1
|
Romanovsky E, Choudhary A, Peles D, Abu-Akel A, Stern S. Uncovering convergence and divergence between autism and schizophrenia using genomic tools and patients' neurons. Mol Psychiatry 2025; 30:1019-1028. [PMID: 39237719 DOI: 10.1038/s41380-024-02740-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Autism spectrum disorders (ASDs) are highly heritable and result in abnormal repetitive behaviors and impairment in communication and cognitive skills. Previous studies have focused on the genetic correlation between ASDs and other neuropsychiatric disorders, but an in-depth understanding of the correlation to other disorders is required. We conducted an extensive meta-analysis of common variants identified in ASDs by genome-wide association studies (GWAS) and compared it to the consensus genes and single nucleotide polymorphisms (SNPs) of Schizophrenia (SCZ). We found approximately 75% of the GWAS genes that are associated with ASD are also associated with SCZ. We further investigated the cellular phenotypes of neurons derived from induced pluripotent stem cell (iPSC) models in ASD and SCZ. Our findings revealed that ASD and SCZ neurons initially follow divergent developmental trajectories compared to control neurons. However, despite these early diametrical differences, both ASD and SCZ neurons ultimately display similar deficits in synaptic activity as they mature. This significant genetic overlap between ASD and SCZ, coupled with the convergence towards similar synaptic deficits, highlights the intricate interplay of genetic and developmental factors in shaping the shared underlying mechanisms of these complex neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Eva Romanovsky
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ahmad Abu-Akel
- School of Psychological Sciences, University of Haifa, Haifa, Israel
- The Haifa Brain and Behavior Hub, University of Haifa, Haifa, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| |
Collapse
|
2
|
Montanucci L, Brünger T, Bhattarai N, Boßelmann CM, Kim S, Allen JP, Zhang J, Klöckner C, Krey I, Fariselli P, May P, Lemke JR, Myers SJ, Yuan H, Traynelis SF, Lal D. Ligand distances as key predictors of pathogenicity and function in NMDA receptors. Hum Mol Genet 2025; 34:128-139. [PMID: 39535073 PMCID: PMC11780861 DOI: 10.1093/hmg/ddae156] [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: 05/25/2024] [Revised: 10/10/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Genetic variants in the genes GRIN1, GRIN2A, GRIN2B, and GRIN2D, which encode subunits of the N-methyl-D-aspartate receptor (NMDAR), have been associated with severe and heterogeneous neurologic and neurodevelopmental disorders, including early onset epilepsy, developmental and epileptic encephalopathy, intellectual disability, and autism spectrum disorders. Missense variants in these genes can result in gain or loss of the NMDAR function, requiring opposite therapeutic treatments. Computational methods that predict pathogenicity and molecular functional effects of missense variants are therefore crucial for therapeutic applications. We assembled 223 missense variants from patients, 631 control variants from the general population, and 160 missense variants characterized by electrophysiological readouts that show whether they can enhance or reduce the function of the receptor. This includes new functional data from 33 variants reported here, for the first time. By mapping these variants onto the NMDAR protein structures, we found that pathogenic/benign variants and variants that increase/decrease the channel function were distributed unevenly on the protein structure, with spatial proximity to ligands bound to the agonist and antagonist binding sites being a key predictive feature for both variant pathogenicity and molecular functional consequences. Leveraging distances from ligands, we developed two machine-learning based predictors for NMDA variants: a pathogenicity predictor which outperforms currently available predictors and the first molecular function (increase/decrease) predictor. Our findings can have direct application to patient care by improving diagnostic yield for genetic neurodevelopmental disorders and by guiding personalized treatment informed by the knowledge of the molecular disease mechanism.
Collapse
Affiliation(s)
- Ludovica Montanucci
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 1133 John Freeman Blvd, Houston, TX 77030, United States
| | - Tobias Brünger
- Cologne Center for Genomics, University of Cologne, University Hospital Cologne, Weyertal 115b, Cologne 50937, Germany
| | - Nisha Bhattarai
- Epilepsy Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44106, United States
| | - Christian M Boßelmann
- Epilepsy Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44106, United States
| | - Sukhan Kim
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - James P Allen
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Jing Zhang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Chiara Klöckner
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Philipp-Rosenthal-street 55, Leipzig 04103, Germany
| | - Ilona Krey
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Philipp-Rosenthal-street 55, Leipzig 04103, Germany
| | - Piero Fariselli
- Department of Medical Sciences, University of Torino, Via Santena 19,Torino, 10123, Italy
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7 Av. des Hauts-Fourneaux, Esch-sur-Alzette, 4362, Luxembourg
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Philipp-Rosenthal-street 55, Leipzig 04103, Germany
| | - Scott J Myers
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Dennis Lal
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 1133 John Freeman Blvd, Houston, TX 77030, United States
- Epilepsy Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44106, United States
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (M.I.T.) and Harvard, 415 Main St, Cambridge, MA 02142, United States
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and M.I.T, 415 Main St., Cambridge, MA 02142, United States
| |
Collapse
|
3
|
Cui C, Li H, Bao Y, Han Y, Yu H, Song H, Zhang B. Association between GRIN2B polymorphism and Parkinson's disease risk, age at onset, and progression in Southern China. Front Neurol 2024; 15:1459576. [PMID: 39758784 PMCID: PMC11697588 DOI: 10.3389/fneur.2024.1459576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 11/19/2024] [Indexed: 01/07/2025] Open
Abstract
Background and objectives The role of N-methyl-D-aspartate receptor 2B (GRIN2B) single nucleotide polymorphisms (SNPs) in influencing the risk and progression of Parkinson's disease (PD) is still unclear. This study aimed to assess the impact of GRIN2B genotype status on PD susceptibility and symptom progression. Methods We enrolled 165 individuals with sporadic PD and 154 healthy controls, all of whom had comprehensive clinical data available at the start and during follow-up. We used chi-squared (χ2) analysis to compare the allele and genotype frequency distributions between the patient and control groups. Linear mixed-effect models were employed to investigate the link between the GRIN2B genotype and the progression of motor and cognitive symptoms. Results The prevalence of the GG + GT genotype and G allele was higher in patients compared to controls (p = 0.032 and p = 0.001, respectively). Subgroup analysis revealed that the GG + GT genotype and G allele were significantly more frequently observed in late-onset PD (LOPD) patients compared to early-onset PD (EOPD) patients (p = 0.014 and p = 0.035, respectively). Notably, individuals with the GG + GT genotype exhibited an estimated annual progression rate of 6.10 points on the Unified Parkinson's Disease Rating Scale (UPDRS), which is significantly higher than that of the TT genotype carriers. Furthermore, the GG + GT carriers showed a markedly rapid progression in rigidity. In addition, the GG + GT carriers demonstrated significantly faster progression rates in rigidity (1.83 points/year) and axial impairment (1.2 points/year) compared to the TT carriers. Notably, the GG genotype carriers exhibited a more rapid decline in recall function. Conclusion The GRIN2B rs219882 G allele is associated with increased PD susceptibility, particularly in LOPD. The carriers of the GG + GT genotype exhibited more rapid motor symptom progression, with a pronounced impact on rigidity and axial impairment.
Collapse
Affiliation(s)
- Can Cui
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hongxia Li
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yiwen Bao
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yingying Han
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hongxiang Yu
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huan Song
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Bei Zhang
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
4
|
Zhang L, Duan Y, Ma R, Han J, Pan N, Gao L, Wang Y. Clinical phenotype and functional influence of GRIN2A variants in epilepsy-aphasia syndrome. Epilepsia Open 2024; 9:2306-2318. [PMID: 39474911 PMCID: PMC11633710 DOI: 10.1002/epi4.13057] [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: 11/02/2023] [Revised: 08/28/2024] [Accepted: 09/08/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVE N-methyl-D-aspartate receptors are glutamate-gated ion channels that play a crucial role in brain function. Numerous inherited or de novo variants in the GRIN2A gene, encoding the GluN2A subunit of the receptor, have been identified in patients with epilepsy. In addition, it is worth noting that GRIN2A variants exhibit a strong correlation with epilepsy-aphasia syndromes, a group of age-dependent epileptic, cognitive, and language disorders with a characteristic electroencephalographic pattern. METHODS Whole exome sequencing was conducted in enrolled patients with epilepsy-aphasia syndromes, and GRIN2A variants were screened. The conservation of substituted residues, conformational changes of mutant subunits, and in silico predictions of pathogenicity were thoroughly assessed in our study. Functional alterations of the variants were examined using whole-cell voltage-clamp current recordings while the relative surface expression levels of subunit proteins were assessed via immunofluorescence assays. A summary of previously published GRIN2A missense variants was conducted to investigate the genotypic-phenotypic-functional correlations. RESULTS Two missense GRIN2A variants (c. 2482A >G/p. M828V, c. 2627 T >C/p. I876T) were identified, which are located in the transmembrane helix M4 and C-terminus domain of the GluN2A subunit, respectively. Both variants exhibited reduced current density of NMDARs and surface/total expression levels of GluN2A subunits, while M828V showed a decreased extent of desensitization as well. A further summary of the previously reported GRIN2A variants demonstrated that more variable phenotypes were observed for variants situated in the C-terminus domain or those with loss-of-function effects. SIGNIFICANCE Our study expands the phenotypic and functional range of GRIN2A-related disorders. In order to optimally establish the domain-function-phenotype correlations in GRIN2A variants, it is imperative to gather a more extensive set of clinical and functional data. PLAIN LANGUAGE SUMMARY This study has identified two genetic variants of the GRIN2A gene in patients with epilepsy-aphasia syndrome. We assess the variants' harmfulness through a variety of functional experiments, including evaluating the expression level of the mutated protein and the resulting changes in electrophysiological activities. Also, we reviewed previously published papers about GRIN2A variants in epilepsy to learn more about the correlations between their locations, functional changes, and clinical manifestations.
Collapse
Affiliation(s)
- Lu Zhang
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Yiran Duan
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
- Present address:
Department of NeurologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Rui Ma
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
- Present address:
Department of Endocrinology, Genetics and MetabolismNational Center for Children's Health, Beijing Children's Hospital, Capital Medical UniversityBeijingChina
| | - Jiaqi Han
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Na Pan
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Lehong Gao
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Yuping Wang
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
- Center of Epilepsy, Beijing Institute for Brain DisordersCapital Medical University, Ministry of Science and TechnologyBeijingChina
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| |
Collapse
|
5
|
Viswanathan S, Oliver KL, Regan BM, Schneider AL, Myers CT, Mehaffey MG, LaCroix AJ, Antony J, Webster R, Cardamone M, Subramanian GM, Chiu ATG, Roza E, Teleanu RI, Malone S, Leventer RJ, Gill D, Berkovic SF, Hildebrand MS, Goad BS, Howell KB, Symonds JD, Brunklaus A, Sadleir LG, Zuberi SM, Mefford HC, Scheffer IE. Solving the Etiology of Developmental and Epileptic Encephalopathy with Spike-Wave Activation in Sleep (D/EE-SWAS). Ann Neurol 2024; 96:932-943. [PMID: 39096015 PMCID: PMC11496008 DOI: 10.1002/ana.27041] [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: 02/23/2024] [Revised: 05/31/2024] [Accepted: 07/11/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE To understand the etiological landscape and phenotypic differences between 2 developmental and epileptic encephalopathy (DEE) syndromes: DEE with spike-wave activation in sleep (DEE-SWAS) and epileptic encephalopathy with spike-wave activation in sleep (EE-SWAS). METHODS All patients fulfilled International League Against Epilepsy (ILAE) DEE-SWAS or EE-SWAS criteria with a Core cohort (n = 91) drawn from our Epilepsy Genetics research program, together with 10 etiologically solved patients referred by collaborators in the Expanded cohort (n = 101). Detailed phenotyping and analysis of molecular genetic results were performed. We compared the phenotypic features of individuals with DEE-SWAS and EE-SWAS. Brain-specific gene co-expression analysis was performed for D/EE-SWAS genes. RESULTS We identified the etiology in 42/91 (46%) patients in our Core cohort, including 29/44 (66%) with DEE-SWAS and 13/47 (28%) with EE-SWAS. A genetic etiology was identified in 31/91 (34%). D/EE-SWAS genes were highly co-expressed in brain, highlighting the importance of channelopathies and transcriptional regulators. Structural etiologies were found in 12/91 (13%) individuals. We identified 10 novel D/EE-SWAS genes with a range of functions: ATP1A2, CACNA1A, FOXP1, GRIN1, KCNMA1, KCNQ3, PPFIA3, PUF60, SETD1B, and ZBTB18, and 2 novel copy number variants, 17p11.2 duplication and 5q22 deletion. Although developmental regression patterns were similar in both syndromes, DEE-SWAS was associated with a longer duration of epilepsy and poorer intellectual outcome than EE-SWAS. INTERPRETATION DEE-SWAS and EE-SWAS have highly heterogeneous genetic and structural etiologies. Phenotypic analysis highlights valuable clinical differences between DEE-SWAS and EE-SWAS which inform clinical care and prognostic counseling. Our etiological findings pave the way for the development of precision therapies. ANN NEUROL 2024;96:932-943.
