. Treatments aimed at either blocking downstream AKT signaling or inactivating reelin restored migration. These findings suggest a central AKT-FOXG1-reelin signaling pathway in FMCD and support pathway inhibitors as potential treatments or therapies for some forms of focal epilepsy.
Collapse
Affiliation(s)
- Seung Tae Baek
- Laboratory of Pediatric Brain Diseases, Rockefeller University, New York, New York, USA
- Department of Neurosciences, University of California San Diego (UCSD), La Jolla, California, USA
| | - Brett Copeland
- Laboratory of Pediatric Brain Diseases, Rockefeller University, New York, New York, USA
| | - Eun-Jin Yun
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Seok-Kyu Kwon
- Department of Neuroscience, Columbia University, New York, New York, USA
| | - Alicia Guemez-Gamboa
- Laboratory of Pediatric Brain Diseases, Rockefeller University, New York, New York, USA
| | - Ashleigh E Schaffer
- Department of Neurosciences, University of California San Diego (UCSD), La Jolla, California, USA
| | - Sangwoo Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Hoon-Chul Kang
- Laboratory of Pediatric Brain Diseases, Rockefeller University, New York, New York, USA
- Department of Pediatrics, Division of Pediatric Neurology, Pediatric Epilepsy Clinics, Severance Children's Hospital, Seoul, South Korea
- Epilepsy Research Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Saera Song
- Laboratory of Pediatric Brain Diseases, Rockefeller University, New York, New York, USA
| | - Gary W Mathern
- Department of Neurosurgery, Mattel Children's Hospital, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Department of Psychiatry and Biobehavioral Sciences, Mattel Children's Hospital, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Joseph G Gleeson
- Laboratory of Pediatric Brain Diseases, Rockefeller University, New York, New York, USA
- Department of Neurosciences, University of California San Diego (UCSD), La Jolla, California, USA
- Neurogenetics Laboratory, Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
370
|
Kang HC, Baek ST, Song S, Gleeson JG. Clinical and Genetic Aspects of the Segmental Overgrowth Spectrum Due to Somatic Mutations in PIK3CA. J Pediatr 2015; 167:957-62. [PMID: 26340871 DOI: 10.1016/j.jpeds.2015.07.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 06/10/2015] [Accepted: 07/27/2015] [Indexed: 02/07/2023]
Affiliation(s)
- Hoon-Chul Kang
- Laboratory of Pediatric Brain Disease, Howard Hughes Medical Institute, The Rockefeller University, New York, NY; Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University, College of Medicine, Seoul, Republic of Korea
| | - Seung Tae Baek
- Laboratory of Pediatric Brain Disease, Howard Hughes Medical Institute, The Rockefeller University, New York, NY
| | - Saera Song
- Laboratory of Pediatric Brain Disease, Howard Hughes Medical Institute, The Rockefeller University, New York, NY
| | - Joseph G Gleeson
- Laboratory of Pediatric Brain Disease, Howard Hughes Medical Institute, The Rockefeller University, New York, NY.
| |
Collapse
|
371
|
Guerrini R, Duchowny M, Jayakar P, Krsek P, Kahane P, Tassi L, Melani F, Polster T, Andre VM, Cepeda C, Krueger DA, Cross JH, Spreafico R, Cosottini M, Gotman J, Chassoux F, Ryvlin P, Bartolomei F, Bernasconi A, Stefan H, Miller I, Devaux B, Najm I, Giordano F, Vonck K, Barba C, Blumcke I. Diagnostic methods and treatment options for focal cortical dysplasia. Epilepsia 2015; 56:1669-86. [DOI: 10.1111/epi.13200] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Renzo Guerrini
- Pediatric Neurology and Neurogenetics Unit and Laboratories; Children's Hospital Meyer-University of Florence; Florence Italy
- IRCCS Stella Maris Foundation; Pisa Italy
| | - Michael Duchowny
- Neuroscience Program and the Comprehensive Epilepsy Center; Miami Children's Hospital; Miami Florida U.S.A
| | - Prasanna Jayakar
- Department of Neurology; Miami Children's Hospital; Miami Florida U.S.A
| | - Pavel Krsek
- Department of Pediatric Neurology; 2nd Faculty of Medicine; Motol University Hospital; Charles University; Prague Czech Republic
| | - Philippe Kahane
- INSERM U836; University of Grenoble Alpes, GIN; Grenoble; France
- Epilepsy Unit; Michallon Hospital; Grenoble France
| | - Laura Tassi
- Epilepsy Surgery Center; Niguarda Hospital; Milan Italy
| | - Federico Melani
- Pediatric Neurology and Neurogenetics Unit and Laboratories; Children's Hospital Meyer-University of Florence; Florence Italy
| | - Tilman Polster
