1
|
Mantoan Ritter L, Annear NMP, Baple EL, Ben-Chaabane LY, Bodi I, Brosson L, Cadwgan JE, Coslett B, Crosby AH, Davies DM, Daykin N, Dedeurwaerdere S, Dühring Fenger C, Dunlop EA, Elmslie FV, Girodengo M, Hambleton S, Jansen AC, Johnson SR, Kearley KC, Kingswood JC, Laaniste L, Lachlan K, Latchford A, Madsen RR, Mansour S, Mihaylov SR, Muhammed L, Oliver C, Pepper T, Rawlins LE, Schim van der Loeff I, Siddiqui A, Takhar P, Tatton-Brown K, Tee AR, Tibarewal P, Tye C, Ultanir SK, Vanhaesebroeck B, Zare B, Pal DK, Bateman JM. mTOR pathway diseases: challenges and opportunities from bench to bedside and the mTOR node. Orphanet J Rare Dis 2025; 20:256. [PMID: 40426219 PMCID: PMC12107773 DOI: 10.1186/s13023-025-03740-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a highly conserved serine/threonine kinase that regulates key cellular processes including cell growth, autophagy and metabolism. Hyperactivation of the mTOR pathway causes a group of rare and ultrarare genetic diseases. mTOR pathway diseases have diverse clinical manifestations that are managed by distinct medical disciplines but share a common underlying molecular basis. There is a now a deep understanding of the molecular underpinning that regulates the mTOR pathway but effective treatments for most mTOR pathway diseases are lacking. Translating scientific knowledge into clinical applications to benefit the unmet clinical needs of patients is a major challenge common to many rare diseases. In this article we expound how mTOR pathway diseases provide an opportunity to coordinate basic and translational disease research across the group, together with industry, medical research foundations, charities and patient groups, by pooling expertise and driving progress to benefit patients. We outline the germline and somatic mutations in the mTOR pathway that cause rare diseases and summarise the prevalence, genetic basis, clinical manifestations, pathophysiology and current treatments for each disease in this group. We describe the challenges and opportunities for progress in elucidating the underlying mechanisms, improving diagnosis and prognosis, as well as the development and approval of new therapies for mTOR pathway diseases. We illustrate the crucial role of patient public involvement and engagement in rare disease and mTOR pathway disease research. Finally, we explain how the mTOR Pathway Diseases node, part of the Research Disease Research UK Platform, will address these challenges to improve the understanding, diagnosis and treatment of mTOR pathway diseases.
Collapse
Affiliation(s)
- Laura Mantoan Ritter
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | - Nicholas M P Annear
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | - Leila Y Ben-Chaabane
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | - Istvan Bodi
- King's College Hospital NHS Foundation Trust, London, UK
| | | | | | | | | | | | | | | | | | | | - Frances V Elmslie
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | - Marie Girodengo
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
- The Francis Crick Institute, London, UK
| | - Sophie Hambleton
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Simon R Johnson
- Centre for Respiratory Research, NIHR Nottingham Biomedical Research Centre and Biodiscovery Institute, Translational Medical Sciences, University of Nottingham, Nottingham, UK
| | - Kelly C Kearley
- mTOR Node Advisory Panel (MAP), London, UK
- PTEN UK and Ireland Patient Group, London, UK
| | - John C Kingswood
- St George's University Hospitals NHS Foundation Trust, London, UK
| | | | - Katherine Lachlan
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Andrew Latchford
- Polyposis Registry, St Mark's Hospital, London, UK
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Sahar Mansour
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | | | | | - Tom Pepper
- PTEN Research, Cheltenham, Gloucestershire, UK
| | | | - Ina Schim van der Loeff
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ata Siddiqui
- King's College Hospital NHS Foundation Trust, London, UK
| | | | - Katrina Tatton-Brown
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | | | - Charlotte Tye
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | | | | | | | - Deb K Pal
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | - Joseph M Bateman
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK.
| |
Collapse
|
2
|
Boff MO, Xavier FAC, Diz FM, Gonçalves JB, Ferreira LM, Zambeli J, Pazzin DB, Previato TTR, Erwig HS, Gonçalves JIB, Bruzzo FTK, Marinowic D, da Costa JC, Zanirati G. mTORopathies in Epilepsy and Neurodevelopmental Disorders: The Future of Therapeutics and the Role of Gene Editing. Cells 2025; 14:662. [PMID: 40358185 PMCID: PMC12071303 DOI: 10.3390/cells14090662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 05/15/2025] Open
Abstract
mTORopathies represent a group of neurodevelopmental disorders linked to dysregulated mTOR signaling, resulting in conditions such as tuberous sclerosis complex, focal cortical dysplasia, hemimegalencephaly, and Smith-Kingsmore Syndrome. These disorders often manifest with epilepsy, cognitive impairments, and, in some cases, structural brain anomalies. The mTOR pathway, a central regulator of cell growth and metabolism, plays a crucial role in brain development, where its hyperactivation leads to abnormal neuroplasticity, tumor formation, and heightened neuronal excitability. Current treatments primarily rely on mTOR inhibitors, such as rapamycin, which reduce seizure frequency and tumor size but fail to address underlying genetic causes. Advances in gene editing, particularly via CRISPR/Cas9, offer promising avenues for precision therapies targeting the genetic mutations driving mTORopathies. New delivery systems, including viral and non-viral vectors, aim to enhance the specificity and efficacy of these therapies, potentially transforming the management of these disorders. While gene editing holds curative potential, challenges remain concerning delivery, long-term safety, and ethical considerations. Continued research into mTOR mechanisms and innovative gene therapies may pave the way for transformative, personalized treatments for patients affected by these complex neurodevelopmental conditions.
Collapse
Affiliation(s)
- Marina Ottmann Boff
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Fernando Antônio Costa Xavier
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Medicine and Health Sciences, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Fernando Mendonça Diz
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Júlia Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Laura Meireles Ferreira
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Jean Zambeli
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, University of the Valley of the Rio dos Sinos (UNISINOS), São Leopoldo 93022-750, RS, Brazil
| | - Douglas Bottega Pazzin
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Thales Thor Ramos Previato
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Helena Scartassini Erwig
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Health and Life, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Fernanda Thays Konat Bruzzo
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Daniel Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Health and Life, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Jaderson Costa da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Gabriele Zanirati
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| |
Collapse
|
3
|
Liu J, Huang R, Tang F, Ma Y, Kwan P. A missense variant in DEPDC5 resulted in abnormal morphology and increased seizure susceptibility and mortality through regulating mTOR signaling. Neurobiol Dis 2025; 207:106842. [PMID: 39954744 DOI: 10.1016/j.nbd.2025.106842] [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: 06/28/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025] Open
Abstract
Dishevelled, Egl-10 and Pleckstrin domain-containing 5 (DEPDC5), a key inhibitor of the mammalian/mechanistic target of rapamycin (mTOR) pathway, is frequently associated with epilepsy. However, the functional consequences of most DEPDC5 variants rely on in silico predictions and have not been experimentally confirmed.This study aimed to determine the functional consequences of a DEPDC5 variant identified in patients with epilepsy across multiple generations in a Chinese family. We identified a missense heterozygous variant (c. 2055C > A; p. Phe685Leu) in DEPDC5 in Chinese family affected by epilepsy across three generations. This variant has not been previously reported in the Chinese population. Primary neuron cultures transfected with the mutant plasmid exhibited altered subcellular localization. To explore the mechanisms of epilepsy linked to this variant, we created nervous system-specific conditional human DEPDC5 knock-in mouse using Cre-recombination under the Nestin promotor (hDEPDC5WT mice, hDEPDC5F685L mice). Compared to wildtype (WT) and hDEPDC5WT mice, hDEPDC5F685L mice exhibited histological signs of mTOR hyperactivation, enlarged neuronal soma, abnormal neurons, and heightened susceptibility to seizures and mortality. Administering rapamycin to hDEPDC5F685L mice starting two weeks after birth normalized neuronal size and mTOR activity, decreased seizure susceptibility and mortality, and showed no effects in the WT or hDEPDC5WT mice. Collectively, these results indicate that the DEPDC5 variant causes abnormal morphology and increased seizure vulnerability through modulation of mTOR signaling.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Rui Huang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Fenglin Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yuanlin Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China; Chongqing Key Laboratory of Cerebrovascular Disease Research, Chongqing, 402160, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, 402160, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China.
| | - Patrick Kwan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Epilepsy Unit, Brain Program, Alfred Hospital, Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
Shao W, Liu L, Gu J, Yang Y, Wu Y, Zhang Z, Xu Q, Wang Y, Shen Y, Gu L, Cheng Y, Zhang H. Spotlight on mechanism of sudden unexpected death in epilepsy in Dravet syndrome. Transl Psychiatry 2025; 15:84. [PMID: 40097380 PMCID: PMC11914262 DOI: 10.1038/s41398-025-03304-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 02/17/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
Dravet syndrome (DS) is a severe and catastrophic epilepsy with childhood onset. The incidence and prevalence of sudden unexpected death in epilepsy (SUDEP) are significantly higher in DS patients than in general epileptic populations. Although extensive research conducted, the underlying mechanisms of SUDEP occurring in DS patients remain unclear. This review focuses on the link between DS and SUDEP and analyzes the potential pathogenesis. We summarize the genetic basis of DS and SUDEP and elucidate the pathophysiological mechanisms of SUDEP in DS. Furthermore, given the drug-resistant nature of this disorder, the pharmacological approach has limited efficacy and often causes side effects, therefore, the non-pharmacological approaches and precise treatment can reduce the risk of SUDEP in this condition, open a new window to cure this disease, and provide a widened landscape of treatment options for patients.