Collapse
Affiliation(s)
- Sindhu Viswanathan
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
- Department of Paediatrics, Hospital Pulau Pinang, Pulau Pinang, Malaysia
| | - Karen L. Oliver
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
- Population Health and Immunity Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, the University of Melbourne, Melbourne, VIC 3010, Australia
| | - Brigid M. Regan
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
| | - Amy L. Schneider
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
| | - Candace T. Myers
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Michele G. Mehaffey
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, USA
| | - Amy J. LaCroix
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, USA
| | - Jayne Antony
- T.Y. Nelson Department of Neurology and Neurosurgery, The Children’s Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Richard Webster
- T.Y. Nelson Department of Neurology and Neurosurgery, The Children’s Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Michael Cardamone
- Sydney Children’s Hospital, Randwick; School of Clinical Medicine, UNSW Sydney, New South Wales, Australia
| | | | - Annie TG Chiu
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
| | - Eugenia Roza
- Faculty of Medicine, Clinical Neurosciences Department, Paediatric Neurology, Carol Davila University of Medicine and Pharmacy, Romania
- Pediatric Neurology Department, Dr. Victor Gomoiu Children’s Hospital, Romania
| | - Raluca I. Teleanu
- Faculty of Medicine, Clinical Neurosciences Department, Paediatric Neurology, Carol Davila University of Medicine and Pharmacy, Romania
- Pediatric Neurology Department, Dr. Victor Gomoiu Children’s Hospital, Romania
| | - Stephen Malone
- Centre for Advanced Imaging, University of Queensland, St Lucia, Australia
- Neurosciences Department, Queensland Children’s Hospital, South Brisbane Queensland, Australia
| | - Richard J. Leventer
- Department of Neurology, Royal Children’s Hospital, University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| | - Deepak Gill
- T.Y. Nelson Department of Neurology and Neurosurgery, The Children’s Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- Kids Neuroscience Centre, Kids Research Institute, Sydney, Australia
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
| | - Michael S. Hildebrand
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
- Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| | - Beatrice S. Goad
- Department of Neurology, Royal Children’s Hospital, University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| | - Katherine B. Howell
- Department of Neurology, Royal Children’s Hospital, University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| | - Joseph D. Symonds
- School of Health and Wellbeing, University of Glasgow, UK
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Glasgow, UK
| | - Andreas Brunklaus
- School of Health and Wellbeing, University of Glasgow, UK
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Glasgow, UK
| | - Lynette G. Sadleir
- Department of Paediatrics and Child Health, University of Otago Wellington, Wellington, New Zealand
| | - Sameer M. Zuberi
- School of Health and Wellbeing, University of Glasgow, UK
- The Paediatric Neurosciences Research Group, Royal Hospital for Children, Glasgow, UK
| | - Heather C. Mefford
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA, USA
- Centre for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN,USA
| | - Ingrid E. Scheffer
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Melbourne, Australia
- Department of Neurology, Royal Children’s Hospital, University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
- The Florey Institute of Neurosciences and Mental Health, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Rubio C, Romo-Parra H, López-Landa A, Rubio-Osornio M. Classification of Current Experimental Models of Epilepsy. Brain Sci 2024; 14:1024. [PMID: 39452036 PMCID: PMC11506208 DOI: 10.3390/brainsci14101024] [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: 09/13/2024] [Revised: 10/03/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
INTRODUCTION This article provides an overview of several experimental models, including in vivo, genetics, chemical, knock-in, knock-out, electrical, in vitro, and optogenetics models, that have been employed to investigate epileptogenesis. The present review introduces a novel categorization of these models, taking into account the fact that the most recent classification that gained widespread acceptance was established by Fisher in 1989. A significant number of such models have become virtually outdated. OBJECTIVE This paper specifically examines the models that have contributed to the investigation of partial seizures, generalized seizures, and status epilepticus. DISCUSSION A description is provided of the primary features associated with the processes that produce and regulate the symptoms of various epileptogenesis models. Numerous experimental epilepsy models in animals have made substantial contributions to the investigation of particular brain regions that are capable of inducing seizures. Experimental models of epilepsy have also enabled the investigation of the therapeutic mechanisms of anti-epileptic medications. Typically, animals are selected for the development and study of experimental animal models of epilepsy based on the specific form of epilepsy being investigated. CONCLUSIONS Currently, it is established that specific animal species can undergo epileptic seizures that resemble those described in humans. Nevertheless, it is crucial to acknowledge that a comprehensive assessment of all forms of human epilepsy has not been feasible. However, these experimental models, both those derived from channelopathies and others, have provided a limited comprehension of the fundamental mechanisms of this disease.
Collapse
Affiliation(s)
- Carmen Rubio
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico; (C.R.); (H.R.-P.); (A.L.-L.)
| | - Héctor Romo-Parra
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico; (C.R.); (H.R.-P.); (A.L.-L.)
- Psychology Department, Universidad Iberoamericana, Mexico City 01219, Mexico
| | - Alejandro López-Landa
- Department of Neurophysiology, Instituto Nacional de Neurología y Neurocirugía, Mexico City 14269, Mexico; (C.R.); (H.R.-P.); (A.L.-L.)
| | - Moisés Rubio-Osornio
- Department of Neurochemistry, Instituto Nacional de Neurología y Neurocirugía, Av. Insurgentes Sur 3877, Mexico City 14269, Mexico
| |
Collapse
|
7
|
Cetica V, Cavallin M, Ricci ML, Mandorlini C, Bartolini E, Parrini E, Guerrini R. Double gonosomal mosaicism as an unusual hereditary mechanism in familial GRIN2A-related disorder. J Med Genet 2024; 61:999-1002. [PMID: 39084904 DOI: 10.1136/jmg-2024-110101] [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: 05/03/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024]
Abstract
We aim to describe double gonosomal mosaicism in the GRIN2A gene in a mother who passed on two different pathogenic variants at the same nucleotide to her two affected children. We studied a boy with epilepsy and intellectual disability, along with his sister and mother who exhibited language impairment and learning difficulties without epilepsy. We identified in the proband a splice-site variant in GRIN2A (c.1008-1G>A) inherited from his mother. Subsequent testing of his sister revealed a different change at the same nucleotide c.1008-1G>T, which was also present in the mother's DNA at 3.9% allele frequency. The co-occurrence of two mutational events at the same nucleotide is extremely rare. Since a chance occurrence is unlikely, we hypothesise that a base mismatch may introduce instability triggering a second event. In this family, the mother carries three alleles, of which one is at very low frequency. This complex genetic landscape poses diagnostic challenges since low-level mosaicism may escape detection via conventional methods. Applying specific technology becomes crucial, as double mosaicism might prove to be more prevalent than anticipated severely impacting diagnostic accuracy and genetic counselling.
Collapse
Affiliation(s)
- Valentina Cetica
- Paediatric Neurology Unit and Laboratories, Neuroscience Department, Member of ERN Epicare and ITHACA, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Mara Cavallin
- Paediatric Neurology Unit and Laboratories, Neuroscience Department, Member of ERN Epicare and ITHACA, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Maria Luisa Ricci
- Paediatric Neurology Unit and Laboratories, Neuroscience Department, Member of ERN Epicare and ITHACA, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Claudia Mandorlini
- Paediatric Neurology Unit and Laboratories, Neuroscience Department, Member of ERN Epicare and ITHACA, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Emanuele Bartolini
- Department of Developmental Neuroscience, IRCCS Foundation Stella Maris, Pisa, Italy
| | - Elena Parrini
- Paediatric Neurology Unit and Laboratories, Neuroscience Department, Member of ERN Epicare and ITHACA, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Renzo Guerrini
- Paediatric Neurology Unit and Laboratories, Neuroscience Department, Member of ERN Epicare and ITHACA, Meyer Children's Hospital IRCCS, Florence, Italy
- University of Florence, Florence, Italy
| |
Collapse
|
8
|
Wang Y, Yang H, Li N, Wang L, Guo C, Ma W, Liu S, Peng C, Chen J, Song H, Chen H, Ma X, Yi J, Lian J, Kong W, Dong J, Tu X, Shah M, Tian X, Huang Z. A Novel Ubiquitin Ligase Adaptor PTPRN Suppresses Seizure Susceptibility through Endocytosis of Na V1.2 Sodium Channels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400560. [PMID: 38874331 PMCID: PMC11304301 DOI: 10.1002/advs.202400560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/06/2024] [Indexed: 06/15/2024]
Abstract
Intrinsic plasticity, a fundamental process enabling neurons to modify their intrinsic properties, plays a crucial role in shaping neuronal input-output function and is implicated in various neurological and psychiatric disorders. Despite its importance, the underlying molecular mechanisms of intrinsic plasticity remain poorly understood. In this study, a new ubiquitin ligase adaptor, protein tyrosine phosphatase receptor type N (PTPRN), is identified as a regulator of intrinsic neuronal excitability in the context of temporal lobe epilepsy. PTPRN recruits the NEDD4 Like E3 Ubiquitin Protein Ligase (NEDD4L) to NaV1.2 sodium channels, facilitating NEDD4L-mediated ubiquitination, and endocytosis of NaV1.2. Knockout of PTPRN in hippocampal granule cells leads to augmented NaV1.2-mediated sodium currents and higher intrinsic excitability, resulting in increased seizure susceptibility in transgenic mice. Conversely, adeno-associated virus-mediated delivery of PTPRN in the dentate gyrus region decreases intrinsic excitability and reduces seizure susceptibility. Moreover, the present findings indicate that PTPRN exerts a selective modulation effect on voltage-gated sodium channels. Collectively, PTPRN plays a significant role in regulating intrinsic excitability and seizure susceptibility, suggesting a potential strategy for precise modulation of NaV1.2 channels' function.
Collapse
Affiliation(s)
- Yifan Wang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Hui Yang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Na Li
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Lili Wang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Chang Guo
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Weining Ma
- Department of NeurologyShengjing Hospital Affiliated to China Medical UniversityShenyang110022China
| | - Shiqi Liu
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Chao Peng
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jiexin Chen
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Huifang Song
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Hedan Chen
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Xinyue Ma
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jingyun Yi
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jingjing Lian
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Weikaixin Kong
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Jie Dong
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Xinyu Tu
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| | - Mala Shah
- UCL School of PharmacyUniversity College LondonLondonWC1N 1AXUK
| | - Xin Tian
- Department of NeurologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of NeurologyChongqing400016China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- IDG/McGovern Institute for Brain ResearchPeking UniversityBeijing100871China
| |
Collapse
|
9
|
Heydarlou D, Asghari A, Ezzati S, Khalil M, Karim S, Lui F. An Unusual Case of GRIN2A Mutation Presenting as Progressive Limbic Encephalopathy in an Adult. Cureus 2024; 16:e63046. [PMID: 39050322 PMCID: PMC11268452 DOI: 10.7759/cureus.63046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
The glutamate ionotropic receptor NMDA (N-methyl-D-aspartate) type subunit 2A gene (GRIN2A) encodes the GluN2A subunit of NMDA receptors, which are essential for synaptic plasticity and memory consolidation. Mutations in GRIN2A can disrupt these processes, often affecting the pediatric population and causing various neurological disorders characterized by epilepsy, intellectual disability, and aphasia, among other neuropsychiatric findings. We report an unusual presentation of adult-onset GRIN2A mutation-associated progressive limbic encephalopathy (LE), characterized by rapidly progressive cortical atrophy, seizures, aphasia, and neuropsychiatric abnormalities, which ultimately led to the patient's sudden demise. Further research into GRIN2A mutations will improve our understanding of such presentations, guiding enhancements in diagnostic methods and therapeutic approaches.