- Department of Child Neurology; Bethel Epilepsy Center; Bielefeld Germany
| | | | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center; David Geffen School of Medicine; University of California at Los Angeles; Los Angeles California U.S.A
| | - Darcy A. Krueger
- Division of Neurology; Department of Pediatrics; Cincinnati Children's Hospital Medical Center; University of Cincinnati College of Medicine; Cincinnati Ohio U.S.A
| | - J. Helen Cross
- UCL-Institute of Child Health; Great Ormond Street Hospital for Children NHS Foundation Trust; London United Kingdom
- Young Epilepsy; Lingfield United Kingdom
| | - Roberto Spreafico
- Clinical Epileptology and Experimental Neurophysiology Unit; Neurological InstituteC. Besta”; Milan Italy
| | - Mirco Cosottini
- Department of Translational Research and New Technologies in Medicine and Surgery; University of Pisa; Pisa Italy
| | - Jean Gotman
- Montreal Neurological Institute and Hospital; McGill University; Montreal Quebec Canada
| | | | - Philippe Ryvlin
- Department of Clinical Neurosciences; CHUV; Lausanne Switzerland
- Translational and Integrative Group in Epilepsy Research (TIGER) and Institute for Epilepsies (IDEE); Lyon's Neuroscience Center; INSERM U1028; CNRS 5292; UCBL; Le Vinatier Hospital; Bron; Lyon France
| | - Fabrice Bartolomei
- Faculty of Medicine; INSERM, U1106; Institute of Neurosciences of Systems; Marseille France
- Faculty of Medicine; Aix Marseille University; Marseille France
- Clinical Neurophysiology Unit; Department of Clinical Neurosciences; CHU Timone; Marseille France
- Henri-Gastaut Hospital; Saint-Paul Center; Marseille France
| | - Andrea Bernasconi
- Neuroimaging of Epilepsy Laboratory; McConnell Brain Imaging Center; Montreal Neurological Institute and Hospital; McGill University; Montreal Quebec Canada
| | - Hermann Stefan
- Epilepsy Center Erlangen (ZEE); University Erlangen-Nürnberg; Erlangen Germany
| | - Ian Miller
- Department of Neurology and Comprehensive Epilepsy Program; Brain Institute; Miami Children's Hospital; Miami Florida U.S.A
| | | | - Imad Najm
- Epilepsy Center; Neurological Institute; Cleveland Clinic; Cleveland OH U.S.A
| | - Flavio Giordano
- Pediatric Neurosurgery Unit; Children's Hospital Meyer-University of Florence; Florence Italy
| | - Kristl Vonck
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology; Department of Neurology; Ghent University; Ghent Belgium
| | - Carmen Barba
- Pediatric Neurology and Neurogenetics Unit and Laboratories; Children's Hospital Meyer-University of Florence; Florence Italy
| | - Ingmar Blumcke
- Department of Neuropathology; University Hospital Erlangen; Erlangen Germany
| |
Collapse
|
372
|
Patil VV, Guzman M, Carter AN, Rathore G, Yoshor D, Curry D, Wilfong A, Agadi S, Swann JW, Adesina AM, Bhattacharjee MB, Anderson AE. Activation of extracellular regulated kinase and mechanistic target of rapamycin pathway in focal cortical dysplasia. Neuropathology 2015; 36:146-56. [PMID: 26381727 DOI: 10.1111/neup.12242] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/31/2015] [Accepted: 08/01/2015] [Indexed: 12/26/2022]
Abstract
Neuropathology of resected brain tissue has revealed an association of focal cortical dysplasia (FCD) with drug-resistant epilepsy (DRE). Recent studies have shown that the mechanistic target of rapamycin (mTOR) pathway is hyperactivated in FCD as evidenced by increased phosphorylation of the ribosomal protein S6 (S6) at serine 240/244 (S(240/244) ), a downstream target of mTOR. Moreover, extracellular regulated kinase (ERK) has been shown to phosphorylate S6 at serine 235/236 (S(235/236) ) and tuberous sclerosis complex 2 (TSC2) at serine 664 (S(664) ) leading to hyperactive mTOR signaling. We evaluated ERK phosphorylation of S6 and TSC2 in two types of FCD (FCD I and FCD II) as a candidate mechanism contributing to mTOR pathway dysregulation. Tissue samples from patients with tuberous sclerosis (TS) served as a positive control. Immunostaining for phospho-S6 (pS6(240/244) and pS6(235/236) ), phospho-ERK (pERK), and phospho-TSC2 (pTSC2) was performed on resected brain tissue with FCD and TS. We found increased pS6(240/244) and pS6(235/236) staining in FCD I, FCD II and TS compared to normal-appearing tissue, while pERK and pTSC2 staining was increased only in FCD IIb and TS tissue. Our results suggest that both the ERK and mTOR pathways are dysregulated in FCD and TS; however, the signaling alterations are different for FCD I as compared to FCD II and TS.