Collapse
Affiliation(s)
- WeiHui Shao
- Department of Anesthesiology, the Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Lu Liu
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - JiaXuan Gu
- Department of Anesthesiology, the Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Yue Yang
- Department of Anesthesiology, the Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - YaXuan Wu
- Department of Anesthesiology, the Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - ZhuoYue Zhang
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qing Xu
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - YuLing Wang
- Department of Anesthesiology, the Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Yue Shen
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, 310006, China
| | - LeYuan Gu
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yuan Cheng
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, 310006, China.
| | - HongHai Zhang
- Department of Anesthesiology, the Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China.
- Department of Anesthesiology, Zhejiang University School of Medicine, Hangzhou, 310006, China.
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
5
|
Moloney PB, Delanty N. An overview of the value of mTOR inhibitors to the treatment of epilepsy: the evidence to date. Expert Rev Neurother 2025:1-17. [PMID: 39903448 DOI: 10.1080/14737175.2025.2462280] [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: 10/17/2024] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/06/2025]
Abstract
INTRODUCTION Dysregulated mechanistic target of rapamycin (mTOR) activity is implicated in seizure development in epilepsies caused by variants in mTOR pathway genes. Sirolimus and everolimus, allosteric mTOR inhibitors, are widely used in transplant medicine and oncology. Everolimus is approved for treating seizures in tuberous sclerosis complex (TSC), the prototype mTORopathy. Emerging evidence suggests that mTOR inhibitors could also be effective in other mTORopathies, such as DEPDC5-related epilepsy and focal cortical dysplasia type 2 (FCD2). AREAS COVERED This narrative review summarizes key regulatory proteins in the mTOR cascade and outlines epilepsy syndromes linked to variants in genes encoding these proteins, particularly TSC, GATOR1-related epilepsies, and FCD2. It discusses the clinical pharmacology of mTOR inhibitors and the evidence supporting their efficacy as antiseizure medications (ASM) in mTORopathies. Lastly, potential benefits of next-generation mTOR inhibitors for CNS indications are evaluated. EXPERT OPINION The therapeutic benefits of mTOR inhibitors in TSC are well-established, but their value in other mTORopathies remains uncertain. Despite targeting the underlying disease biology, their efficacy in TSC is not significantly different from other ASM, likely due in part to pharmacokinetic constraints. Next-generation mTOR inhibitors that address these limitations may offer improved response rates, but they are in the preclinical development phase.
Collapse
Affiliation(s)
- Patrick B Moloney
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Epilepsy, Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Norman Delanty
- Department of Neurology, Beaumont Hospital, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Research Ireland FutureNeuro Centre, Dublin, Ireland
| |
Collapse
|
6
|
Dentel B, Angeles-Perez L, Flores AY, Lei K, Ren C, Sanchez AP, Tsai PT. Neuronal cell type specific roles for Nprl2 in neurodevelopmental disorder-relevant behaviors. Neurobiol Dis 2025; 205:106790. [PMID: 39765274 DOI: 10.1016/j.nbd.2025.106790] [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: 05/20/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Loss of function in the subunits of the GTPase-activating protein (GAP) activity toward Rags-1 (GATOR1) complex, an amino-acid sensitive negative regulator of the mechanistic target of rapamycin complex 1 (mTORC1), is implicated in both genetic familial epilepsies and Neurodevelopmental Disorders (NDDs) (Baldassari et al., 2018). Previous studies have found seizure phenotypes and increased activity resulting from conditional deletion of GATOR1 function from forebrain excitatory neurons (Yuskaitis et al., 2018; Dentel et al., 2022); however, studies focused on understanding mechanisms contributing to NDD-relevant behaviors are lacking, especially studies understanding the contributions of GATOR1's critical GAP catalytic subunit, nitrogen permease regulator like-2 (Nprl2). Given the clinical phenotypes observed in patients with Nprl2 mutations, in this study, we sought to investigate the neuronal cell type contributions of Nprl2 to NDD behaviors. We conditionally deleted Nprl2 broadly in most neurons (Synapsin1cre), in inhibitory neurons only (Vgatcre), and in Purkinje cells within the cerebellum (L7cre). Broad neuronal deletion of Nprl2 resulted in seizures, social and learning deficits, and hyperactivity. In contrast, deleting Nprl2 from inhibitory neurons led to increased motor learning, hyperactive behavior, in addition to social and learning deficits. Lastly, Purkinje cell (PC) loss of Nprl2 also led to learning and social deficits but did not affect locomotor activity. These phenotypes enhance understanding of the spectrum of disease found in human populations with GATOR1 loss of function and highlight the significance of distinct cellular populations to NDD-related behaviors. SIGNIFICANCE STATEMENT: We aim to elucidate the neuronal-specific contributions of nitrogen permease regulator like-2 (Nprl2) to its neurodevelopmental disorder (NDD)-relevant phenotypes. We conditionally deleted Nprl2 broadly in neurons (Syn1cre), in inhibitory neurons (Vgatcre), and in cerebellar Purkinje cells (L7cre). We identify seizures only in the Syn1cre conditional mutant (cKO); hyperactivity, learning difficulties, social deficits, and impulsivity in the Syn1cre and Vgatcre cKOs; and social deficits, and fear learning deficits in L7cre cKOs. To our knowledge, we are the first to describe the behavioral contributions of Nprl2's function across multiple cell types. Our findings highlight both critical roles for Nprl2 in learning and behavior and also distinct contributions of select neuronal populations to these NDD-relevant behaviors.
Collapse
Affiliation(s)
- Brianne Dentel
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Lidiette Angeles-Perez
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Abigail Y Flores
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Katherine Lei
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Chongyu Ren
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Andrea Pineda Sanchez
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Peter T Tsai
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Psychiatry, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Pediatrics, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Neuroscience; O'Donnell Brain Institute, Dallas, TX, United States of America.
| |
Collapse
|
7
|
Sambri I, Ferniani M, Ballabio A. Ragopathies and the rising influence of RagGTPases on human diseases. Nat Commun 2024; 15:5812. [PMID: 38987251 PMCID: PMC11237164 DOI: 10.1038/s41467-024-50034-4] [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: 02/23/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024] Open
Abstract
RagGTPases (Rags) play an essential role in the regulation of cell metabolism by controlling the activities of both mechanistic target of rapamycin complex 1 (mTORC1) and Transcription factor EB (TFEB). Several diseases, herein named ragopathies, are associated to Rags dysfunction. These diseases may be caused by mutations either in genes encoding the Rags, or in their upstream regulators. The resulting phenotypes may encompass a variety of clinical features such as cataract, kidney tubulopathy, dilated cardiomyopathy and several types of cancer. In this review, we focus on the key clinical, molecular and physio-pathological features of ragopathies, aiming to shed light on their underlying mechanisms.
Collapse
Affiliation(s)
- Irene Sambri
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, (NA), Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program (GEM), Naples, Italy
| | - Marco Ferniani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, (NA), Italy.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy.
| |
Collapse
|
8
|
Okoh J, Mays J, Bacq A, Oses-Prieto JA, Tyanova S, Chen CJ, Imanbeyev K, Doladilhe M, Zhou H, Jafar-Nejad P, Burlingame A, Noebels J, Baulac S, Costa-Mattioli M. Targeted suppression of mTORC2 reduces seizures across models of epilepsy. Nat Commun 2023; 14:7364. [PMID: 37963879 PMCID: PMC10645975 DOI: 10.1038/s41467-023-42922-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023] Open
Abstract
Epilepsy is a neurological disorder that poses a major threat to public health. Hyperactivation of mTOR complex 1 (mTORC1) is believed to lead to abnormal network rhythmicity associated with epilepsy, and its inhibition is proposed to provide some therapeutic benefit. However, mTOR complex 2 (mTORC2) is also activated in the epileptic brain, and little is known about its role in seizures. Here we discover that genetic deletion of mTORC2 from forebrain neurons is protective against kainic acid-induced behavioral and EEG seizures. Furthermore, inhibition of mTORC2 with a specific antisense oligonucleotide robustly suppresses seizures in several pharmacological and genetic mouse models of epilepsy. Finally, we identify a target of mTORC2, Nav1.2, which has been implicated in epilepsy and neuronal excitability. Our findings, which are generalizable to several models of human seizures, raise the possibility that inhibition of mTORC2 may serve as a broader therapeutic strategy against epilepsy.