Collapse
Affiliation(s)
- Dorsa Heydarlou
- Neurology, California Northstate University College of Medicine, Elk Grove, USA
| | - Arya Asghari
- Neurology, California Northstate University College of Medicine, Elk Grove, USA
| | - Shawyon Ezzati
- Neurology, California Northstate University College of Medicine, Elk Grove, USA
| | - Mariam Khalil
- Neurology, California Northstate University College of Medicine, Elk Grove, USA
| | | | - Forshing Lui
- Clinical Sciences, California Northstate University College of Medicine, Elk Grove, USA
| |
Collapse
|
10
|
Korinek M, Candelas Serra M, Abdel Rahman F, Dobrovolski M, Kuchtiak V, Abramova V, Fili K, Tomovic E, Hrcka Krausova B, Krusek J, Cerny J, Vyklicky L, Balik A, Smejkalova T. Disease-Associated Variants in GRIN1, GRIN2A and GRIN2B genes: Insights into NMDA Receptor Structure, Function, and Pathophysiology. Physiol Res 2024; 73:S413-S434. [PMID: 38836461 PMCID: PMC11412357 DOI: 10.33549/physiolres.935346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are a subtype of ionotropic glutamate receptors critical for synaptic transmission and plasticity, and for the development of neural circuits. Rare or de-novo variants in GRIN genes encoding NMDAR subunits have been associated with neurodevelopmental disorders characterized by intellectual disability, developmental delay, autism, schizophrenia, or epilepsy. In recent years, some disease-associated variants in GRIN genes have been characterized using recombinant receptors expressed in non-neuronal cells, and a few variants have also been studied in neuronal preparations or animal models. Here we review the current literature on the functional evaluation of human disease-associated variants in GRIN1, GRIN2A and GRIN2B genes at all levels of analysis. Focusing on the impact of different patient variants at the level of receptor function, we discuss effects on receptor agonist and co-agonist affinity, channel open probability, and receptor cell surface expression. We consider how such receptor-level functional information may be used to classify variants as gain-of-function or loss-of-function, and discuss the limitations of this classification at the synaptic, cellular, or system level. Together this work by many laboratories worldwide yields valuable insights into NMDAR structure and function, and represents significant progress in the effort to understand and treat GRIN disorders. Keywords: NMDA receptor , GRIN genes, Genetic variants, Electrophysiology, Synapse, Animal models.
Collapse
Affiliation(s)
- M Korinek
- Department of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Ng ACH, Scantlebury MH. Successful treatment of epileptic encephalopathy with spike wave activation in sleep with anakinra. Epilepsy Behav Rep 2024; 27:100678. [PMID: 38881883 PMCID: PMC11177074 DOI: 10.1016/j.ebr.2024.100678] [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: 02/23/2024] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 06/18/2024] Open
Abstract
Patients with epileptic encephalopathy with spike wave activation in sleep (EE-SWAS) often display drug-resistant epilepsy. The activation of epileptic activity during sleep is associated temporally with neurocognitive impairment and causes a spectrum of disorders within the epilepsy-aphasia syndrome. The prognosis is dependent on promptness of treatment and etiology. However, there is no clear consensus with regards to the optimal management for patients with EE-SWAS. We queried our Pediatric Epilepsy Outcome-Informatics Project (PEOIP) database for all patients treated with anakinra in our centre. We herein report a case of a female with EE-SWAS, who demonstrated remarkable neurocognitive improvement with anakinra. We suggest that a trial of anakinra may be an option for patients with EE-SWAS due to non-structural and possibly inflammatory etiology.
Collapse
Affiliation(s)
- Andy Cheuk-Him Ng
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Division of Neurology, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta and Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Morris H Scantlebury
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
12
|
Montanucci L, Brünger T, Bhattarai N, Boßelmann CM, Kim S, Allen JP, Zhang J, Klöckner C, Fariselli P, May P, Lemke JR, Myers SJ, Yuan H, Traynelis SF, Lal D. Distances from ligands as main predictive features for pathogenicity and functional effect of variants in NMDA receptors. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.06.24306939. [PMID: 38766179 PMCID: PMC11100844 DOI: 10.1101/2024.05.06.24306939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Genetic variants in genes GRIN1 , GRIN2A , GRIN2B , and GRIN2D , which encode subunits of the N-methyl-D-aspartate receptor (NMDAR), have been associated with severe and heterogeneous neurologic diseases. Missense variants in these genes can result in gain or loss of the NMDAR function, requiring opposite therapeutic treatments. Computational methods that predict pathogenicity and molecular functional effects are therefore crucial for accurate diagnosis and therapeutic applications. We assembled missense variants: 201 from patients, 631 from general population, and 159 characterized by electrophysiological readouts showing whether they can enhance or reduce the receptor function. This includes new functional data from 47 variants reported here, for the first time. We found that pathogenic/benign variants and variants that increase/decrease the channel function were distributed unevenly on the protein structure, with spatial proximity to ligands bound to the agonist and antagonist binding sites being key predictive features. Leveraging distances from ligands, we developed two independent machine learning-based predictors for NMDAR missense variants: a pathogenicity predictor which outperforms currently available predictors (AUC=0.945, MCC=0.726), and the first binary predictor of molecular function (increase or decrease) (AUC=0.809, MCC=0.523). Using these, we reclassified variants of uncertain significance in the ClinVar database and refined a previous genome-informed epidemiological model to estimate the birth incidence of molecular mechanism-defined GRIN disorders. Our findings demonstrate that distance from ligands is an important feature in NMDARs that can enhance variant pathogenicity prediction and enable functional prediction. Further studies with larger numbers of phenotypically and functionally characterized variants will enhance the potential clinical utility of this method.
Collapse
|
13
|
Morgan AT, Amor DJ, St John MD, Scheffer IE, Hildebrand MS. Genetic architecture of childhood speech disorder: a review. Mol Psychiatry 2024; 29:1281-1292. [PMID: 38366112 PMCID: PMC11189821 DOI: 10.1038/s41380-024-02409-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 02/18/2024]
Abstract
Severe speech disorders lead to poor literacy, reduced academic attainment and negative psychosocial outcomes. As early as the 1950s, the familial nature of speech disorders was recognized, implying a genetic basis; but the molecular genetic basis remained unknown. In 2001, investigation of a large three generational family with severe speech disorder, known as childhood apraxia of speech (CAS), revealed the first causative gene; FOXP2. A long hiatus then followed for CAS candidate genes, but in the past three years, genetic analysis of cohorts ascertained for CAS have revealed over 30 causative genes. A total of 36 pathogenic variants have been identified from 122 cases across 3 cohorts in this nascent field. All genes identified have been in coding regions to date, with no apparent benefit at this stage for WGS over WES in identifying monogenic conditions associated with CAS. Hence current findings suggest a remarkable one in three children have a genetic variant that explains their CAS, with significant genetic heterogeneity emerging. Around half of the candidate genes identified are currently supported by medium (6 genes) to strong (9 genes) evidence supporting the association between the gene and CAS. Despite genetic heterogeneity; many implicated proteins functionally converge on pathways involved in chromatin modification or transcriptional regulation, opening the door to precision diagnosis and therapies. Most of the new candidate genes for CAS are associated with previously described neurodevelopmental conditions that include intellectual disability, autism and epilepsy; broadening the phenotypic spectrum to a distinctly milder presentation defined by primary speech disorder in the setting of normal intellect. Insights into the genetic bases of CAS, a severe, rare speech disorder, are yet to translate to understanding the heritability of more common, typically milder forms of speech or language impairment such as stuttering or phonological disorder. These disorders likely follow complex inheritance with polygenic contributions in many cases, rather than the monogenic patterns that underly one-third of patients with CAS. Clinical genetic testing for should now be implemented for individuals with CAS, given its high diagnostic rate, which parallels many other neurodevelopmental disorders where this testing is already standard of care. The shared mechanisms implicated by gene discovery for CAS highlight potential new targets for future precision therapies.
Collapse
Affiliation(s)
- Angela T Morgan
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.
- Speech Pathology, University of Melbourne, Melbourne, VIC, Australia.
- Speech Pathology, Royal Children's Hospital, Melbourne, VIC, Australia.
| | - David J Amor
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Miya D St John
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Speech Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Ingrid E Scheffer
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Epilepsy Research Centre, Austin Health, Melbourne, VIC, Australia
| | - Michael S Hildebrand
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Epilepsy Research Centre, Austin Health, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Li GX, Chang RZ, Liu TT, Jin GN, Lu K, Yong TY, Li Z, Liu JH, Zhang B, Zhang WG, Ding ZY. GRIN2A mutation is a novel indicator of stratifying beneficiaries of immune checkpoint inhibitors in multiple cancers. Cancer Gene Ther 2024; 31:586-598. [PMID: 38267623 DOI: 10.1038/s41417-024-00730-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/26/2024]
Abstract
Glutamate-NMDAR receptors (GRINs) have been reported to influence cancer immunogenicity; however, the relationship between GRIN alterations and the response to immune checkpoint inhibitors (ICIs) has not been determined. This study combined clinical characteristics and mutational profiles from multiple cohorts to form a discovery cohort (n = 901). The aim of this study was to investigate the correlation between the mutation status of the GRIN gene and the response to ICI therapy. Additionally, an independent ICI-treated cohort from the Memorial Sloan Kettering Cancer Center (MSKCC, N = 1513) was used for validation. Furthermore, this study explored the associations between GRIN2A mutations and intrinsic and extrinsic immunity using multiomics analysis. In the discovery cohort, patients with GRIN2A-MUTs had improved clinical outcomes, as indicated by a higher objective response rate (ORR: 36.8% vs 25.8%, P = 0.020), durable clinical benefit (DCB: 55.2% vs 38.7%, P = 0.005), prolonged progression-free survival (PFS: HR = 0.65; 95% CI 0.49 to 0.87; P = 0.003), and increased overall survival (OS: HR = 0.67; 95% CI 0.50 to 0.89; P = 0.006). Similar results were observed in the validation cohort, in which GRIN2A-MUT patients exhibited a significant improvement in overall survival (HR = 0.66; 95% CI = 0.49 to 0.88; P = 0.005; adjusted P = 0.045). Moreover, patients with GRIN2A-MUTs exhibited an increase in tumor mutational burden, high expression of costimulatory molecules, increased activity of antigen-processing machinery, and infiltration of various immune cells. Additionally, gene sets associated with cell cycle regulation and the interferon response were enriched in GRIN2A-mutated tumors. In conclusion, GRIN2A mutation is a novel biomarker associated with a favorable response to ICIs in multiple cancers.
Collapse
Affiliation(s)
- Gan-Xun Li
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rui-Zhi Chang
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tong-Tong Liu
- Department of Anesthesiology, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guan-Nan Jin
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Kan Lu
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tu-Ying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430071, Hubei, China
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430071, Hubei, China
| | - Ji-Hong Liu
- Department and Institute of Urology, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bixiang Zhang
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wan-Guang Zhang
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Ze-Yang Ding
- Hepatic Surgery Center, and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, National Medical Center for Major Public Health Events, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
15
|
Lu Y, Mu L, Elstrott J, Fu C, Sun C, Su T, Ma X, Yan J, Jiang H, Hanson JE, Geng Y, Chen Y. Differential depletion of GluN2A induces heterogeneous schizophrenia-related phenotypes in mice. EBioMedicine 2024; 102:105045. [PMID: 38471394 PMCID: PMC10943646 DOI: 10.1016/j.ebiom.2024.105045] [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: 07/14/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Schizophrenia, a debilitating psychiatric disorder, displays considerable interindividual variation in clinical presentations. The ongoing debate revolves around whether this heterogeneity signifies a continuum of severity linked to a singular causative factor or a collection of distinct subtypes with unique origins. Within the realm of schizophrenia, the functional impairment of GluN2A, a subtype of the NMDA receptor, has been associated with an elevated risk. Despite GluN2A's expression across various neuronal types throughout the brain, its specific contributions to schizophrenia and its involvement in particular cell types or brain regions remain unexplored. METHODS We generated age-specific, cell type-specific or brain region-specific conditional knockout mice targeting GluN2A and conducted a comprehensive analysis using tests measuring phenotypes relevant to schizophrenia. FINDINGS Through the induction of germline ablation of GluN2A, we observed the emergence of numerous schizophrenia-associated abnormalities in adult mice. Intriguingly, GluN2A knockout performed at different ages, in specific cell types and within distinct brain regions, we observed overlapping yet distinct schizophrenia-related phenotypes in mice. INTERPRETATION Our interpretation suggests that the dysfunction of GluN2A is sufficient to evoke heterogeneous manifestations associated with schizophrenia, indicating that GluN2A stands as a prominent risk factor and a potential therapeutic target for schizophrenia. FUNDING This project received support from the Shanghai Municipal Science and Technology Major Project (Grant No. 2019SHZDZX02) awarded to Y.C. and the Natural Science Foundation of Shanghai (Grant No. 19ZR1468600 and 201409003800) awarded to G.Y.