Collapse
Affiliation(s)
- Vinit V Patil
- Program in Translational Biology and Molecular Medicine, Texas Children's Hospital, Houston, Texas, USA.,Cain Foundation Laboratories, Texas Children's Hospital, Houston, Texas, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA.,Department of Pathology, Saint Louis University, Saint Louis, Missouri
| | - Miguel Guzman
- Department of Pathology, Saint Louis University, Saint Louis, Missouri
| | - Angela N Carter
- Department of Neuroscience, Texas Children's Hospital, Houston, Texas, USA.,Cain Foundation Laboratories, Texas Children's Hospital, Houston, Texas, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Geetanjali Rathore
- Department of Pediatrics, Texas Children's Hospital, Houston, Texas, USA
| | - Daniel Yoshor
- Department of Neurosurgery, Texas Children's Hospital, Houston, Texas, USA
| | - Daniel Curry
- Department of Neurosurgery, Texas Children's Hospital, Houston, Texas, USA
| | - Angus Wilfong
- Department of Neurology, Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Texas Children's Hospital, Houston, Texas, USA
| | - Satish Agadi
- Department of Neurology, Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Texas Children's Hospital, Houston, Texas, USA
| | - John W Swann
- Department of Neuroscience, Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Texas Children's Hospital, Houston, Texas, USA.,Program in Translational Biology and Molecular Medicine, Texas Children's Hospital, Houston, Texas, USA.,Cain Foundation Laboratories, Texas Children's Hospital, Houston, Texas, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | | | - Meenakshi B Bhattacharjee
- Department of Pathology and Laboratory Medicine, University of Texas Medical School, Houston, Texas, USA
| | - Anne E Anderson
- Department of Neurology, Texas Children's Hospital, Houston, Texas, USA.,Department of Neuroscience, Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Texas Children's Hospital, Houston, Texas, USA.,Program in Translational Biology and Molecular Medicine, Texas Children's Hospital, Houston, Texas, USA.,Cain Foundation Laboratories, Texas Children's Hospital, Houston, Texas, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
373
|
Abstract
Epilepsy is a group of disorders characterized by recurrent seizures, and is one of the most common neurological conditions. The genetic basis of epilepsy is clear from epidemiological studies and from rare gene discoveries in large families. The three major classes of epilepsy disorders are genetic generalized, focal and encephalopathic epilepsies, with several specific disorders within each class. Advances in genomic technologies that facilitate genome-wide discovery of both common and rare variants have led to a rapid increase in our understanding of epilepsy genetics. Copy number variant and genome-wide association studies have contributed to our understanding of the complex genetic architecture of generalized epilepsy, while genetic insights into the focal epilepsies and epileptic encephalopathies have come primarily from exome sequencing. It is increasingly clear that epilepsy is genetically heterogeneous, and novel gene discoveries have moved the field beyond the known contribution of ion channels to implicate chromatin remodeling, transcriptional regulation and regulation of the mammalian target of rapamycin (mTOR) protein in the etiology of epilepsy. Such discoveries pave the way for new therapeutics, some of which are already being studied. In this review, we discuss the rapid pace of gene discovery in epilepsy, as facilitated by genomic technologies, and highlight several novel genes and potential therapies.