Collapse
Affiliation(s)
- James Okoh
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX, USA
- Altos Labs Inc, Bay Area Institute, Redwood City, CA, USA
| | - Jacqunae Mays
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Alexandre Bacq
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, F-75013, Paris, France
| | - Juan A Oses-Prieto
- Departments of Chemistry and Pharmaceutical Chemistry, University of California San Fransisco, San Fransisco, CA, USA
| | - Stefka Tyanova
- Altos Labs Inc, Bay Area Institute, Redwood City, CA, USA
| | - Chien-Ju Chen
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX, USA
- Novartis Inc, Boston, MA, USA
| | - Khalel Imanbeyev
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Marion Doladilhe
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, F-75013, Paris, France
| | - Hongyi Zhou
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX, USA
- Altos Labs Inc, Bay Area Institute, Redwood City, CA, USA
| | | | - Alma Burlingame
- Departments of Chemistry and Pharmaceutical Chemistry, University of California San Fransisco, San Fransisco, CA, USA
| | - Jeffrey Noebels
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Stephanie Baulac
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, F-75013, Paris, France
| | - Mauro Costa-Mattioli
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX, USA.
- Altos Labs Inc, Bay Area Institute, Redwood City, CA, USA.
| |
Collapse
|
9
|
Kao HY, Yao Y, Yang T, Ziobro J, Zylinski M, Mir MY, Hu S, Cao R, Borna NN, Banerjee R, Parent JM, Wang S, Leventhal DK, Li P, Wang Y. Sudden Unexpected Death in Epilepsy and Respiratory Defects in a Mouse Model of DEPDC5-Related Epilepsy. Ann Neurol 2023; 94:812-824. [PMID: 37606181 PMCID: PMC10592102 DOI: 10.1002/ana.26773] [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: 01/19/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023]
Abstract
OBJECTIVES DEPDC5 is a common causative gene in familial focal epilepsy with or without malformations of cortical development. Its pathogenic variants also confer a significantly higher risk for sudden unexpected death in epilepsy (SUDEP), providing opportunities to investigate the pathophysiology intersecting neurodevelopment, epilepsy, and cardiorespiratory function. There is an urgent need to gain a mechanistic understanding of DEPDC5-related epilepsy and SUDEP, identify biomarkers for patients at high risk, and develop preventive interventions. METHODS Depdc5 was specifically deleted in excitatory or inhibitory neurons in the mouse brain to determine neuronal subtypes that drive epileptogenesis and SUDEP. Electroencephalogram (EEG), cardiac, and respiratory recordings were performed to determine cardiorespiratory phenotypes associated with SUDEP. Baseline respiratory function and the response to hypoxia challenge were also studied in these mice. RESULTS Depdc5 deletion in excitatory neurons in cortical layer 5 and dentate gyrus caused frequent generalized tonic-clonic seizures and SUDEP in young adult mice, but Depdc5 deletion in cortical interneurons did not. EEG suppression immediately following ictal offset was observed in fatal and non-fatal seizures, but low amplitude rhythmic theta frequency activity was lost only in fatal seizures. In addition, these mice developed baseline respiratory dysfunction prior to SUDEP, during which ictal apnea occurred long before terminal cardiac asystole. INTERPRETATION Depdc5 deletion in excitatory neurons is sufficient to cause DEPDC5-related epilepsy and SUDEP. Ictal apnea and respiratory dysregulation play critical roles in SUDEP. Our study also provides a novel mouse model to investigate the underlying mechanisms of DEPDC5-related epilepsy and SUDEP. ANN NEUROL 2023;94:812-824.
Collapse
Affiliation(s)
- Hsin-Yi Kao
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Yilong Yao
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Tao Yang
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Julie Ziobro
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Mary Zylinski
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Mohd Yaqub Mir
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Shuntong Hu
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Runnan Cao
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Rajat Banerjee
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Jack M. Parent
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Michgian Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Shuo Wang
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Daniel K. Leventhal
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Parkinson Disease Foundation Research Center of Excellence, University of Michigan, Ann Arbor, MI, USA
| | - Peng Li
- Michgian Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biologic and Material Sciences, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Yu Wang
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Michgian Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
10
|
Moloney PB, Kearney H, Benson KA, Costello DJ, Cavalleri GL, Gorman KM, Lynch BJ, Delanty N. Everolimus precision therapy for the GATOR1-related epilepsies: A case series. Eur J Neurol 2023; 30:3341-3346. [PMID: 37422919 DOI: 10.1111/ene.15975] [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: 04/28/2023] [Revised: 06/19/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Pathogenic variants in the GAP activity towards RAGs 1 (GATOR1) complex genes (DEPDC5, NPRL2, NPRL3) cause focal epilepsy through hyperactivation of the mechanistic target of rapamycin pathway. We report our experience using everolimus in patients with refractory GATOR1-related epilepsy. METHODS We performed an open-label observational study of everolimus for drug-resistant epilepsy caused by variants in DEPDC5, NPRL2 and NPRL3. Everolimus was titrated to a target serum concentration (5-15 ng/mL). The primary outcome measure was change in mean monthly seizure frequency compared with baseline. RESULTS Five patients were treated with everolimus. All had highly active (median baseline seizure frequency, 18/month) and refractory focal epilepsy (failed 5-16 prior anti-seizure medications). Four had DEPDC5 variants (three loss-of-function, one missense) and one had a NPRL3 splice-site variant. All patients with DEPDC5 loss-of-function variants had significantly reduced seizures (74.3%-86.1%), although one stopped everolimus after 12 months due to psychiatric symptoms. Everolimus was less effective in the patient with a DEPDC5 missense variant (43.9% seizure frequency reduction). The patient with NPRL3-related epilepsy had seizure worsening. The most common adverse event was stomatitis. CONCLUSIONS Our study provides the first human data on the potential benefit of everolimus precision therapy for epilepsy caused by DEPDC5 loss-of-function variants. Further studies are needed to support our findings.
Collapse
Affiliation(s)
- Patrick B Moloney
- School of Pharmacy and Biomolecular Sciences, The Royal College of Surgeons in Ireland, Dublin, Ireland
- FutureNeuro SFI Research Centre, Dublin, Ireland
- Blackrock Clinic, Dublin, Ireland
- Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Hugh Kearney
- FutureNeuro SFI Research Centre, Dublin, Ireland
- Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Katherine A Benson
- School of Pharmacy and Biomolecular Sciences, The Royal College of Surgeons in Ireland, Dublin, Ireland
- FutureNeuro SFI Research Centre, Dublin, Ireland
| | - Daniel J Costello
- FutureNeuro SFI Research Centre, Dublin, Ireland
- Department of Neurology, Cork University Hospital and College of Medicine and Health, University College Cork, Cork, Ireland
| | - Gianpiero L Cavalleri
- School of Pharmacy and Biomolecular Sciences, The Royal College of Surgeons in Ireland, Dublin, Ireland
- FutureNeuro SFI Research Centre, Dublin, Ireland
| | - Kathleen M Gorman
- Department of Neurology, Children's Health Ireland at Temple Street, Dublin, Ireland
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Bryan J Lynch
- Department of Neurology, Children's Health Ireland at Temple Street, Dublin, Ireland
| | - Norman Delanty
- School of Pharmacy and Biomolecular Sciences, The Royal College of Surgeons in Ireland, Dublin, Ireland
- FutureNeuro SFI Research Centre, Dublin, Ireland
- Department of Neurology, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
11
|
Bagnall RD, Perucca P. ILAE Genetic Literacy Series: Postmortem Genetic Testing in Sudden Unexpected Death in Epilepsy. Epileptic Disord 2023; 25:472-479. [PMID: 37340991 DOI: 10.1002/epd2.20090] [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: 03/15/2023] [Revised: 05/31/2023] [Accepted: 06/17/2023] [Indexed: 06/22/2023]
Abstract
A 24-year-old man with non-lesional bitemporal lobe epilepsy since age 16 years was found dead in bed around midday. He was last seen the previous night when he was witnessed to have a tonic-clonic seizure. Before his death, he was experiencing weekly focal impaired awareness seizures and up to two focal-to-bilateral tonic-clonic seizures each year. He had trialed several antiseizure medications and was on levetiracetam 1500 mg/day, lamotrigine 400 mg/day, and clobazam 10 mg/day at the time of death. Other than epilepsy, his medical history was unremarkable. Of note, he had an older brother with a history of febrile seizures and a paternal first cousin with epilepsy. No cause of death was identified following a comprehensive postmortem investigation. The coroner classified the death as "sudden unexpected death in epilepsy" (SUDEP), and it would qualify as "definite SUDEP" using the current definitions.1 This left the family with many questions unanswered; in particular, they wish to know what caused the death and whether it could happen to other family members. Could postmortem genetic testing identify a cause of death, provide closure to the family, and facilitate cascade genetic testing of first-degree family members who may be at risk of sudden death? While grieving family members struggle with uncertainty about the cause of death, we as clinicians also face similar uncertainties about genetic contributions to SUDEP, especially when the literature is sparse, and the utility of genetic testing is still being worked out. We aim to shed some light on this topic, highlighting areas where data is emerging but also areas where uncertainty remains, keeping our case in mind as we examine this clinically important area.