Collapse
Affiliation(s)
- Yi Lu
- Interdisciplinary Research Centre on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, No.100 Haike Rd., Pudong New District, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Longyu Mu
- Interdisciplinary Research Centre on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, No.100 Haike Rd., Pudong New District, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Justin Elstrott
- Department of Translational Imaging, Genentech Inc., South San Francisco, CA 94080, USA
| | - Chaoying Fu
- Interdisciplinary Research Centre on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, No.100 Haike Rd., Pudong New District, Shanghai 201210, China
| | - Cailu Sun
- Interdisciplinary Research Centre on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, No.100 Haike Rd., Pudong New District, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tonghui Su
- Interdisciplinary Research Centre on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, No.100 Haike Rd., Pudong New District, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaofan Ma
- Department of Anaesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai 200011, China
| | - Jia Yan
- Department of Anaesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai 200011, China
| | - Hong Jiang
- Department of Anaesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, Shanghai 200011, China
| | - Jesse E Hanson
- Department of Neuroscience, Genentech Inc., South San Francisco, CA 94080, USA
| | - Yang Geng
- Interdisciplinary Research Centre on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, No.100 Haike Rd., Pudong New District, Shanghai 201210, China.
| | - Yelin Chen
- Interdisciplinary Research Centre on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, No.100 Haike Rd., Pudong New District, Shanghai 201210, China.
| |
Collapse
|
16
|
Lin ZJ, Li B, Lin PX, Song W, Yan LM, Meng H, He N. Clinical application of trio-based whole-exome sequencing in idiopathic generalized epilepsy. Seizure 2024; 116:24-29. [PMID: 36842888 DOI: 10.1016/j.seizure.2023.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/13/2023] Open
Abstract
PURPOSE Idiopathic generalized epilepsies (IGEs) are a common group of genetic generalized epilepsies with high genetic heterogeneity and complex inheritance. However, the genetic basis is still largely unknown. This study aimed to explore the genetic etiologies in IGEs. METHODS Trio-based whole-exome sequencing was performed in 60 cases with IGEs. The pathogenicity of candidate genetic variants was evaluated by the criteria of the American College of Medical Genetics and Genomics (ACMG), and the clinical causality was assessed by concordance between the observed phenotype and the reported phenotype. RESULTS Seven candidate variants were detected in seven unrelated cases with IGE (11.7%, 7/60). According to ACMG, a de novo SLC2A1 (c.376C>T/p.Arg126Cys) variant identified in childhood absence epilepsy was evaluated as pathogenic with clinical concordance. Six variants were assessed to be uncertain significance by ACMG, but then considered causative after evaluation of clinical concordance. These variants included CLCN4 hemizygous variant (c.2044G>A/p.Glu682Lys) and IQSEC2 heterozygous variant (c.4315C>T/p.Pro1439Ser) in juvenile absence epilepsy, EFHC1 variant (c.1504C>T/p.Arg502Trp) and CACNA1H (c.589G>T/p.Ala197Ser) both with incomplete penetrance in juvenile myoclonic epilepsy, and GRIN2A variant (c.2011C>G/p.Gln671Glu) and GABRB1 variant (c.1075G>A/p.Val359Ile) both co-segregated with juvenile myoclonic epilepsy. Among them, GABRB1 was for the first time identified as potential novel causative gene for IGE. SIGNIFICANCE Considering the genetic heterogeneity and complex inheritance of IGEs, a comprehensive evaluation combined the ACMG scoring and assessment of clinical concordance is suggested for the pathogenicity analysis of variants identified in clinical screening. GABRB1 is probably a novel causative gene for IGE, which warrants further studies.
Collapse
Affiliation(s)
- Zhi-Jian Lin
- Department of Neurology, the Affiliated Hospital of Putian University, Putian 351100, Fujian Province, China; Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Bin Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Peng-Xing Lin
- Department of Neurology, the Affiliated Hospital of Putian University, Putian 351100, Fujian Province, China
| | - Wang Song
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Li-Min Yan
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, 570311 Haikou, Hainan, China
| | - Heng Meng
- Department of Neurology, the First Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| | - Na He
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
17
|
Iacobucci GJ, Popescu GK. Calcium- and calmodulin-dependent inhibition of NMDA receptor currents. Biophys J 2024; 123:277-293. [PMID: 38140727 PMCID: PMC10870176 DOI: 10.1016/j.bpj.2023.12.018] [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: 09/18/2023] [Revised: 12/05/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023] Open
Abstract
Calcium ions (Ca2+) reduce NMDA receptor currents through several distinct mechanisms. Among these, calmodulin (CaM)-dependent inhibition (CDI) accomplishes rapid, reversible, and incomplete reduction of the NMDA receptor currents in response to elevations in intracellular Ca2+. Quantitative and mechanistic descriptions of CDI of NMDA receptor-mediated signals have been marred by variability originating, in part, from differences in the conditions and metrics used to evaluate this process across laboratories. Recent ratiometric approaches to measure the magnitude and kinetics of NMDA receptor CDI have facilitated rapid insights into this phenomenon. Notably, the kinetics and magnitude of NMDA receptor CDI depend on the degree of saturation of its CaM binding sites, which represent the bona fide calcium sensor for this type of inhibition, the kinetics and magnitude of the Ca2+ signal, which depends on the biophysical properties of the NMDA receptor or of adjacent Ca2+ sources, and on the relative distribution of Ca2+ sources and CaM molecules. Given that all these factors vary widely during development, across cell types, and with physiological and pathological states, it is important to understand how NMDA receptor CDI develops and how it contributes to signaling in the central nervous system. Here, we review briefly these recent advances and highlight remaining questions about the structural and kinetic mechanisms of NMDA receptor CDI. Given that pathologies can arise from several sources, including mutations in the NMDA receptor and in CaM, understanding how CaM responds to intracellular Ca2+ signals to initiate conformational changes in NMDA receptors, and mapping the structural domains responsible will help to envision novel therapeutic strategies to neuropsychiatric diseases, which presently have limited available treatments.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, New York
| | - Gabriela K Popescu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, New York.
| |
Collapse
|
18
|
Shepard N, Baez-Nieto D, Iqbal S, Kurganov E, Budnik N, Campbell AJ, Pan JQ, Sheng M, Farsi Z. Differential functional consequences of GRIN2A mutations associated with schizophrenia and neurodevelopmental disorders. Sci Rep 2024; 14:2798. [PMID: 38307912 PMCID: PMC10837427 DOI: 10.1038/s41598-024-53102-3] [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: 08/01/2023] [Accepted: 01/27/2024] [Indexed: 02/04/2024] Open
Abstract
Human genetic studies have revealed rare missense and protein-truncating variants in GRIN2A, encoding for the GluN2A subunit of the NMDA receptors, that confer significant risk for schizophrenia (SCZ). Mutations in GRIN2A are also associated with epilepsy and developmental delay/intellectual disability (DD/ID). However, it remains enigmatic how alterations to the same protein can result in diverse clinical phenotypes. Here, we performed functional characterization of human GluN1/GluN2A heteromeric NMDA receptors that contain SCZ-linked GluN2A variants, and compared them to NMDA receptors with GluN2A variants associated with epilepsy or DD/ID. Our findings demonstrate that SCZ-associated GRIN2A variants were predominantly loss-of-function (LoF), whereas epilepsy and DD/ID-associated variants resulted in both gain- and loss-of-function phenotypes. We additionally show that M653I and S809R, LoF GRIN2A variants associated with DD/ID, exert a dominant-negative effect when co-expressed with a wild-type GluN2A, whereas E58Ter and Y698C, SCZ-linked LoF variants, and A727T, an epilepsy-linked LoF variant, do not. These data offer a potential mechanism by which SCZ/epilepsy and DD/ID-linked variants can cause different effects on receptor function and therefore result in divergent pathological outcomes.
Collapse
Affiliation(s)
- Nate Shepard
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David Baez-Nieto
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sumaiya Iqbal
- The Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Erkin Kurganov
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nikita Budnik
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Arthur J Campbell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jen Q Pan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Morgan Sheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Zohreh Farsi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
19
|
Posar A, Visconti P. Continuous Spike-Waves during Slow Sleep Today: An Update. CHILDREN (BASEL, SWITZERLAND) 2024; 11:169. [PMID: 38397281 PMCID: PMC10887038 DOI: 10.3390/children11020169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/19/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024]
Abstract
In the context of childhood epilepsy, the concept of continuous spike-waves during slow sleep (CSWS) includes several childhood-onset heterogeneous conditions that share electroencephalograms (EEGs) characterized by a high frequency of paroxysmal abnormalities during sleep, which have negative effects on the cognitive development and behavior of the child. These negative effects may have the characteristics of a clear regression or of a slowdown in development. Seizures are very often present, but not constantly. The above makes it clear why CSWS have been included in epileptic encephalopathies, in which, by definition, frequent EEG paroxysmal abnormalities have an unfavorable impact on cognitive functions, including socio-communicative skills, causing autistic features, even regardless of the presence of clinically overt seizures. Although several decades have passed since the original descriptions of the electroclinical condition of CSWS, there are still many areas that are little-known and deserve to be further studied, including the EEG diagnostic criteria, the most effective electrophysiological parameter for monitoring the role of the thalamus in CSWS pathogenesis, its long-term evolution, the nosographic location of Landau-Kleffner syndrome, standardized neuropsychological and behavioral assessments, and pharmacological and non-pharmacological therapies.
Collapse
Affiliation(s)
- Annio Posar
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOSI Disturbi dello Spettro Autistico, 40139 Bologna, Italy;
- Department of Biomedical and Neuromotor Sciences, Bologna University, 40139 Bologna, Italy
| | - Paola Visconti
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOSI Disturbi dello Spettro Autistico, 40139 Bologna, Italy;
| |
Collapse
|
20
|
Licchetta L, Di Giorgi L, Santucci M, Taruffi L, Stipa C, Minardi R, Carelli V, Bisulli F. Biallelic pathogenic variants of PARS2 cause developmental and epileptic encephalopathy with spike-and-wave activation in sleep. Mol Genet Genomic Med 2024; 12:e2311. [PMID: 38087948 PMCID: PMC10767575 DOI: 10.1002/mgg3.2311] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Biallelic pathogenic variants in the mitochondrial prolyl-tRNA synthetase 2 gene (PARS2, OMIM * 612036) have been associated with Developmental and Epileptic Encephalopathy-75 (DEE-75, MIM #618437). This condition is typically characterized by early-onset refractory infantile spasms with hypsarrhythmia, intellectual disability, microcephaly, cerebral atrophy with hypomyelination, lactic acidemia, and cardiomyopathy. Most affected individuals do not survive beyond the age of 10 years. METHODS We describe a patient with early-onset DEE, consistently showing an EEG pattern of Spike-and-Wave Activation in Sleep (SWAS) since childhood. The patient underwent extensive clinical, metabolic and genetic investigations, including whole exome sequencing (WES). RESULTS WES analysis identified compound heterozygous variants in PARS2 that have been already reported as pathogenic. A literature review of PARS2-associated DEE, focusing mainly on the electroclinical phenotype, did not reveal the association of SWAS with pathogenic variants in PARS2. Notably, unlike previously reported cases with the same genotype, this patient had longer survival without cardiac involvement or lactic acidosis, suggesting potential genetic modifiers contributing to disease variability. CONCLUSION These findings widen the genetic heterogeneity of DEE-SWAS, including PARS2 as a causative gene in this syndromic entity, and highlight the importance of prolonged sleep EEG recording for the recognition of SWAS as a possible electroclinical evolution of PARS2-related DEE.
Collapse
Affiliation(s)
- Laura Licchetta
- IRCCS Istituto delle Scienze Neurologiche di BolognaFull member of the European Reference Network EpiCARE BolognaBolognaItaly
| | - Lucia Di Giorgi
- IRCCS Istituto delle Scienze Neurologiche di BolognaFull member of the European Reference Network EpiCARE BolognaBolognaItaly
- Department of Biomedicine, Neuroscience and Advanced DiagnosticsUniversity of PalermoPalermoItaly
| | - Margherita Santucci
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Lisa Taruffi
- IRCCS Istituto delle Scienze Neurologiche di BolognaFull member of the European Reference Network EpiCARE BolognaBolognaItaly
| | - Carlotta Stipa
- IRCCS Istituto delle Scienze Neurologiche di BolognaFull member of the European Reference Network EpiCARE BolognaBolognaItaly
| | - Raffaella Minardi
- IRCCS Istituto delle Scienze Neurologiche di BolognaFull member of the European Reference Network EpiCARE BolognaBolognaItaly
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di BolognaFull member of the European Reference Network EpiCARE BolognaBolognaItaly
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Francesca Bisulli
- IRCCS Istituto delle Scienze Neurologiche di BolognaFull member of the European Reference Network EpiCARE BolognaBolognaItaly
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| |
Collapse
|
21
|
Peall KJ, Owen MJ, Hall J. Rare genetic brain disorders with overlapping neurological and psychiatric phenotypes. Nat Rev Neurol 2024; 20:7-21. [PMID: 38001363 DOI: 10.1038/s41582-023-00896-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 11/26/2023]
Abstract
Understanding rare genetic brain disorders with overlapping neurological and psychiatric phenotypes is of increasing importance given the potential for developing disease models that could help to understand more common, polygenic disorders. However, the traditional clinical boundaries between neurology and psychiatry result in frequent segregation of these disorders into distinct silos, limiting cross-specialty understanding that could facilitate clinical and biological advances. In this Review, we highlight multiple genetic brain disorders in which neurological and psychiatric phenotypes are observed, but for which in-depth, cross-spectrum clinical phenotyping is rarely undertaken. We describe the combined phenotypes observed in association with genetic variants linked to epilepsy, dystonia, autism spectrum disorder and schizophrenia. We also consider common underlying mechanisms that centre on synaptic plasticity, including changes to synaptic and neuronal structure, calcium handling and the balance of excitatory and inhibitory neuronal activity. Further investigation is needed to better define and replicate these phenotypes in larger cohorts, which would help to gain greater understanding of the pathophysiological mechanisms and identify common therapeutic targets.