Collapse
Affiliation(s)
- Candace T Myers
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Heather C Mefford
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
374
|
Delineation of New Disorders and Phenotypic Expansion of Known Disorders Through Whole Exome Sequencing. CURRENT GENETIC MEDICINE REPORTS 2015. [DOI: 10.1007/s40142-015-0079-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
375
|
Gong X, Zhang L, Huang T, Lin TV, Miyares L, Wen J, Hsieh L, Bordey A. Activating the translational repressor 4E-BP or reducing S6K-GSK3β activity prevents accelerated axon growth induced by hyperactive mTOR in vivo. Hum Mol Genet 2015. [PMID: 26220974 DOI: 10.1093/hmg/ddv295] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Abnormal axonal connectivity and hyperactive mTOR complex 1 (mTORC1) are shared features of several neurological disorders. Hyperactive mTORC1 alters axon length and polarity of hippocampal neurons in vitro, but the impact of hyperactive mTORC1 on axon growth in vivo and the mechanisms underlying those effects remain unclear. Using in utero electroporation during corticogenesis, we show that increasing mTORC1 activity accelerates axon growth without multiple axon formation. This was prevented by counteracting mTORC1 signaling through p70S6Ks (S6K1/2) or eukaryotic initiation factor 4E-binding protein (4E-BP1/2), which both regulate translation. In addition to regulating translational targets, S6K1 indirectly signals through GSK3β, a regulator of axogenesis. Although blocking GSK3β activity did not alter axon growth under physiological conditions in vivo, blocking it using a dominant-negative mutant or lithium chloride prevented mTORC1-induced accelerated axon growth. These data reveal the contribution of translational and non-translational downstream effectors such as GSK3β to abnormal axon growth in neurodevelopmental mTORopathies and open new therapeutic options for restoring long-range connectivity.
Collapse
Affiliation(s)
- Xuan Gong
- Department of Neurosurgery, Xiangya Hospital, Central South University, 85 Xiangya Street, Changsha 410008, China, Department of Neurosurgery and Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8082, USA
| | - Longbo Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, 85 Xiangya Street, Changsha 410008, China, Department of Neurosurgery and Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8082, USA
| | - Tianxiang Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, 85 Xiangya Street, Changsha 410008, China, Department of Neurosurgery and Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8082, USA
| | - Tiffany V Lin
- Department of Neurosurgery and Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8082, USA
| | - Laura Miyares
- Department of Neurosurgery and Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8082, USA
| | - John Wen
- Department of Neurosurgery and Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8082, USA
| | - Lawrence Hsieh
- Department of Neurosurgery and Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8082, USA
| | - Angélique Bordey
- Department of Neurosurgery and Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8082, USA
| |
Collapse
|
376
|
Nakashima M, Saitsu H, Takei N, Tohyama J, Kato M, Kitaura H, Shiina M, Shirozu H, Masuda H, Watanabe K, Ohba C, Tsurusaki Y, Miyake N, Zheng Y, Sato T, Takebayashi H, Ogata K, Kameyama S, Kakita A, Matsumoto N. Somatic Mutations in the MTOR gene cause focal cortical dysplasia type IIb. Ann Neurol 2015; 78:375-86. [PMID: 26018084 DOI: 10.1002/ana.24444] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 05/26/2015] [Accepted: 05/26/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Focal cortical dysplasia (FCD) type IIb is a cortical malformation characterized by cortical architectural abnormalities, dysmorphic neurons, and balloon cells. It has been suggested that FCDs are caused by somatic mutations in cells in the developing brain. Here, we explore the possible involvement of somatic mutations in FCD type IIb. METHODS We collected a total of 24 blood-brain paired samples with FCD, including 13 individuals with FCD type IIb, 5 with type IIa, and 6 with type I. We performed whole-exome sequencing using paired samples from 9 of the FCD type IIb subjects. Somatic MTOR mutations were identified and further investigated using all 24 paired samples by deep sequencing of the entire gene's coding region. Somatic MTOR mutations were confirmed by droplet digital polymerase chain reaction. The effect of MTOR mutations on mammalian target of rapamycin (mTOR) kinase signaling was evaluated by immunohistochemistry and Western blotting analyses of brain samples and by in vitro transfection experiments. RESULTS We identified four lesion-specific somatic MTOR mutations in 6 of 13 (46%) individuals with FCD type IIb showing mutant allele rates of 1.11% to 9.31%. Functional analyses showed that phosphorylation of ribosomal protein S6 in FCD type IIb brain tissues with MTOR mutations was clearly elevated, compared to control samples. Transfection of any of the four MTOR mutants into HEK293T cells led to elevated phosphorylation of 4EBP, the direct target of mTOR kinase. INTERPRETATION We found low-prevalence somatic mutations in MTOR in FCD type IIb, indicating that activating somatic mutations in MTOR cause FCD type IIb.