Collapse
Affiliation(s)
- Richard D Bagnall
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Piero Perucca
- Department of Medicine (Austin Health), Epilepsy Research Centre, The University of Melbourne, Melbourne, Victoria, Australia
- Bladin-Berkovic Comprehensive Epilepsy Program, Department of Neurology, Austin Health, Melbourne, Victoria, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
- Department of Neurology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Mannick JB, Lamming DW. Targeting the biology of aging with mTOR inhibitors. NATURE AGING 2023; 3:642-660. [PMID: 37142830 PMCID: PMC10330278 DOI: 10.1038/s43587-023-00416-y] [Citation(s) in RCA: 139] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/07/2023] [Indexed: 05/06/2023]
Abstract
Inhibition of the protein kinase mechanistic target of rapamycin (mTOR) with the Food and Drug Administration (FDA)-approved therapeutic rapamycin promotes health and longevity in diverse model organisms. More recently, specific inhibition of mTORC1 to treat aging-related conditions has become the goal of basic and translational scientists, clinicians and biotechnology companies. Here, we review the effects of rapamycin on the longevity and survival of both wild-type mice and mouse models of human diseases. We discuss recent clinical trials that have explored whether existing mTOR inhibitors can safely prevent, delay or treat multiple diseases of aging. Finally, we discuss how new molecules may provide routes to the safer and more selective inhibition of mTOR complex 1 (mTORC1) in the decade ahead. We conclude by discussing what work remains to be done and the questions that will need to be addressed to make mTOR inhibitors part of the standard of care for diseases of aging.
Collapse
Affiliation(s)
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
13
|
Auvin S, Baulac S. mTOR-therapy and targeted treatment opportunities in mTOR-related epilepsies associated with cortical malformations. Rev Neurol (Paris) 2023; 179:337-344. [PMID: 36906459 DOI: 10.1016/j.neurol.2022.12.007] [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: 10/27/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 03/11/2023]
Abstract
Dysregulation of the mTOR pathway is now well documented in several neurodevelopmental disorders associated with epilepsy. Mutations of mTOR pathway genes are involved in tuberous sclerosis complex (TSC) as well as in a range of cortical malformations from hemimegalencephaly (HME) to type II focal cortical dysplasia (FCD II), leading to the concept of "mTORopathies" (mTOR pathway-related malformations). This suggests that mTOR inhibitors (notably rapamycin (sirolimus), and everolimus) could be used as antiseizure medication. In this review, we provide an overview of pharmacological treatments targeting the mTOR pathway for epilepsy based on lectures from the ILAE French Chapter meeting in October 2022 in Grenoble. There is strong preclinical evidence for the antiseizure effects of mTOR inhibitors in TSC and cortical malformation mouse models. There are also open studies on the antiseizure effects of mTOR inhibitors, as well as one phase III study showing the antiseizure effect of everolimus in TSC patients. Finally, we discuss to which extent mTOR inhibitors might have properties beyond the antiseizure effect on associated neuropsychiatric comorbidities. We also discuss a new way of treatment on the mTOR pathways.
Collapse
Affiliation(s)
- S Auvin
- Service de neurologie pédiatrique, EpiCARE ERN membre, Hôpital Robert Debré, AP-HP, Paris, France; Université Paris-Cité, Inserm NeuroDiderot, Paris, France; Institut Universitaire de France (IUF), Paris, France.
| | - S Baulac
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpital de la Pitié Salpêtrière, 75013 Paris, France
| |
Collapse
|
14
|
Kovačević M, Janković M, Branković M, Milićević O, Novaković I, Sokić D, Ristić A, Shamsani J, Vojvodić N. Novel GATOR1 variants in focal epilepsy. Epilepsy Behav 2023; 141:109139. [PMID: 36848747 DOI: 10.1016/j.yebeh.2023.109139] [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: 12/07/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/27/2023]
Abstract
INTRODUCTION Variants in GATOR1 genes are well established in focal epilepsy syndromes. A strong association of GATOR1 variants with drug-resistant epilepsy as well as an increased risk of sudden unexplained death in epilepsy warrants developing strategies to facilitate the identification of patients who could potentially benefit from genetic testing and precision medicine. We aimed to determine the yield of GATOR1 gene sequencing in patients with focal epilepsy typically referred for genetic testing, establish novel GATOR1 variants and determine clinical, electroencephalographic, and radiological characteristics of variant carriers. PATIENTS AND METHODS Ninety-six patients with clinical suspicion of genetic focal epilepsy with previous comprehensive diagnostic epilepsy evaluation in The Neurology Clinic, University Clinical Center of Serbia, were included in the study. Sequencing was performed using a custom gene panel encompassing DEPDC5, NPRL2, and NPRL3. Variants of interest (VOI) were classified according to criteria proposed by the American College of Medical Genetics and the Association for Molecular Pathology. RESULTS Four previously unreported VOI in 4/96 (4.2%) patients were found in our cohort. Three likely pathogenic variants were determined in 3/96 (3.1%) patients, one frameshift variant in DEPDC5 in a patient with nonlesional frontal lobe epilepsy, one splicogenic DEPDC5 variant in a patient with nonlesional posterior quadrant epilepsy, and one frameshift variant in NPRL2 in a patient with temporal lobe epilepsy associated with hippocampal sclerosis. Only one VOI, a missense variant in NPRL3, found in 1/96 (1.1%) patients, was classified as a variant of unknown significance. CONCLUSION GATOR1 gene sequencing was diagnostic in 3.1% of our cohort and revealed three novel likely pathogenic variants, including a previously unreported association of temporal lobe epilepsy with hippocampal sclerosis with an NPRL2 variant. Further research is essential for a better understanding of the clinical scope of GATOR1 gene-associated epilepsy.
Collapse
Affiliation(s)
- Maša Kovačević
- Neurology Clinic, University Clinical Center of Serbia, Serbia; Faculty of Medicine, University of Belgrade, Serbia.
| | - Milena Janković
- Neurology Clinic, University Clinical Center of Serbia, Serbia
| | | | | | | | - Dragoslav Sokić
- Neurology Clinic, University Clinical Center of Serbia, Serbia; Faculty of Medicine, University of Belgrade, Serbia
| | - Aleksandar Ristić
- Neurology Clinic, University Clinical Center of Serbia, Serbia; Faculty of Medicine, University of Belgrade, Serbia
| | | | - Nikola Vojvodić
- Neurology Clinic, University Clinical Center of Serbia, Serbia; Faculty of Medicine, University of Belgrade, Serbia
| |
Collapse
|
15
|
Sun S, Wang H. Clocking Epilepsies: A Chronomodulated Strategy-Based Therapy for Rhythmic Seizures. Int J Mol Sci 2023; 24:4223. [PMID: 36835631 PMCID: PMC9962262 DOI: 10.3390/ijms24044223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Epilepsy is a neurological disorder characterized by hypersynchronous recurrent neuronal activities and seizures, as well as loss of muscular control and sometimes awareness. Clinically, seizures have been reported to display daily variations. Conversely, circadian misalignment and circadian clock gene variants contribute to epileptic pathogenesis. Elucidation of the genetic bases of epilepsy is of great importance because the genetic variability of the patients affects the efficacies of antiepileptic drugs (AEDs). For this narrative review, we compiled 661 epilepsy-related genes from the PHGKB and OMIM databases and classified them into 3 groups: driver genes, passenger genes, and undetermined genes. We discuss the potential roles of some epilepsy driver genes based on GO and KEGG analyses, the circadian rhythmicity of human and animal epilepsies, and the mutual effects between epilepsy and sleep. We review the advantages and challenges of rodents and zebrafish as animal models for epileptic studies. Finally, we posit chronomodulated strategy-based chronotherapy for rhythmic epilepsies, integrating several lines of investigation for unraveling circadian mechanisms underpinning epileptogenesis, chronopharmacokinetic and chronopharmacodynamic examinations of AEDs, as well as mathematical/computational modeling to help develop time-of-day-specific AED dosing schedules for rhythmic epilepsy patients.