Collapse
Affiliation(s)
- Kathryn J Peall
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK.
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.
| | - Michael J Owen
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
22
|
Gjerulfsen CE, Krey I, Klöckner C, Rubboli G, Lemke JR, Møller RS. Spectrum of NMDA Receptor Variants in Neurodevelopmental Disorders and Epilepsy. Methods Mol Biol 2024; 2799:1-11. [PMID: 38727899 DOI: 10.1007/978-1-0716-3830-9_1] [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] [Indexed: 07/03/2024]
Abstract
N-methyl-D-aspartate receptors (NMDAR) are ligand-gated ion channels mediating excitatory neurotransmission and are important for normal brain development, cognitive abilities, and motor functions. Pathogenic variants in the Glutamate receptor Ionotropic N-methyl-D-aspartate (GRIN) genes (GRIN1, GRIN2A-D) encoding NMDAR subunits have been associated with a wide spectrum of neurodevelopmental disorders and epilepsies ranging from treatable focal epilepsies to devastating early-onset developmental and epileptic encephalopathies. Genetic variants in NMDA receptor genes can cause a range of complex alterations to receptor properties resulting in various degrees of loss-of-function, gain-of-function, or mixtures thereof. Understanding how genetic variants affect the function of the receptors, therefore, represents an important first step in the ongoing development towards targeted therapies. Currently, targeted treatment options for GRIN-related diseases are limited. However, treatment with memantine has been reported to significantly reduce seizure frequency in a few individuals with developmental and epileptic encephalopathies harboring de novo gain-of-function GRIN2A missense variants, and supplementary treatment with L-serine has been associated with improved motor and cognitive performance as well as reduced seizure frequency in patients with GRIN2B loss-of-function missense variants as well as GRIN2A and GRIN2B null variants.
Collapse
Affiliation(s)
- Cathrine E Gjerulfsen
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark
| | - Ilona Krey
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Chiara Klöckner
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Guido Rubboli
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark.
- Department of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
23
|
Hanson JE, Yuan H, Perszyk RE, Banke TG, Xing H, Tsai MC, Menniti FS, Traynelis SF. Therapeutic potential of N-methyl-D-aspartate receptor modulators in psychiatry. Neuropsychopharmacology 2024; 49:51-66. [PMID: 37369776 PMCID: PMC10700609 DOI: 10.1038/s41386-023-01614-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/24/2023] [Accepted: 05/15/2023] [Indexed: 06/29/2023]
Abstract
N-methyl-D-aspartate (NMDA) receptors mediate a slow component of excitatory synaptic transmission, are widely distributed throughout the central nervous system, and regulate synaptic plasticity. NMDA receptor modulators have long been considered as potential treatments for psychiatric disorders including depression and schizophrenia, neurodevelopmental disorders such as Rett Syndrome, and neurodegenerative conditions such as Alzheimer's disease. New interest in NMDA receptors as therapeutic targets has been spurred by the findings that certain inhibitors of NMDA receptors produce surprisingly rapid and robust antidepressant activity by a novel mechanism, the induction of changes in the brain that well outlast the presence of drug in the body. These findings are driving research into an entirely new paradigm for using NMDA receptor antagonists in a host of related conditions. At the same time positive allosteric modulators of NMDA receptors are being pursued for enhancing synaptic function in diseases that feature NMDA receptor hypofunction. While there is great promise, developing the therapeutic potential of NMDA receptor modulators must also navigate the potential significant risks posed by the use of such agents. We review here the emerging pharmacology of agents that target different NMDA receptor subtypes, offering new avenues for capturing the therapeutic potential of targeting this important receptor class.
Collapse
Affiliation(s)
- Jesse E Hanson
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Riley E Perszyk
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Tue G Banke
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Hao Xing
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ming-Chi Tsai
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Frank S Menniti
- MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA.
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
24
|
McTague A, Scheffer IE, Kullmann DM, Sisodiya S. Epilepsies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 203:157-184. [PMID: 39174247 DOI: 10.1016/b978-0-323-90820-7.00016-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Recent advances in genetic diagnosis have revealed the underlying etiology of many epilepsies and have identified pathogenic, causative variants in numerous ion and ligand-gated channel genes. This chapter describes the clinical presentations of epilepsy associated with different channelopathies including classic electroclinical syndromes and emerging gene-specific phenotypes. Also discussed are the archetypal epilepsy channelopathy, SCN1A-Dravet syndrome, considering the expanding phenotype. Clinical presentations where a channelopathy is suspected, such as sleep-related hypermotor epilepsy and epilepsy in association with movement disorders, are reviewed. Channelopathies pose an intriguing problem for the development of gene therapies. Design of targeted therapies requires physiologic insights into the often multifaceted impact of a pathogenic variant, coupled with an understanding of the phenotypic spectrum of a gene. As gene-specific novel therapies come online for the channelopathies, it is essential that clinicians are able to recognize epilepsy phenotypes likely to be due to channelopathy and institute early genetic testing in both children and adults. These findings are likely to have immediate management implications and to inform prognostic and reproductive counseling.
Collapse
Affiliation(s)
- Amy McTague
- Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, United Kingdom; Department of Neurology, Great Ormond Street Hospital for Children, London, United Kingdom.
| | - Ingrid E Scheffer
- Austin Health and Royal Children's Hospital, Florey and Murdoch Children's Research Institutes, University of Melbourne, Melbourne, VIC, Australia
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom; National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
| | - Sanjay Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom; National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
| |
Collapse
|
25
|
Varlamova EG, Borisova EV, Evstratova YA, Newman AG, Kuldaeva VP, Gavrish MS, Kondakova EV, Tarabykin VS, Babaev AA, Turovsky EA. Socrates: A Novel N-Ethyl-N-nitrosourea-Induced Mouse Mutant with Audiogenic Epilepsy. Int J Mol Sci 2023; 24:17104. [PMID: 38069426 PMCID: PMC10707124 DOI: 10.3390/ijms242317104] [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: 10/25/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Epilepsy is one of the common neurological diseases that affects not only adults but also infants and children. Because epilepsy has been studied for a long time, there are several pharmacologically effective anticonvulsants, which, however, are not suitable as therapy for all patients. The genesis of epilepsy has been extensively investigated in terms of its occurrence after injury and as a concomitant disease with various brain diseases, such as tumors, ischemic events, etc. However, in the last decades, there are multiple reports that both genetic and epigenetic factors play an important role in epileptogenesis. Therefore, there is a need for further identification of genes and loci that can be associated with higher susceptibility to epileptic seizures. Use of mouse knockout models of epileptogenesis is very informative, but it has its limitations. One of them is due to the fact that complete deletion of a gene is not, in many cases, similar to human epilepsy-associated syndromes. Another approach to generating mouse models of epilepsy is N-Ethyl-N-nitrosourea (ENU)-directed mutagenesis. Recently, using this approach, we generated a novel mouse strain, soc (socrates, formerly s8-3), with epileptiform activity. Using molecular biology methods, calcium neuroimaging, and immunocytochemistry, we were able to characterize the strain. Neurons isolated from soc mutant brains retain the ability to differentiate in vitro and form a network. However, soc mutant neurons are characterized by increased spontaneous excitation activity. They also demonstrate a high degree of Ca2+ activity compared to WT neurons. Additionally, they show increased expression of NMDA receptors, decreased expression of the Ca2+-conducting GluA2 subunit of AMPA receptors, suppressed expression of phosphoinositol 3-kinase, and BK channels of the cytoplasmic membrane involved in protection against epileptogenesis. During embryonic and postnatal development, the expression of several genes encoding ion channels is downregulated in vivo, as well. Our data indicate that soc mutation causes a disruption of the excitation-inhibition balance in the brain, and it can serve as a mouse model of epilepsy.
Collapse
Affiliation(s)
- Elena G. Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia;
| | - Ekaterina V. Borisova
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (E.V.B.); (A.G.N.)
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (V.P.K.); (M.S.G.); (E.V.K.); (A.A.B.)
| | - Yuliya A. Evstratova
- Federal State Budgetary Educational Institution of Higher Education “MIREA—Russian Technological University”, 78, Vernadskogo Ave., 119454 Moscow, Russia;
| | - Andrew G. Newman
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (E.V.B.); (A.G.N.)
| | - Vera P. Kuldaeva
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (V.P.K.); (M.S.G.); (E.V.K.); (A.A.B.)
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 10 Nab. Ushaiki, 634050 Tomsk, Russia
| | - Maria S. Gavrish
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (V.P.K.); (M.S.G.); (E.V.K.); (A.A.B.)
| | - Elena V. Kondakova
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (V.P.K.); (M.S.G.); (E.V.K.); (A.A.B.)
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 10 Nab. Ushaiki, 634050 Tomsk, Russia
| | - Victor S. Tarabykin
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (E.V.B.); (A.G.N.)
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (V.P.K.); (M.S.G.); (E.V.K.); (A.A.B.)
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, 10 Nab. Ushaiki, 634050 Tomsk, Russia
| | - Alexey A. Babaev
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (V.P.K.); (M.S.G.); (E.V.K.); (A.A.B.)
| | - Egor A. Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia;
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia; (V.P.K.); (M.S.G.); (E.V.K.); (A.A.B.)
| |
Collapse
|
26
|
Bortolami A, Sesti F. Ion channels in neurodevelopment: lessons from the Integrin-KCNB1 channel complex. Neural Regen Res 2023; 18:2365-2369. [PMID: 37282454 PMCID: PMC10360111 DOI: 10.4103/1673-5374.371347] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
Ion channels modulate cellular excitability by regulating ionic fluxes across biological membranes. Pathogenic mutations in ion channel genes give rise to epileptic disorders that are among the most frequent neurological diseases affecting millions of individuals worldwide. Epilepsies are triggered by an imbalance between excitatory and inhibitory conductances. However, pathogenic mutations in the same allele can give rise to loss-of-function and/or gain-of-function variants, all able to trigger epilepsy. Furthermore, certain alleles are associated with brain malformations even in the absence of a clear electrical phenotype. This body of evidence argues that the underlying epileptogenic mechanisms of ion channels are more diverse than originally thought. Studies focusing on ion channels in prenatal cortical development have shed light on this apparent paradox. The picture that emerges is that ion channels play crucial roles in landmark neurodevelopmental processes, including neuronal migration, neurite outgrowth, and synapse formation. Thus, pathogenic channel mutants can not only cause epileptic disorders by altering excitability, but further, by inducing morphological and synaptic abnormalities that are initiated during neocortex formation and may persist into the adult brain.