Collapse
Affiliation(s)
- Mitsuko Nakashima
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hirotomo Saitsu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuyuki Takei
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Jun Tohyama
- Department of Child Neurology, Nishi-Niigata Chuo National Hospital, Niigata, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Hiroki Kitaura
- Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Masaaki Shiina
- Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroshi Shirozu
- Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
| | - Hiroshi Masuda
- Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
| | - Keisuke Watanabe
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Chihiro Ohba
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yoshinori Tsurusaki
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yingjun Zheng
- Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Tatsuhiro Sato
- Division of Biochemistry, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kazuhiro Ogata
- Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shigeki Kameyama
- Department of Functional Neurosurgery, Epilepsy Center, Nishi-Niigata Chuo National Hospital, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
377
|
Kim J, Kim S, Nam H, Kim S, Lee D. SoloDel: a probabilistic model for detecting low-frequent somatic deletions from unmatched sequencing data. Bioinformatics 2015; 31:3105-13. [PMID: 26071141 DOI: 10.1093/bioinformatics/btv358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 06/05/2015] [Indexed: 01/26/2023] Open
Abstract
MOTIVATION Finding somatic mutations from massively parallel sequencing data is becoming a standard process in genome-based biomedical studies. There are a number of robust methods developed for detecting somatic single nucleotide variations However, detection of somatic copy number alteration has been substantially less explored and remains vulnerable to frequently raised sampling issues: low frequency in cell population and absence of the matched control samples. RESULTS We developed a novel computational method SoloDel that accurately classifies low-frequent somatic deletions from germline ones with or without matched control samples. We first constructed a probabilistic, somatic mutation progression model that describes the occurrence and propagation of the event in the cellular lineage of the sample. We then built a Gaussian mixture model to represent the mixed population of somatic and germline deletions. Parameters of the mixture model could be estimated using the expectation-maximization algorithm with the observed distribution of read-depth ratios at the points of discordant-read based initial deletion calls. Combined with conventional structural variation caller, SoloDel greatly increased the accuracy in classifying somatic mutations. Even without control, SoloDel maintained a comparable performance in a wide range of mutated subpopulation size (10-70%). SoloDel could also successfully recall experimentally validated somatic deletions from previously reported neuropsychiatric whole-genome sequencing data. AVAILABILITY AND IMPLEMENTATION Java-based implementation of the method is available at http://sourceforge.net/projects/solodel/ CONTACT swkim@yuhs.ac or dhlee@biosoft.kaist.ac.kr SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Junho Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 120-752, Korea, Department of Bio and Brain Engineering, KAIST, Yuseong-Gu, Daejeon 305-701, Korea
| | - Sanghyeon Kim
- Stanley Brain Research Laboratory, Stanley Medical Research Institute, Rockville, MD 20850, USA and
| | - Hojung Nam
- School of Information and Communications, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | - Sangwoo Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 120-752, Korea
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, Yuseong-Gu, Daejeon 305-701, Korea
| |
Collapse
|
378
|
Ferguson SM. Beyond indigestion: emerging roles for lysosome-based signaling in human disease. Curr Opin Cell Biol 2015; 35:59-68. [PMID: 25950843 DOI: 10.1016/j.ceb.2015.04.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/16/2015] [Accepted: 04/19/2015] [Indexed: 01/01/2023]
Abstract
Lysosomes are becoming increasingly recognized as a hub that integrates diverse signals in order to control multiple aspects of cell physiology. This is illustrated by the discovery of a growing number of lysosome-localized proteins that respond to changes in growth factor and nutrient availability to regulate mTORC1 signaling as well as the identification of MiT/TFE transcription factors (MITF, TFEB and TFE3) as proteins that shuttle between lysosomes and the nucleus to elicit a transcriptional response to ongoing changes in lysosome status. These findings have been paralleled by advances in human genetics that connect mutations in genes involved in lysosomal signaling to a broad range of human illnesses ranging from cancer to neurological disease. This review summarizes these new discoveries at the interface between lysosome cell biology and human disease.
Collapse
Affiliation(s)
- Shawn M Ferguson
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06510, United States; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, United States.
| |
Collapse
|