Collapse
Affiliation(s)
- Sha Sun
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou 215123, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215123, China
| |
Collapse
|
16
|
Emx1-Cre Is Expressed in Peripheral Autonomic Ganglia That Regulate Central Cardiorespiratory Functions. eNeuro 2022; 9:ENEURO.0093-22.2022. [PMID: 36192157 PMCID: PMC9581573 DOI: 10.1523/eneuro.0093-22.2022] [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: 02/24/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 12/15/2022] Open
Abstract
The Emx1-IRES-Cre transgenic mouse is commonly used to direct genetic recombination in forebrain excitatory neurons. However, the original study reported that Emx1-Cre is also expressed embryonically in peripheral autonomic ganglia, which could potentially affect the interpretation of targeted circuitry contributing to systemic phenotypes. Here, we report that Emx1-Cre is expressed in the afferent vagus nerve system involved in autonomic cardiorespiratory regulatory pathways. Our imaging studies revealed expression of Emx1-Cre driven tdtomato fluorescence in the afferent vagus nerve innervating the dorsal medulla of brainstem, cell bodies in the nodose ganglion, and their potential target structures at the carotid bifurcation such as the carotid sinus and the superior cervical ganglion (SCG). Photostimulation of the afferent terminals in the nucleus tractus solitarius (NTS) in vitro using Emx1-Cre driven ChR2 reliably evoked EPSCs in the postsynaptic neurons with electrophysiological characteristics consistent with the vagus afferent nerves. In addition, optogenetic stimulation targeting the Emx1-Cre expressing structures identified in this study, such as vagus nerve, carotid bifurcation, and the dorsal medulla surface transiently depressed cardiorespiratory rate in urethane anesthetized mice in vivo Together, our study demonstrates that Emx1-IRES-Cre is expressed in the key peripheral autonomic nerve system and can modulate cardiorespiratory function independently of forebrain expression. These results raise caution when interpreting systemic phenotypes of Emx1-IRES-Cre conditional recombinant mice, and also suggest the utility of this line to investigate modulators of the afferent vagal system.
Collapse
|
17
|
Yuskaitis CJ, Modasia JB, Schrötter S, Rossitto LA, Groff KJ, Morici C, Mithal DS, Chakrabarty RP, Chandel NS, Manning BD, Sahin M. DEPDC5-dependent mTORC1 signaling mechanisms are critical for the anti-seizure effects of acute fasting. Cell Rep 2022; 40:111278. [PMID: 36044864 PMCID: PMC9508617 DOI: 10.1016/j.celrep.2022.111278] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/11/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022] Open
Abstract
Caloric restriction and acute fasting are known to reduce seizures but through unclear mechanisms. mTOR signaling has been suggested as a potential mechanism for seizure protection from fasting. We demonstrate that brain mTORC1 signaling is reduced after acute fasting of mice and that neuronal mTORC1 integrates GATOR1 complex-mediated amino acid and tuberous sclerosis complex (TSC)-mediated growth factor signaling. Neuronal mTORC1 is most sensitive to withdrawal of leucine, arginine, and glutamine, which are dependent on DEPDC5, a component of the GATOR1 complex. Metabolomic analysis reveals that Depdc5 neuronal-specific knockout mice are resistant to sensing significant fluctuations in brain amino acid levels after fasting. Depdc5 neuronal-specific knockout mice are resistant to the protective effects of fasting on seizures or seizure-induced death. These results establish that acute fasting reduces seizure susceptibility in a DEPDC5-dependent manner. Modulation of nutrients upstream of GATOR1 and mTORC1 could offer a rational therapeutic strategy for epilepsy treatment.
Collapse
Affiliation(s)
- Christopher J Yuskaitis
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jinita B Modasia
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sandra Schrötter
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Leigh-Ana Rossitto
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Karenna J Groff
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher Morici
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Divakar S Mithal
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Section of Neurology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Ram P Chakrabarty
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Navdeep S Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Brendan D Manning
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
18
|
Wang G, Wu W, Xu Y, Yang Z, Xiao B, Long L. Imaging Genetics in Epilepsy: Current Knowledge and New Perspectives. Front Mol Neurosci 2022; 15:891621. [PMID: 35706428 PMCID: PMC9189397 DOI: 10.3389/fnmol.2022.891621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/06/2022] [Indexed: 12/11/2022] Open
Abstract
Epilepsy is a neurological network disease with genetics playing a much greater role than was previously appreciated. Unfortunately, the relationship between genetic basis and imaging phenotype is by no means simple. Imaging genetics integrates multidimensional datasets within a unified framework, providing a unique opportunity to pursue a global vision for epilepsy. This review delineates the current knowledge of underlying genetic mechanisms for brain networks in different epilepsy syndromes, particularly from a neural developmental perspective. Further, endophenotypes and their potential value are discussed. Finally, we highlight current challenges and provide perspectives for the future development of imaging genetics in epilepsy.
Collapse
Affiliation(s)
- Ge Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Epileptic Disease of Hunan Province, Central South University, Changsha, China
| | - Wenyue Wu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Yuchen Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhuanyi Yang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Epileptic Disease of Hunan Province, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Epileptic Disease of Hunan Province, Central South University, Changsha, China
| | - Lili Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Epileptic Disease of Hunan Province, Central South University, Changsha, China
- *Correspondence: Lili Long
| |
Collapse
|
19
|
Dentel B, Angeles-Perez L, Ren C, Jakkamsetti V, Holley AJ, Caballero D, Oh E, Gibson J, Pascual JM, Huber KM, Tu BP, Tsai PT. Increased glycine contributes to synaptic dysfunction and early mortality in Nprl2 seizure model. iScience 2022; 25:104334. [PMID: 35602938 PMCID: PMC9118754 DOI: 10.1016/j.isci.2022.104334] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 09/16/2021] [Accepted: 04/26/2022] [Indexed: 10/25/2022] Open
Abstract
Targeted therapies for epilepsies associated with the mTORC1 signaling negative regulator GATOR1 are lacking. NPRL2 is a subunit of the GATOR1 complex and mutations in GATOR1 subunits, including NPRL2, are associated with epilepsy. To delineate the mechanisms underlying NPRL2-related epilepsies, we created a mouse (Mus musculus) model with neocortical loss of Nprl2. Mutant mice have increased mTORC1 signaling and exhibit spontaneous seizures. They also display abnormal synaptic function characterized by increased evoked and spontaneous EPSC and decreased evoked and spontaneous IPSC frequencies, respectively. Proteomic and metabolomics studies of Nprl2 mutants revealed alterations in known epilepsy-implicated proteins and metabolic pathways, including increases in the neurotransmitter, glycine. Furthermore, glycine actions on the NMDA receptor contribute to the electrophysiological and survival phenotypes of these mice. Taken together, in this neuronal Nprl2 model, we delineate underlying molecular, metabolic, and electrophysiological mechanisms contributing to mTORC1-related epilepsy, providing potential therapeutic targets for epilepsy.
Collapse
Affiliation(s)
- Brianne Dentel
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | | | - Chongyu Ren
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Vikram Jakkamsetti
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Andrew J. Holley
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Daniel Caballero
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Emily Oh
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Jay Gibson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Juan M. Pascual
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Kimberly M. Huber
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Benjamin P. Tu
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Peter T. Tsai
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX 75235, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75235, USA
- Departments of Pediatrics and Psychiatry, UT Southwestern Medical Center, Dallas, TX 75235, USA
| |
Collapse
|
20
|
Samanta D. DEPDC5-related epilepsy: A comprehensive review. Epilepsy Behav 2022; 130:108678. [PMID: 35429726 DOI: 10.1016/j.yebeh.2022.108678] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/07/2022] [Accepted: 03/21/2022] [Indexed: 11/03/2022]
Abstract
DEPDC5-related epilepsy, caused by pathogenic germline variants(with or without additional somatic variants in the brain) of DEPDC5 (Dishevelled, Egl-10 and Pleckstrin domain-containing protein 5) gene, is a newly discovered predominantly focal epilepsy linked to enhanced mTORC1 pathway. DEPDC5-related epilepsy includes several familial epilepsy syndromes, including familial focal epilepsy with variable foci (FFEVF) and rare sporadic nonlesional focal epilepsy. DEPDC5 has been identified as one of the more common epilepsy genes linked to infantile spasms and sudden unexpected death (SUDEP). Although intelligence usually is unaffected in DEPDC5-related epilepsy, some people have been diagnosed with intellectual disabilities, autism spectrum disorder, and other psychiatric problems. DEPDC5 variants have also been found in 20% of individuals with various brain abnormalities, challenging the traditional distinction between lesional and nonlesional epilepsies. The most exciting development of DEPDC5 variants is the possibility of precision therapeutics using mTOR inhibitors, as evidenced with phenotypic rescue in many animal models. However, more research is needed to better understand the functional impact of diverse (particularly missense or splice-region) variants, the specific involvement of DEPDC5 in epileptogenesis, and the creation and utilization of precision therapies in humans. Precision treatments for DEPDC5-related epilepsy will benefit not only a small number of people with the condition, but they will also pave the way for new therapeutic approaches in epilepsy (including acquired epilepsies in which mTORC1 activation occurs, for example, post-traumatic epilepsy) and other neurological disorders involving a dysfunctional mTOR pathway.