Collapse
Affiliation(s)
- Alessandro Bortolami
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, West Piscataway, NJ, USA
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, West Piscataway, NJ, USA
| |
Collapse
|
27
|
Pavinato L, Stanic J, Barzasi M, Gurgone A, Chiantia G, Cipriani V, Eberini I, Palazzolo L, Di Luca M, Costa A, Marcantoni A, Biamino E, Spada M, Hiatt SM, Kelley WV, Vestito L, Sisodiya SM, Efthymiou S, Chand P, Kaiyrzhanov R, Bruselles A, Cardaropoli S, Tartaglia M, De Rubeis S, Buxbaum JD, Smedley D, Ferrero GB, Giustetto M, Gardoni F, Brusco A. Missense variants in RPH3A cause defects in excitatory synaptic function and are associated with a clinically variable neurodevelopmental disorder. Genet Med 2023; 25:100922. [PMID: 37403762 DOI: 10.1016/j.gim.2023.100922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 06/22/2023] [Accepted: 06/25/2023] [Indexed: 07/06/2023] Open
Abstract
PURPOSE RPH3A encodes a protein involved in the stabilization of GluN2A subunit of N-methyl-D-aspartate (NMDA)-type glutamate receptors at the cell surface, forming a complex essential for synaptic plasticity and cognition. We investigated the effect of variants in RPH3A in patients with neurodevelopmental disorders. METHODS By using trio-based exome sequencing, GeneMatcher, and screening of 100,000 Genomes Project data, we identified 6 heterozygous variants in RPH3A. In silico and in vitro models, including rat hippocampal neuronal cultures, have been used to characterize the effect of the variants. RESULTS Four cases had a neurodevelopmental disorder with untreatable epileptic seizures [p.(Gln73His)dn; p.(Arg209Lys); p.(Thr450Ser)dn; p.(Gln508His)], and 2 cases [p.(Arg235Ser); p.(Asn618Ser)dn] showed high-functioning autism spectrum disorder. Using neuronal cultures, we demonstrated that p.(Thr450Ser) and p.(Asn618Ser) reduce the synaptic localization of GluN2A; p.(Thr450Ser) also increased the surface levels of GluN2A. Electrophysiological recordings showed increased GluN2A-dependent NMDA ionotropic glutamate receptor currents for both variants and alteration of postsynaptic calcium levels. Finally, expression of the Rph3AThr450Ser variant in neurons affected dendritic spine morphology. CONCLUSION Overall, we provide evidence that missense gain-of-function variants in RPH3A increase GluN2A-containing NMDA ionotropic glutamate receptors at extrasynaptic sites, altering synaptic function and leading to a clinically variable neurodevelopmental presentation ranging from untreatable epilepsy to autism spectrum disorder.
Collapse
Affiliation(s)
- Lisa Pavinato
- Department of Medical Sciences, University of Turin, Turin, Italy; Institute of Oncology Research (IOR), Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland
| | - Jennifer Stanic
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Marta Barzasi
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Antonia Gurgone
- Department of Neuroscience, University of Turin, Turin, Italy
| | | | - Valentina Cipriani
- William Harvey Research Institute, Clinical Pharmacology Precision Medicine, Queen Mary University of London, Charterhouse Square, United Kingdom
| | - Ivano Eberini
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Luca Palazzolo
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Alex Costa
- Department of Biosciences, University of the Studies of Milan, Milan, Italy; Institute of Biophysics, Consiglio Nazionale delle Ricerche (CNR), Milan, Italy
| | - Andrea Marcantoni
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Elisa Biamino
- Department of Pediatrics, Regina Margherita Children Hospital, Turin, Italy
| | - Marco Spada
- Department of Pediatrics, Regina Margherita Children Hospital, Turin, Italy
| | - Susan M Hiatt
- HudsonAlpha Institute for Biotechnology, Huntsville, AL
| | | | - Letizia Vestito
- William Harvey Research Institute, Clinical Pharmacology Precision Medicine, Queen Mary University of London, Charterhouse Square, United Kingdom
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom; Chalfont Centre for Epilepsy Bucks, Chalfont St Peter, United Kingdom
| | - Stephanie Efthymiou
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Prem Chand
- Department of Paediatric and Child Health, Aga Khan University Hospital, Karachi, Pakistan
| | - Rauan Kaiyrzhanov
- University College London, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Cardaropoli
- Department of Public Health and Pediatric Sciences, University of Torino, Torino, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Damian Smedley
- William Harvey Research Institute, Clinical Pharmacology Precision Medicine, Queen Mary University of London, Charterhouse Square, United Kingdom
| | | | | | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, DiSFeB, University of the Studies of Milan, Milan, Italy
| | - Alfredo Brusco
- Department of Medical Sciences, University of Turin, Turin, Italy; Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Turin, Italy.
| |
Collapse
|
28
|
Boerma T, Ter Haar S, Ganga R, Wijnen F, Blom E, Wierenga CJ. What risk factors for Developmental Language Disorder can tell us about the neurobiological mechanisms of language development. Neurosci Biobehav Rev 2023; 154:105398. [PMID: 37741516 DOI: 10.1016/j.neubiorev.2023.105398] [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: 04/21/2023] [Revised: 07/03/2023] [Accepted: 09/17/2023] [Indexed: 09/25/2023]
Abstract
Language is a complex multidimensional cognitive system that is connected to many neurocognitive capacities. The development of language is therefore strongly intertwined with the development of these capacities and their neurobiological substrates. Consequently, language problems, for example those of children with Developmental Language Disorder (DLD), are explained by a variety of etiological pathways and each of these pathways will be associated with specific risk factors. In this review, we attempt to link previously described factors that may interfere with language development to putative underlying neurobiological mechanisms of language development, hoping to uncover openings for future therapeutical approaches or interventions that can help children to optimally develop their language skills.
Collapse
Affiliation(s)
- Tessel Boerma
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands
| | - Sita Ter Haar
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands; Cognitive Neurobiology and Helmholtz Institute, Department of Psychology, Utrecht University/Translational Neuroscience, University Medical Center Utrecht, the Netherlands
| | - Rachida Ganga
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands
| | - Frank Wijnen
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands
| | - Elma Blom
- Department of Development and Education of youth in Diverse Societies (DEEDS), Utrecht University, Utrecht, the Netherlands; Department of Language and Culture, The Arctic University of Norway UiT, Tromsø, Norway.
| | - Corette J Wierenga
- Biology Department, Faculty of Science, Utrecht University, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
29
|
Camp CR, Vlachos A, Klöckner C, Krey I, Banke TG, Shariatzadeh N, Ruggiero SM, Galer P, Park KL, Caccavano A, Kimmel S, Yuan X, Yuan H, Helbig I, Benke TA, Lemke JR, Pelkey KA, McBain CJ, Traynelis SF. Loss of Grin2a causes a transient delay in the electrophysiological maturation of hippocampal parvalbumin interneurons. Commun Biol 2023; 6:952. [PMID: 37723282 PMCID: PMC10507040 DOI: 10.1038/s42003-023-05298-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 08/29/2023] [Indexed: 09/20/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ligand-gated ionotropic glutamate receptors that mediate a calcium-permeable component to fast excitatory neurotransmission. NMDARs are heterotetrameric assemblies of two obligate GluN1 subunits (GRIN1) and two GluN2 subunits (GRIN2A-GRIN2D). Sequencing data shows that 43% (297/679) of all currently known NMDAR disease-associated genetic variants are within the GRIN2A gene, which encodes the GluN2A subunit. Here, we show that unlike missense GRIN2A variants, individuals affected with disease-associated null GRIN2A variants demonstrate a transient period of seizure susceptibility that begins during infancy and diminishes near adolescence. We show increased circuit excitability and CA1 pyramidal cell output in juvenile mice of both Grin2a+/- and Grin2a-/- mice. These alterations in somatic spiking are not due to global upregulation of most Grin genes (including Grin2b). Deeper evaluation of the developing CA1 circuit led us to uncover age- and Grin2a gene dosing-dependent transient delays in the electrophysiological maturation programs of parvalbumin (PV) interneurons. We report that Grin2a+/+ mice reach PV cell electrophysiological maturation between the neonatal and juvenile neurodevelopmental timepoints, with Grin2a+/- mice not reaching PV cell electrophysiological maturation until preadolescence, and Grin2a-/- mice not reaching PV cell electrophysiological maturation until adulthood. Overall, these data may represent a molecular mechanism describing the transient nature of seizure susceptibility in disease-associated null GRIN2A patients.
Collapse
Affiliation(s)
- Chad R Camp
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Anna Vlachos
- Section on Cellular and Synaptic Physiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chiara Klöckner
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Ilona Krey
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Tue G Banke
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Nima Shariatzadeh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Sarah M Ruggiero
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- The Epilepsy NeuroGenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Peter Galer
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA
| | - Kristen L Park
- University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA
| | - Adam Caccavano
- Section on Cellular and Synaptic Physiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sarah Kimmel
- Section on Cellular and Synaptic Physiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiaoqing Yuan
- Section on Cellular and Synaptic Physiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Functional Evaluation of Rare Variants, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ingo Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- The Epilepsy NeuroGenetics Initiative, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Tim A Benke
- University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
- Center for Rare Diseases, University of Leipzig Medical Center, Leipzig, Germany
| | - Kenneth A Pelkey
- Section on Cellular and Synaptic Physiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chris J McBain
- Section on Cellular and Synaptic Physiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Functional Evaluation of Rare Variants, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
30
|
Chinappen DM, Ostrowski LM, Spencer ER, Kwon H, Kramer MA, Hämäläinen MS, Chu CJ. Decreased thalamocortical connectivity in resolved Rolandic epilepsy. Clin Neurophysiol 2023; 153:21-27. [PMID: 37419052 PMCID: PMC10520846 DOI: 10.1016/j.clinph.2023.05.013] [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: 03/28/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 07/09/2023]
Abstract
OBJECTIVE Median nerve somatosensory evoked fields (SEFs) conduction times reflect the integrity of neural transmission across the thalamocortical circuit. We hypothesized median nerve SEF conduction time would be abnormal in children with Rolandic epilepsy (RE). METHODS 22 children with RE (10 active; 12 resolved) and 13 age-matched controls underwent structural and diffusion MRI and median nerve and visual stimulation during magnetoencephalography (MEG). N20 SEF responses were identified in contralateral somatosensory cortices. P100 were identified in contralateral occipital cortices as controls. Conduction times were compared between groups in linear models controlling for height. N20 conduction time was also compared to thalamic volume and Rolandic thalamocortical structural connectivity inferred using probabilistic tractography. RESULTS The RE group had slower N20 conduction compared to controls (p = 0.042, effect size 0.6 ms) and this difference was driven by the resolved RE group (p = 0.046). There was no difference in P100 conduction time between groups (p = 0.83). Ventral thalamic volume positively correlated with N20 conduction time (p = 0.014). CONCLUSIONS Children with resolved RE have focally decreased Rolandic thalamocortical connectivity. SIGNIFICANCE These results identify a persistent focal thalamocortical circuit abnormality in resolved RE and suggest that decreased Rolandic thalamocortical connectivity may support symptom resolution in this self-limited epilepsy.
Collapse
Affiliation(s)
- Dhinakaran M Chinappen
- Massachusetts General Hospital, Department of Neurology, Boston, MA 02114, USA; Graduate Program in Neuroscience, Boston University, Boston, MA 02215, USA.
| | - Lauren M Ostrowski
- Massachusetts General Hospital, Department of Neurology, Boston, MA 02114, USA
| | - Elizabeth R Spencer
- Massachusetts General Hospital, Department of Neurology, Boston, MA 02114, USA; Graduate Program in Neuroscience, Boston University, Boston, MA 02215, USA
| | - Hunki Kwon
- Massachusetts General Hospital, Department of Neurology, Boston, MA 02114, USA
| | - Mark A Kramer
- Department of Mathematics and Statistics and Center for Systems Neuroscience, Boston University, Boston, MA 02215, USA
| | - Matti S Hämäläinen
- Massachusetts General Hospital, Department of Radiology, Boston, MA 02114, USA; Athinoula A, Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Catherine J Chu
- Massachusetts General Hospital, Department of Neurology, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
31
|
Freibauer AE, RamachandranNair R, Jain P, Jones KC, Whitney R. The genetic landscape of developmental and epileptic encephalopathy with spike-and-wave activation in sleep. Seizure 2023; 110:119-125. [PMID: 37352690 DOI: 10.1016/j.seizure.2023.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023] Open
Abstract
OBJECTIVE Epileptic Encephalopathy / Developmental Epileptic Encephalopathy with spike-and-wave activation in sleep (EE/DEE-SWAS) is defined as an epilepsy syndrome characterized by neurodevelopmental regression temporally related to the emergence of significant activation of spike-wave discharges in EEG during sleep. The availability of genetic testing has made it evident that monogenic and chromosomal abnormalities play an aetiological role in the development of EE/DEE-SWAS. We sought to review the literature to better understand the genetic landscape of EE/DEE-SWAS. METHODS In this systematic review, we reviewed cases of EE/DEE-SWAS associated with a genetic aetiology, collecting information related to the underlying aetiology, onset, management, and EEG patterns. RESULTS One hundred and seventy-two cases of EE/DEE-SWAS were identified. Genetic causes of note included pathogenic variants in GRIN2A, ZEB2, CNKSR2 and chromosome 17q21.31 deletions, each of which demonstrated unique clinical characteristics, EEG patterns, and age of onset. Factors identified to raise suspicion of a potential genetic aetiology included the presentation of DEE-SWAS and onset of SWAS under the age of five years. Treatment of EE/DEE-SWAS due to genetic causes was diverse, including a combination of anti-seizure medications, steroids, and other clinical strategies, with no clear consensus on a preferred or superior treatment. Data collected was significantly heterogeneous, with a lack of consistent use of neuropsychology testing, EEG patterns, or use of established clinical definitions. CONCLUSIONS Uniformity concerning the new definition of EE/DEE-SWAS, guidelines for management and more frequent genetic screening will be needed to guide best practices for the treatment of patients with EE/DEE-SWAS.