Collapse
Affiliation(s)
- Debopam Samanta
- Child Neurology Section, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
21
|
Yang T, Hu S, Chang WC, Kao HY, Wang Y. Perineuronal Nets Degradation and Parvalbumin Interneuron Loss in a Mouse Model of DEPDC5-Related Epilepsy. Dev Neurosci 2022; 44:671-677. [PMID: 35580549 PMCID: PMC9669280 DOI: 10.1159/000525039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022] Open
Abstract
DEPDC5, the key gene within the mechanistic target of rapamycin (mTOR) pathway, is one of the most common causative genes in patients with epilepsy and malformation of cortical development (MCD). Although somatic mutations in the dorsal cortical progenitors generate the malformed cortex, its pathogenesis of hyperexcitability is complex and remains unclear. We specifically deleted Depdc5 in the mouse forebrain dorsal progenitors to model DEPDC5-related epilepsy and investigated whether and how parvalbumin interneurons were non-cell autonomously affected in the malformed cortex. We showed that long before seizures, coincident with microglia inflammation, proteolytic enzymes degraded perineuronal nets (PNNs) in the malformed cortex, resulting in parvalbumin (PV+) interneuron loss and presynaptic inhibition impairment. Our studies, therefore, uncovered the hitherto unknown role of PNN in mTOR-related MCD, providing a new framework for mechanistic-based therapeutic development.
Collapse
Affiliation(s)
- Tao Yang
- Department of Neurology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Shuntong Hu
- Department of Neurology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Wei-Chih Chang
- Department of Neurology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Hsin-Yi Kao
- Department of Neurology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Yu Wang
- Department of Neurology, University of Michigan, Ann Arbor, MI, U.S.A
| |
Collapse
|
22
|
Bacq A, Roussel D, Bonduelle T, Zagaglia S, Maletic M, Ribierre T, Adle‐Biassette H, Marchal C, Jennesson M, An I, Picard F, Navarro V, Sisodiya SM, Baulac S. Cardiac Investigations in Sudden Unexpected Death in DEPDC5-Related Epilepsy. Ann Neurol 2022; 91:101-116. [PMID: 34693554 PMCID: PMC9299146 DOI: 10.1002/ana.26256] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Germline loss-of-function mutations in DEPDC5, and in its binding partners (NPRL2/3) of the mammalian target of rapamycin (mTOR) repressor GATOR1 complex, cause focal epilepsies and increase the risk of sudden unexpected death in epilepsy (SUDEP). Here, we asked whether DEPDC5 haploinsufficiency predisposes to primary cardiac defects that could contribute to SUDEP and therefore impact the clinical management of patients at high risk of SUDEP. METHODS Clinical cardiac investigations were performed in 16 patients with pathogenic variants in DEPDC5, NPRL2, or NPRL3. Two novel Depdc5 mouse strains, a human HA-tagged Depdc5 strain and a Depdc5 heterozygous knockout with a neuron-specific deletion of the second allele (Depdc5c/- ), were generated to investigate the role of Depdc5 in SUDEP and cardiac activity during seizures. RESULTS Holter, echocardiographic, and electrocardiographic (ECG) examinations provided no evidence for altered clinical cardiac function in the patient cohort, of whom 3 DEPDC5 patients succumbed to SUDEP and 6 had a family history of SUDEP. There was no cardiac injury at autopsy in a postmortem DEPDC5 SUDEP case. The HA-tagged Depdc5 mouse revealed expression of Depdc5 in the brain, heart, and lungs. Simultaneous electroencephalographic-ECG records on Depdc5c/- mice showed that spontaneous epileptic seizures resulting in a SUDEP-like event are not preceded by cardiac arrhythmia. INTERPRETATION Mouse and human data show neither structural nor functional cardiac damage that might underlie a primary contribution to SUDEP in the spectrum of DEPDC5-related epilepsies. ANN NEUROL 2022;91:101-116.
Collapse
Affiliation(s)
- Alexandre Bacq
- Sorbonne University, Paris Brain Institute (ICM), Inserm, CNRS, AP‐HP, Pitié‐Salpêtrière HospitalParisFrance
| | - Delphine Roussel
- Sorbonne University, Paris Brain Institute (ICM), Inserm, CNRS, AP‐HP, Pitié‐Salpêtrière HospitalParisFrance
| | - Thomas Bonduelle
- Sorbonne University, Paris Brain Institute (ICM), Inserm, CNRS, AP‐HP, Pitié‐Salpêtrière HospitalParisFrance
- Epilepsy and Neurology Department, Bordeaux University Hospital CenterBordeauxFrance
| | - Sara Zagaglia
- Department of Clinical and Experimental EpilepsyUniversity College London Queen Square Institute of NeurologyLondonUK
- Chalfont Centre for EpilepsyBucksUK
| | - Marina Maletic
- Sorbonne University, Paris Brain Institute (ICM), Inserm, CNRS, AP‐HP, Pitié‐Salpêtrière HospitalParisFrance
| | - Théo Ribierre
- Sorbonne University, Paris Brain Institute (ICM), Inserm, CNRS, AP‐HP, Pitié‐Salpêtrière HospitalParisFrance
| | - Homa Adle‐Biassette
- Pathological Anatomy Department, University of Paris, AP‐HP, Lariboisière Hospital, DMU, DREAM, UMR 1141, INSERMParisFrance
| | - Cécile Marchal
- Epilepsy and Neurology Department, Bordeaux University Hospital CenterBordeauxFrance
| | - Mélanie Jennesson
- Department of PediatricsAmerican Memorial Hospital, Reims University Hospital CenterReimsFrance
| | - Isabelle An
- Epileptology Unit and Reference Center of Rare Epilepsies, Pitié‐Salpêtrière Hospital, AP‐HPParisFrance
| | - Fabienne Picard
- EEG and Epilepsy Unit, Department of Clinical NeurosciencesUniversity Hospitals and Faculty of Medicine of GenevaGenevaSwitzerland
| | - Vincent Navarro
- Sorbonne University, Paris Brain Institute (ICM), Inserm, CNRS, AP‐HP, Pitié‐Salpêtrière HospitalParisFrance
- Epileptology Unit and Reference Center of Rare Epilepsies, Pitié‐Salpêtrière Hospital, AP‐HPParisFrance
| | - Sanjay M. Sisodiya
- Department of Clinical and Experimental EpilepsyUniversity College London Queen Square Institute of NeurologyLondonUK
- Chalfont Centre for EpilepsyBucksUK
| | - Stéphanie Baulac
- Sorbonne University, Paris Brain Institute (ICM), Inserm, CNRS, AP‐HP, Pitié‐Salpêtrière HospitalParisFrance
| |
Collapse
|
23
|
Ishida S, Zhao D, Sawada Y, Hiraoka Y, Mashimo T, Tanaka K. Dorsal telencephalon-specific Nprl2- and Nprl3-knockout mice: novel mouse models for GATORopathy. Hum Mol Genet 2021; 31:1519-1530. [PMID: 34965576 PMCID: PMC9071434 DOI: 10.1093/hmg/ddab337] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 09/28/2021] [Accepted: 11/12/2021] [Indexed: 11/30/2022] Open
Abstract
The most frequent genetic cause of focal epilepsies is variations in the GAP activity toward RAGs 1 complex genes DEP domain containing 5 (DEPDC5), nitrogen permease regulator 2-like protein (NPRL2) and nitrogen permease regulator 3-like protein (NPRL3). Because these variations are frequent and associated with a broad spectrum of focal epilepsies, a unique pathology categorized as GATORopathy can be conceptualized. Animal models recapitulating the clinical features of patients are essential to decipher GATORopathy. Although several genetically modified animal models recapitulate DEPDC5-related epilepsy, no models have been reported for NPRL2- or NPRL3-related epilepsies. Here, we conditionally deleted Nprl2 and Nprl3 from the dorsal telencephalon in mice [Emx1cre/+; Nprl2f/f (Nprl2-cKO) and Emx1cre/+; Nprl3f/f (Nprl3-cKO)] and compared their phenotypes with Nprl2+/−, Nprl3+/− and Emx1cre/+; Depdc5f/f (Depdc5-cKO) mice. Nprl2-cKO and Nprl3-cKO mice recapitulated the major abnormal features of patients—spontaneous seizures, and dysmorphic enlarged neuronal cells with increased mechanistic target of rapamycin complex 1 signaling—similar to Depdc5-cKO mice. Chronic postnatal rapamycin administration dramatically prolonged the survival period and inhibited seizure occurrence but not enlarged neuronal cells in Nprl2-cKO and Nprl3-cKO mice. However, the benefit of rapamycin after withdrawal was less durable in Nprl2- and Nprl3-cKO mice compared with Depdc5-cKO mice. Further studies using these conditional knockout mice will be useful for understanding GATORopathy and for the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Saeko Ishida
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510, Japan
- Division of Animal Genetics, Laboratory Animal Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Di Zhao
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510, Japan
- Division of Animal Genetics, Laboratory Animal Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Yuta Sawada
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510, Japan
| | - Yuichi Hiraoka
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510, Japan
- Laboratory of Genome Editing for Biomedical Research, MRI, TMDU, Tokyo, 101-0062, Japan
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510, Japan
- Center for Brain Integration Research (CBIR), TMDU, Tokyo, 113-8510, Japan
| |
Collapse
|
24
|
Marini C, Giardino M. Novel treatments in epilepsy guided by genetic diagnosis. Br J Clin Pharmacol 2021; 88:2539-2551. [PMID: 34778987 DOI: 10.1111/bcp.15139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 10/22/2021] [Accepted: 11/04/2021] [Indexed: 12/21/2022] Open
Abstract
In recent years, precision medicine has emerged as a new paradigm for improved and more individualized patient care. Its key objective is to provide the right treatment, to the right patient at the right time, by basing medical decisions on individual characteristics, including specific genetic biomarkers. In order to realize this objective researchers and physicians must first identify the underlying genetic cause; over the last 10 years, advances in genetics have made this possible for several monogenic epilepsies. Through next generation techniques, a precise genetic aetiology is attainable in 30-50% of genetic epilepsies beginning in the paediatric age. While committed in such search for novel genes carrying disease-causing variants, progress in the study of experimental models of epilepsy has also provided a better understanding of the mechanisms underlying the condition. Such advances are already being translated into improving care, management and treatment of some patients. Identification of a precise genetic aetiology can already direct physicians to prescribe treatments correcting specific metabolic defects, avoid antiseizure medicines that might aggravate functional consequences of the disease-causing variant or select the drugs that counteract the underlying, genetically determined, functional disturbance. Personalized, tailored treatments should not just focus on how to stop seizures but possibly prevent their onset and cure the disorder, often consisting of seizures and its comorbidities including cognitive, motor and behaviour deficiencies. This review discusses the therapeutic implications following a specific genetic diagnosis and the correlation between genetic findings, pathophysiological mechanisms and tailored seizure treatment, emphasizing the impact on current clinical practice.
Collapse
Affiliation(s)
- Carla Marini
- Child Neurology and Psychiatric Unit, Pediatric Hospital G. Salesi, United Hospitals of Ancona, Ancona, Italy
| | - Maria Giardino
- Child Neurology and Psychiatric Unit, Pediatric Hospital G. Salesi, United Hospitals of Ancona, Ancona, Italy
| |
Collapse
|
25
|
Patodia S, Somani A, Thom M. Review: Neuropathology findings in autonomic brain regions in SUDEP and future research directions. Auton Neurosci 2021; 235:102862. [PMID: 34411885 PMCID: PMC8455454 DOI: 10.1016/j.autneu.2021.102862] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/16/2021] [Accepted: 07/24/2021] [Indexed: 12/21/2022]
Abstract
Autonomic dysfunction is implicated from clinical, neuroimaging and experimental studies in sudden and unexpected death in epilepsy (SUDEP). Neuropathological analysis in SUDEP series enable exploration of acquired, seizure-related cellular adaptations in autonomic and brainstem autonomic centres of relevance to dysfunction in the peri-ictal period. Alterations in SUDEP compared to control groups have been identified in the ventrolateral medulla, amygdala, hippocampus and central autonomic regions. These involve neuropeptidergic, serotonergic and adenosine systems, as well as specific regional astroglial and microglial populations, as potential neuronal modulators, orchestrating autonomic dysfunction. Future research studies need to extend to clinically and genetically characterized epilepsies, to explore if common or distinct pathways of autonomic dysfunction mediate SUDEP. The ultimate objective of SUDEP research is the identification of disease biomarkers for at risk patients, to improve post-mortem recognition and disease categorisation, but ultimately, for exposing potential treatment targets of pharmacologically modifiable and reversible cellular alterations.
Collapse
Affiliation(s)
- Smriti Patodia
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Alyma Somani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Maria Thom
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK.
| |
Collapse
|
26
|
Guerrini R, Balestrini S, Wirrell EC, Walker MC. Monogenic Epilepsies: Disease Mechanisms, Clinical Phenotypes, and Targeted Therapies. Neurology 2021; 97:817-831. [PMID: 34493617 PMCID: PMC10336826 DOI: 10.1212/wnl.0000000000012744] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/18/2021] [Indexed: 02/05/2023] Open
Abstract
A monogenic etiology can be identified in up to 40% of people with severe epilepsy. To address earlier and more appropriate treatment strategies, clinicians are required to know the implications that specific genetic causes might have on pathophysiology, natural history, comorbidities, and treatment choices. In this narrative review, we summarize concepts on the genetic epilepsies based on the underlying pathophysiologic mechanisms and present the current knowledge on treatment options based on evidence provided by controlled trials or studies with lower classification of evidence. Overall, evidence robust enough to guide antiseizure medication (ASM) choices in genetic epilepsies remains limited to the more frequent conditions for which controlled trials and observational studies have been possible. Most monogenic disorders are very rare and ASM choices for them are still based on inferences drawn from observational studies and early, often anecdotal, experiences with precision therapies. Precision medicine remains applicable to only a narrow number of patients with monogenic epilepsies and may target only part of the actual functional defects. Phenotypic heterogeneity is remarkable, and some genetic mutations activate epileptogenesis through their developmental effects, which may not be reversed postnatally. Other genes seem to have pure functional consequences on excitability, acting through either loss- or gain-of-function effects, and these may have opposite treatment implications. In addition, the functional consequences of missense mutations may be difficult to predict, making precision treatment approaches considerably more complex than estimated by deterministic interpretations. Knowledge of genetic etiologies can influence the approach to surgical treatment of focal epilepsies. Identification of germline mutations in specific genes contraindicates surgery while mutations in other genes do not. Identification, quantification, and functional characterization of specific somatic mutations before surgery using CSF liquid biopsy or after surgery in brain specimens will likely be integrated in planning surgical strategies and reintervention after a first unsuccessful surgery as initial evidence suggests that mutational load may correlate with the epileptogenic zone. Promising future directions include gene manipulation by DNA or mRNA targeting; although most are still far from clinical use, some are in early phase clinical development.