Collapse
Affiliation(s)
- Alexander E Freibauer
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON, Canada
| | | | - Puneet Jain
- Epilepsy Program, Division of Neurology, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Kevin C Jones
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON, Canada
| | - Robyn Whitney
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
32
|
Halász P, Szũcs A. Self-limited childhood epilepsies are disorders of the perisylvian communication system, carrying the risk of progress to epileptic encephalopathies-Critical review. Front Neurol 2023; 14:1092244. [PMID: 37388546 PMCID: PMC10301767 DOI: 10.3389/fneur.2023.1092244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/04/2023] [Indexed: 07/01/2023] Open
Abstract
"Sleep plasticity is a double-edged sword: a powerful machinery of neural build-up, with a risk to epileptic derailment." We aimed to review the types of self-limited focal epilepsies..."i.e. keep as two separate paragraphs" We aimed to review the types of self-limited focal epilepsies: (1) self-limited focal childhood epilepsy with centrotemporal spikes, (2) atypical Rolandic epilepsy, and (3) electrical status epilepticus in sleep with mental consequences, including Landau-Kleffner-type acquired aphasia, showing their spectral relationship and discussing the debated topics. Our endeavor is to support the system epilepsy concept in this group of epilepsies, using them as models for epileptogenesis in general. The spectral continuity of the involved conditions is evidenced by several features: language impairment, the overarching presence of centrotemporal spikes and ripples (with changing electromorphology across the spectrum), the essential timely and spatial independence of interictal epileptic discharges from seizures, NREM sleep relatedness, and the existence of the intermediate-severity "atypical" forms. These epilepsies might be the consequences of a genetically determined transitory developmental failure, reflected by widespread neuropsychological symptoms originating from the perisylvian network that have distinct time and space relations from secondary epilepsy itself. The involved epilepsies carry the risk of progression to severe, potentially irreversible encephalopathic forms.
Collapse
Affiliation(s)
- Péter Halász
- Department of Neurology, University Medical School, Pécs, Hungary
| | - Anna Szũcs
- Institute of Behavioral Sciences, Semmelweis University, Budapest, Hungary
| |
Collapse
|
33
|
Donnan AM, Schneider AL, Russ-Hall S, Churilov L, Scheffer IE. Rates of Status Epilepticus and Sudden Unexplained Death in Epilepsy in People With Genetic Developmental and Epileptic Encephalopathies. Neurology 2023; 100:e1712-e1722. [PMID: 36750385 PMCID: PMC10115508 DOI: 10.1212/wnl.0000000000207080] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 01/05/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The genetic developmental and epileptic encephalopathies (DEEs) comprise a large group of severe epilepsy syndromes, with a wide phenotypic spectrum. Currently, the rates of convulsive status epilepticus (CSE), nonconvulsive status epilepticus (NCSE), and sudden unexplained death in epilepsy (SUDEP) in these diseases are not well understood. We aimed to describe the proportions of patients with frequently observed genetic DEEs who developed CSE, NCSE, mortality, and SUDEP. Understanding the risks of these serious presentations in each genetic DEE will enable earlier diagnosis and appropriate management. METHODS In this retrospective analysis of patients with a genetic DEE, we estimated the proportions with CSE, NCSE, and SUDEP and the overall and SUDEP-specific mortality rates for each genetic diagnosis. We included patients with a pathogenic variant in the genes SCN1A, SCN2A, SCN8A, SYNGAP1, NEXMIF, CHD2, PCDH19, STXBP1, GRIN2A, KCNT1, and KCNQ2 and with Angelman syndrome (AS). RESULTS The cohort comprised 510 individuals with a genetic DEE, in whom we observed CSE in 47% and NCSE in 19%. The highest proportion of CSE occurred in patients with SCN1A-associated DEEs, including 181/203 (89%; 95% CI 84-93) patients with Dravet syndrome and 8/15 (53%; 95% CI 27-79) non-Dravet SCN1A-DEEs. CSE was also notable in patients with pathogenic variants in KCNT1 (6/10; 60%; 95% CI 26-88) and SCN2A (8/15; 53%; 95% CI 27-79). NCSE was common in patients with non-Dravet SCN1A-DEEs (8/15; 53%; 95% CI 27-79) and was notable in patients with CHD2-DEEs (6/14; 43%; 95% CI 18-71) and AS (6/19; 32%; 95% CI 13-57). There were 42/510 (8%) deaths among the cohort, producing a mortality rate of 6.1 per 1,000 person-years (95% CI 4.4-8.3). Cases of SUDEP accounted for 19/42 (48%) deaths. Four genes were associated with SUDEP: SCN1A, SCN2A, SCN8A, and STXBP1. The estimated SUDEP rate was 2.8 per 1,000 person-years (95% CI 1.6-4.3). DISCUSSION We showed that proportions of patients with CSE, NCSE, and SUDEP differ for commonly encountered genetic DEEs. The estimates for each genetic DEE studied will inform early diagnosis and management of status epilepticus and SUDEP and inform disease-specific counseling for patients and families in this high-risk group of conditions.
Collapse
Affiliation(s)
- Alice M Donnan
- From the Epilepsy Research Centre (A.M.D., A.L.S., S.R.-H., I.E.S.), Department of Medicine, The University of Melbourne, Austin Health; Melbourne Medical School (L.C.), Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville; The Florey Institute of Neurosciences and Mental Health (L.C., I.E.S.), Melbourne; and Department of Paediatrics (I.E.S.), The University of Melbourne, Royal Children's Hospital, and Murdoch Children's Research Institute, Victoria, Australia
| | - Amy L Schneider
- From the Epilepsy Research Centre (A.M.D., A.L.S., S.R.-H., I.E.S.), Department of Medicine, The University of Melbourne, Austin Health; Melbourne Medical School (L.C.), Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville; The Florey Institute of Neurosciences and Mental Health (L.C., I.E.S.), Melbourne; and Department of Paediatrics (I.E.S.), The University of Melbourne, Royal Children's Hospital, and Murdoch Children's Research Institute, Victoria, Australia
| | - Sophie Russ-Hall
- From the Epilepsy Research Centre (A.M.D., A.L.S., S.R.-H., I.E.S.), Department of Medicine, The University of Melbourne, Austin Health; Melbourne Medical School (L.C.), Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville; The Florey Institute of Neurosciences and Mental Health (L.C., I.E.S.), Melbourne; and Department of Paediatrics (I.E.S.), The University of Melbourne, Royal Children's Hospital, and Murdoch Children's Research Institute, Victoria, Australia
| | - Leonid Churilov
- From the Epilepsy Research Centre (A.M.D., A.L.S., S.R.-H., I.E.S.), Department of Medicine, The University of Melbourne, Austin Health; Melbourne Medical School (L.C.), Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville; The Florey Institute of Neurosciences and Mental Health (L.C., I.E.S.), Melbourne; and Department of Paediatrics (I.E.S.), The University of Melbourne, Royal Children's Hospital, and Murdoch Children's Research Institute, Victoria, Australia
| | - Ingrid E Scheffer
- From the Epilepsy Research Centre (A.M.D., A.L.S., S.R.-H., I.E.S.), Department of Medicine, The University of Melbourne, Austin Health; Melbourne Medical School (L.C.), Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville; The Florey Institute of Neurosciences and Mental Health (L.C., I.E.S.), Melbourne; and Department of Paediatrics (I.E.S.), The University of Melbourne, Royal Children's Hospital, and Murdoch Children's Research Institute, Victoria, Australia.
| |
Collapse
|
34
|
Pruunsild P, Bengtson CP, Loss I, Lohrer B, Bading H. Expression of the primate-specific LINC00473 RNA in mouse neurons promotes excitability and CREB-regulated transcription. J Biol Chem 2023; 299:104671. [PMID: 37019214 DOI: 10.1016/j.jbc.2023.104671] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
The LINC00473 (Lnc473) gene has previously been shown to be associated with cancer and psychiatric disorders. Its expression is elevated in several types of tumors and decreased in the brains of patients diagnosed with schizophrenia or major depression. In neurons, Lnc473 transcription is strongly responsive to synaptic activity, suggesting a role in adaptive, plasticity-related mechanisms. However, the function of Lnc473 is largely unknown. Here, using a recombinant adeno-associated viral vector, we introduced a primate-specific human Lnc473 RNA into mouse primary neurons. We show that this resulted in a transcriptomic shift comprising downregulation of epilepsy-associated genes and a rise in cAMP response element binding protein (CREB) activity, which was driven by augmented CREB-regulated transcription coactivator 1 (CRTC1) nuclear localization. Moreover, we demonstrate that ectopic Lnc473 expression increased neuronal excitability as well as network excitability. These findings suggest that primates may possess a lineage-specific activity-dependent modulator of CREB-regulated neuronal excitability.
Collapse
|
35
|
Herzog LE, Wang L, Yu E, Choi S, Farsi Z, Song BJ, Pan JQ, Sheng M. Mouse mutants in schizophrenia risk genes GRIN2A and AKAP11 show EEG abnormalities in common with schizophrenia patients. Transl Psychiatry 2023; 13:92. [PMID: 36914641 PMCID: PMC10011509 DOI: 10.1038/s41398-023-02393-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023] Open
Abstract
Schizophrenia is a heterogeneous psychiatric disorder with a strong genetic basis, whose etiology and pathophysiology remain poorly understood. Exome sequencing studies have uncovered rare, loss-of-function variants that greatly increase risk of schizophrenia [1], including loss-of-function mutations in GRIN2A (aka GluN2A or NR2A, encoding the NMDA receptor subunit 2A) and AKAP11 (A-Kinase Anchoring Protein 11). AKAP11 and GRIN2A mutations are also associated with bipolar disorder [2], and epilepsy and developmental delay/intellectual disability [1, 3, 4], respectively. Accessible in both humans and rodents, electroencephalogram (EEG) recordings offer a window into brain activity and display abnormal features in schizophrenia patients. Does loss of Grin2a or Akap11 in mice also result in EEG abnormalities? We monitored EEG in heterozygous and homozygous knockout Grin2a and Akap11 mutant mice compared with their wild-type littermates, at 3- and 6-months of age, across the sleep/wake cycle and during auditory stimulation protocols. Grin2a and Akap11 mutants exhibited increased resting gamma power, attenuated auditory steady-state responses (ASSR) at gamma frequencies, and reduced responses to unexpected auditory stimuli during mismatch negativity (MMN) tests. Sleep spindle density was reduced in a gene dose-dependent manner in Akap11 mutants, whereas Grin2a mutants showed increased sleep spindle density. The EEG phenotypes of Grin2a and Akap11 mutant mice show a variety of abnormal features that overlap considerably with human schizophrenia patients, reflecting systems-level changes caused by Grin2a and Akap11 deficiency. These neurophysiologic findings further substantiate Grin2a and Akap11 mutants as genetic models of schizophrenia and identify potential biomarkers for stratification of schizophrenia patients.
Collapse
Affiliation(s)
- Linnea E Herzog
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Lei Wang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eunah Yu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Soonwook Choi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zohreh Farsi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bryan J Song
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jen Q Pan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Morgan Sheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
36
|
Pereira-Nunes J, Sousa JM, Fonseca J, Melo C, Alves D, Sampaio M, Sousa R. Successful Treatment of a Child With Epileptic Encephalopathy With Spike-Wave Activation in Sleep and GRIN2A Variant Using Sulthiame. Cureus 2023; 15:e34686. [PMID: 36909045 PMCID: PMC9996194 DOI: 10.7759/cureus.34686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 02/09/2023] Open
Abstract
Epileptic encephalopathy with spike-wave activation in sleep (EE-SWAS) and developmental EE-SWAS (DEE-SWAS) are characterized by variable combinations of cognitive, language, behavioral, and/or motor regression associated with continuous or near-continuous diffuse spike-and-wave complexes during sleep. Glutamate ionotropic receptor NMDA type subunit 2A (GRIN2A) variants have been associated with EE-SWAS. It encodes the most relevant GluN2 subunit of the N-methyl-D-aspartate receptor (NMDAR). Sulthiame reduces NMDAR-mediated neuronal excitability and has been progressively used as monotherapy in self-limited epilepsy with centrotemporal spikes (SeLECTS) or as add-ontherapy in EE-SWAS/DEE-SWAS. A five-year-old female, with family history of epilepsy, was initially diagnosed with SeLECTS and medicated with valproic acid (VPA). One year later, she presented a focal to bilateral tonic-clonic seizure during sleep and learning difficulty. The electroencephalogram revealed continuous spike-and-wave during sleep leading to the diagnosis of EE-SWAS. Prednisolone was effective, but there was repeated recurrence after its discontinuation and associated adverse effects. As an alternative, sulthiame was added to VPA. Four years later, she remains clinically stable. Genetic testing revealed a GRIN2A missense variant, C.3228C>A (p.Asn1076Lys). Sulthiame appeared effective in this recurrent EE-SWAS child, who presented a GRIN2A missense variant with possible NMDAR gain-of-function and adverse effects of corticosteroids. Functional studies of GRIN2A variants might become a future tool for individualized therapies.