Collapse
Affiliation(s)
- Renzo Guerrini
- From the Neuroscience Department (R.G., S.B.), Meyer Children's Hospital-University of Florence, Italy; Department of Clinical and Experimental Epilepsy (S.B., M.C.W.), UCL Queen Square Institute of Neurology, London; Chalfont Centre for Epilepsy (S.B.), Buckinghamshire, UK; and Divisions of Child and Adolescent Neurology and Epilepsy (E.C.W.), Department of Neurology, Mayo Clinic, Rochester, MN.
| | - Simona Balestrini
- From the Neuroscience Department (R.G., S.B.), Meyer Children's Hospital-University of Florence, Italy; Department of Clinical and Experimental Epilepsy (S.B., M.C.W.), UCL Queen Square Institute of Neurology, London; Chalfont Centre for Epilepsy (S.B.), Buckinghamshire, UK; and Divisions of Child and Adolescent Neurology and Epilepsy (E.C.W.), Department of Neurology, Mayo Clinic, Rochester, MN
| | - Elaine C Wirrell
- From the Neuroscience Department (R.G., S.B.), Meyer Children's Hospital-University of Florence, Italy; Department of Clinical and Experimental Epilepsy (S.B., M.C.W.), UCL Queen Square Institute of Neurology, London; Chalfont Centre for Epilepsy (S.B.), Buckinghamshire, UK; and Divisions of Child and Adolescent Neurology and Epilepsy (E.C.W.), Department of Neurology, Mayo Clinic, Rochester, MN
| | - Matthew C Walker
- From the Neuroscience Department (R.G., S.B.), Meyer Children's Hospital-University of Florence, Italy; Department of Clinical and Experimental Epilepsy (S.B., M.C.W.), UCL Queen Square Institute of Neurology, London; Chalfont Centre for Epilepsy (S.B.), Buckinghamshire, UK; and Divisions of Child and Adolescent Neurology and Epilepsy (E.C.W.), Department of Neurology, Mayo Clinic, Rochester, MN
| |
Collapse
|
27
|
Moloney PB, Cavalleri GL, Delanty N. Epilepsy in the mTORopathies: opportunities for precision medicine. Brain Commun 2021; 3:fcab222. [PMID: 34632383 PMCID: PMC8495134 DOI: 10.1093/braincomms/fcab222] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 01/16/2023] Open
Abstract
The mechanistic target of rapamycin signalling pathway serves as a ubiquitous regulator of cell metabolism, growth, proliferation and survival. The main cellular activity of the mechanistic target of rapamycin cascade funnels through mechanistic target of rapamycin complex 1, which is inhibited by rapamycin, a macrolide compound produced by the bacterium Streptomyces hygroscopicus. Pathogenic variants in genes encoding upstream regulators of mechanistic target of rapamycin complex 1 cause epilepsies and neurodevelopmental disorders. Tuberous sclerosis complex is a multisystem disorder caused by mutations in mechanistic target of rapamycin regulators TSC1 or TSC2, with prominent neurological manifestations including epilepsy, focal cortical dysplasia and neuropsychiatric disorders. Focal cortical dysplasia type II results from somatic brain mutations in mechanistic target of rapamycin pathway activators MTOR, AKT3, PIK3CA and RHEB and is a major cause of drug-resistant epilepsy. DEPDC5, NPRL2 and NPRL3 code for subunits of the GTPase-activating protein (GAP) activity towards Rags 1 complex (GATOR1), the principal amino acid-sensing regulator of mechanistic target of rapamycin complex 1. Germline pathogenic variants in GATOR1 genes cause non-lesional focal epilepsies and epilepsies associated with malformations of cortical development. Collectively, the mTORopathies are characterized by excessive mechanistic target of rapamycin pathway activation and drug-resistant epilepsy. In the first large-scale precision medicine trial in a genetically mediated epilepsy, everolimus (a synthetic analogue of rapamycin) was effective at reducing seizure frequency in people with tuberous sclerosis complex. Rapamycin reduced seizures in rodent models of DEPDC5-related epilepsy and focal cortical dysplasia type II. This review outlines a personalized medicine approach to the management of epilepsies in the mTORopathies. We advocate for early diagnostic sequencing of mechanistic target of rapamycin pathway genes in drug-resistant epilepsy, as identification of a pathogenic variant may point to an occult dysplasia in apparently non-lesional epilepsy or may uncover important prognostic information including, an increased risk of sudden unexpected death in epilepsy in the GATORopathies or favourable epilepsy surgery outcomes in focal cortical dysplasia type II due to somatic brain mutations. Lastly, we discuss the potential therapeutic application of mechanistic target of rapamycin inhibitors for drug-resistant seizures in GATOR1-related epilepsies and focal cortical dysplasia type II.
Collapse
Affiliation(s)
- Patrick B Moloney
- FutureNeuro, the Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin, D02 VN51, Ireland
| | - Gianpiero L Cavalleri
- FutureNeuro, the Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin, D02 VN51, Ireland
| | - Norman Delanty
- FutureNeuro, the Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin, D02 VN51, Ireland
| |
Collapse
|
28
|
Structure-based modification of pyrazolone derivatives to inhibit mTORC1 by targeting the leucyl-tRNA synthetase-RagD interaction. Bioorg Chem 2021; 112:104907. [PMID: 33979735 DOI: 10.1016/j.bioorg.2021.104907] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/21/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022]
Abstract
The enzyme leucyl-tRNA synthetase (LRS) and the amino acid leucine regulate the mechanistic target of rapamycin (mTOR) signaling pathway. Leucine-dependent mTORC1 activation depends on GTPase activating protein events mediated by LRS. In a prior study, compound BC-LI-0186 was discovered and shown to interfere with the mTORC1 signaling pathway by inhibiting the LRS-RagD interaction. However, BC-LI-0186 exhibited poor solubility and was metabolized by human liver microsomes. In this study, in silico physicochemical properties and metabolite analysis of BC-LI-0186 are used to investigate the addition of functional groups to improve solubility and microsomal stability. In vitro experiments demonstrated that 7b and 8a had improved chemical properties while still maintaining inhibitory activity against mTORC1. The results suggest a new strategy for the discovery of novel drug candidates and the treatment of diverse mTORC1-related diseases.
Collapse
|
29
|
Nicotinic Receptors in Sleep-Related Hypermotor Epilepsy: Pathophysiology and Pharmacology. Brain Sci 2020; 10:brainsci10120907. [PMID: 33255633 PMCID: PMC7761363 DOI: 10.3390/brainsci10120907] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/19/2020] [Accepted: 11/21/2020] [Indexed: 12/12/2022] Open
Abstract
Sleep-related hypermotor epilepsy (SHE) is characterized by hyperkinetic focal seizures, mainly arising in the neocortex during non-rapid eye movements (NREM) sleep. The familial form is autosomal dominant SHE (ADSHE), which can be caused by mutations in genes encoding subunits of the neuronal nicotinic acetylcholine receptor (nAChR), Na+-gated K+ channels, as well as non-channel signaling proteins, such as components of the gap activity toward rags 1 (GATOR1) macromolecular complex. The causative genes may have different roles in developing and mature brains. Under this respect, nicotinic receptors are paradigmatic, as different pathophysiological roles are exerted by distinct nAChR subunits in adult and developing brains. The widest evidence concerns α4 and β2 subunits. These participate in heteromeric nAChRs that are major modulators of excitability in mature neocortical circuits as well as regulate postnatal synaptogenesis. However, growing evidence implicates mutant α2 subunits in ADSHE, which poses interpretive difficulties as very little is known about the function of α2-containing (α2*) nAChRs in the human brain. Planning rational therapy must consider that pharmacological treatment could have different effects on synaptic maturation and adult excitability. We discuss recent attempts towards precision medicine in the mature brain and possible approaches to target developmental stages. These issues have general relevance in epilepsy treatment, as the pathogenesis of genetic epilepsies is increasingly recognized to involve developmental alterations.
Collapse
|
30
|
Tafur L, Kefauver J, Loewith R. Structural Insights into TOR Signaling. Genes (Basel) 2020; 11:E885. [PMID: 32759652 PMCID: PMC7464330 DOI: 10.3390/genes11080885] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 12/31/2022] Open
Abstract
The Target of Rapamycin (TOR) is a highly conserved serine/threonine protein kinase that performs essential roles in the control of cellular growth and metabolism. TOR acts in two distinct multiprotein complexes, TORC1 and TORC2 (mTORC1 and mTORC2 in humans), which maintain different aspects of cellular homeostasis and orchestrate the cellular responses to diverse environmental challenges. Interest in understanding TOR signaling is further motivated by observations that link aberrant TOR signaling to a variety of diseases, ranging from epilepsy to cancer. In the last few years, driven in large part by recent advances in cryo-electron microscopy, there has been an explosion of available structures of (m)TORC1 and its regulators, as well as several (m)TORC2 structures, derived from both yeast and mammals. In this review, we highlight and summarize the main findings from these reports and discuss both the fascinating and unexpected molecular biology revealed and how this knowledge will potentially contribute to new therapeutic strategies to manipulate signaling through these clinically relevant pathways.
Collapse
Affiliation(s)
- Lucas Tafur
- Department of Molecular Biology, University of Geneva, 30 quai Ernest-Ansermet, CH1211 Geneva, Switzerland; (L.T.); (J.K.)
| | - Jennifer Kefauver
- Department of Molecular Biology, University of Geneva, 30 quai Ernest-Ansermet, CH1211 Geneva, Switzerland; (L.T.); (J.K.)
| | - Robbie Loewith
- Department of Molecular Biology, University of Geneva, 30 quai Ernest-Ansermet, CH1211 Geneva, Switzerland; (L.T.); (J.K.)
- Swiss National Centre for Competence in Research (NCCR) in Chemical Biology, University of Geneva, Sciences II, Room 3-308, 30 Quai Ernest-Ansermet, CH1211 Geneva, Switzerland
| |
Collapse
|