Collapse
Affiliation(s)
- Joana Pereira-Nunes
- Department of Pediatrics, Centro Hospitalar Universitário de São João, Porto, PRT.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, PRT
| | - José Maria Sousa
- Department of Neuroradiology, Centro Hospitalar Universitário de São João, Porto, PRT
| | - Jacinta Fonseca
- Pediatric Neurology Unit, Department of Pediatrics, Centro Hospitalar Universitário de São João, Porto, PRT
| | - Cláudia Melo
- Pediatric Neurology Unit, Department of Pediatrics, Centro Hospitalar Universitário de São João, Porto, PRT.,Department of Gynecology-Obstetrics and Pediatrics, Faculty of Medicine of Porto University, Porto, PRT
| | - Dílio Alves
- Department of Neurophysiology, Centro Hospitalar Universitário de São João, Porto, PRT
| | - Mafalda Sampaio
- Pediatric Neurology Unit, Department of Pediatrics, Centro Hospitalar Universitário de São João, Porto, PRT
| | - Raquel Sousa
- Pediatric Neurology Unit, Department of Pediatrics, Centro Hospitalar Universitário de São João, Porto, PRT
| |
Collapse
|
37
|
GRIN2A-related epilepsy and speech disorders: A comprehensive overview with a focus on the role of precision therapeutics. Epilepsy Res 2023; 189:107065. [PMID: 36516565 DOI: 10.1016/j.eplepsyres.2022.107065] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/27/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Language dysfunction is a common and serious comorbidity of epilepsy, especially in individuals with epilepsy aphasia spectrum syndromes. Childhood epilepsy with centrotemporal spikes is on the mild end of the spectrum, while epileptic encephalopathy with continuous spike-and-wave during sleep syndrome is on the severe end. Traditional antiseizure medicines and immunotherapy are currently used to treat severely affected patients, but the results are usually disappointing. The discovery that GRIN2A is the primary monogenic etiology of these diseases has opened the door to precision treatments. The GRIN2A gene encodes GluN2A protein, which constitutes a subunit of the NMDA receptor (NMDAR). The GRIN2A pathogenic variants cause gain or loss of function of NMDAR; the former can be treated with uncompetitive NMDAR antagonists, such as memantine, while the latter with NMDAR co-agonist serine. Hyper-precision therapies with various other effective agents are likely to be developed shortly to target the diverse functional effects of different variants. Precision treatments for GRIN2A-related disorders will benefit those who suffer from the condition and pave the way for new therapeutic approaches to a variety of other NMDAR-linked neurodegenerative and psychiatric diseases (schizophrenia, Parkinson's disease, Alzheimer's disease, and so on). Furthermore, more research into GRIN2A-related disorders will help us better understand the neuroinflammatory and neuroimmunological basis of epilepsy, as well as the pathological and physiological network activation mechanisms that cause sleep activation of central-temporal spikes and language impairment.
Collapse
|
38
|
Benítez-Burraco A, Jiménez-Romero MS, Fernández-Urquiza M. Delving into the Genetic Causes of Language Impairment in a Case of Partial Deletion of NRXN1. Mol Syndromol 2023; 13:496-510. [PMID: 36660026 PMCID: PMC9843585 DOI: 10.1159/000524710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/22/2022] [Indexed: 01/22/2023] Open
Abstract
Introduction Copy-number variations (CNVs) impacting on small DNA stretches and associated with language deficits provide a unique window to the role played by specific genes in language function. Methods We report in detail on the cognitive, language, and genetic features of a girl bearing a small deletion (0.186 Mb) in the 2p16.3 region, arr[hg19] 2p16.3(50761778_50947729)×1, affecting exons 3-7 of NRXN1, a neurexin-coding gene previously related to schizophrenia, autism (ASD), attention deficit hyperactivity disorder (ADHD), mood disorder, and intellectual disability (ID). Results The proband exhibits many of the features commonly found in subjects with deletions of NRXN1, like ASD-like traits (including ritualized behaviors, disordered sensory aspects, social disturbances, and impaired theory of mind), ADHD symptoms, moderate ID, and impaired speech and language. Regarding this latter aspect, we observed altered speech production, underdeveloped phonological awareness, minimal syntax, serious shortage of active vocabulary, impaired receptive language, and inappropriate pragmatic behavior (including lack of metapragmatic awareness and communicative use of gaze). Microarray analyses point to the dysregulation of several genes important for language function in the girl compared to her healthy parents. Discussion Although some basic cognitive deficit - such as the impairment of executive function - might contribute to the language problems exhibited by the proband, molecular evidence suggests that they might result, to a great extent, from the abnormal expression of genes directly related to language.
Collapse
Affiliation(s)
- Antonio Benítez-Burraco
- Department of Spanish, Linguistics, and Theory of Literature (Linguistics), University of Seville, Seville, Spain,*Antonio Benítez-Burraco,
| | | | | |
Collapse
|
39
|
Iacobucci GJ, Liu B, Wen H, Sincox B, Zheng W, Popescu GK. Complex functional phenotypes of NMDA receptor disease variants. Mol Psychiatry 2022; 27:5113-5123. [PMID: 36117210 DOI: 10.1038/s41380-022-01774-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/23/2022] [Accepted: 08/31/2022] [Indexed: 01/14/2023]
Abstract
NMDA receptors have essential roles in the physiology of central excitatory synapses and their dysfunction causes severe neuropsychiatric symptoms. Recently, a series of genetic variants have been identified in patients, however, functional information about these variants is sparse and their role in pathogenesis insufficiently known. Here we investigate the mechanism by which two GluN2A variants may be pathogenic. We use molecular dynamics simulation and single-molecule electrophysiology to examine the contribution of GluN2A subunit-residues, P552 and F652, and their pathogenic substitutions, P552R and F652V, affect receptor functions. We found that P552 and F652 interact during the receptors' normal activity cycle; the interaction stabilizes receptors in open conformations and is required for a normal electrical response. Engineering shorter side-chains at these positions (P552A and/or F652V) caused a loss of interaction energy and produced receptors with severe gating, conductance, and permeability deficits. In contrast, the P552R side chain resulted in stronger interaction and produced a distinct, yet still drastically abnormal electrical response. These results identify the dynamic contact between P552 and F652 as a critical step in the NMDA receptor activation, and show that both increased and reduced communication through this interaction cause dysfunction. Results show that subtle differences in NMDA receptor primary structure can generate complex phenotypic alterations whose binary classification is too simplistic to serve as a therapeutic guide.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| | - Beiying Liu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Han Wen
- Department of Physics, College of Arts and Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Brittany Sincox
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Wenjun Zheng
- Department of Physics, College of Arts and Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| | - Gabriela K Popescu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
40
|
Feng Y, Zhang C, Wei Z, Li G, Gan Y, Liu C, Deng Y. Gene variations of glutamate metabolism pathway and epilepsy. ACTA EPILEPTOLOGICA 2022. [DOI: 10.1186/s42494-022-00103-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Epilepsy is a paroxysmal disorder of the brain, caused by an imbalance of neuronal excitation and inhibition. Glutamate is the most important excitatory neurotransmitter in the brain and plays an important role in epileptogenesis. Mutations in genes at any step/component of the glutamate metabolic pathway may lead to the development of epilepsy or epileptic encephalopathy.
Methods
Clinical history of 3 epilepsy patients with genetic variations of the glutamate metabolism pathway was collected. Electroencephalogram recording and magnetic resonance imaging were performed in each patient. We also reviewed recent literature for a variety of the genetic variations involved in epilepsy.
Results
Case 1 was a SLC1A2 mutation-carrier diagnosed with developmental and epileptic encephalopathy (DEE) 41, whose seizures decreased after start of the ketogenic diet. Case 2 carried a GRIN2A gene mutation and was seizure-free for three years after taking levetiracetam and vitamin B6. Case 3 was a GRIN2B mutation-carrier diagnosed with DEE 27, who seizures diminished after taking oxcarbazepine.
Conclusions
Preclinical and clinical evidence supports the therapeutic potential of glutamatergic signaling-targeting treatments for epilepsy. More studies are needed to discover novel DEE-related genetic mutations in the glutamate metabolic pathway.
Collapse
|
41
|
Guo A, Lun P, Chen J, Li Q, Chang K, Li T, Pan D, Zhang J, Zhou J, Wang K, Zhang Q, Yang Q, Gao C, Wu C, Jian X, Wen Y, Wang Z, Shi Y, Zhao X, Sun P, Li Z. Association analysis of risk genes identified by SCHEMA with schizophrenia in the Chinese Han population. Psychiatr Genet 2022; 32:188-193. [PMID: 36125369 DOI: 10.1097/ypg.0000000000000321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Schizophrenia is a chronic brain disorder. Previously, the Schizophrenia Exome Sequencing Meta-analysis consortium identified 10 highest risk genes related to schizophrenia. This study aimed to analyze the relationship between the 10 highest risk genes identified by the SCHEMA and schizophrenia in a Chinese population. METHODS A total of 225 variants in 10 genes were screened in a Chinese population of 6836 using a customized array. All variants were annotated through the Variant Effect Predictor tool, and the functional impacts of missense variants were assessed based on sorting intolerant from tolerant and PolyPhen-2 scores. The SHEsisPlus tool was used to analyze the association between risk genes and schizophrenia at the locus and gene levels. RESULTS At the locus level, no missense variants significantly related to schizophrenia were found, but we detected three missense variants that appeared only in cases, including TRIO p. Arg1185Gln, RB1CC1 p. Arg1514Cys, and HERC1 p. Val4517Leu. At the gene level, five genes (TRIO, RB1CC1, HERC1, GRIN2A, and CACAN1G) with more than one variant analyzed were kept for the gene-level association analysis. Only the association between RB1CC1 and schizophrenia reached a significant level (OR = 1.634; 95% CI, 1.062-2.516; P = 0.025). CONCLUSION In this study, we determined that RB1CC1 might be a risk gene for schizophrenia in the Chinese population. Our results provide new evidence for recognizing the correlation of these risk genes with the Chinese schizophrenia population.
Collapse
Affiliation(s)
- Aiguo Guo
- School of Basic Medicine, Qingdao University
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University
| | - Peng Lun
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai
| | - Qinghua Li
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao
| | - Kaihui Chang
- School of Basic Medicine, Qingdao University
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University
| | - Teng Li
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University
- School of Public Health, Qingdao University, Qingdao
| | - Dun Pan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai
| | - Jinmai Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai
| | - Juan Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai
| | - Ke Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai
| | - Qian Zhang
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University
| | - Qiangzhen Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai
| | - Chengwen Gao
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University
| | - Chuanhong Wu
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University
| | - Xuemin Jian
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University
| | - Yanqin Wen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai
| | - Zhuo Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai
| | - Yongyong Shi
- School of Basic Medicine, Qingdao University
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai
- Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangzhong Zhao
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University
| | - Peng Sun
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao
| | - Zhiqiang Li
- School of Basic Medicine, Qingdao University
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai
- School of Public Health, Qingdao University, Qingdao
- Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
42
|
Al Qahtani X, Multhaupt-Buell T, Sharma N, Dy-Hollins ME. Myoclonus-Dystonia in an Individual with a Mutation in the GRIN2A Gene. JOURNAL OF PEDIATRIC NEUROLOGY 2022. [DOI: 10.1055/s-0042-1756445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
AbstractMutations in the GRIN2A gene are associated with epilepsy-aphasia spectrum disorders and developmental and epileptic encephalopathies. Associations have been linked with disorders, including autism spectrum disorder and Parkinson's disease. Recently, GRIN2A variants have been reported as a cause of movement disorders in individuals without epilepsy, suggesting that movement disorders should be highlighted as a genetic phenotype associated with pathogenic variants in GRIN2A. We present a case of a male with myoclonus dystonia and without epilepsy found on whole-exome sequencing to have a c.1880G > A; p.S627N variant in the GRIN2A gene. Our case contributes to the expanding phenotypic spectrum of GRIN2A-related disorders and highlights another genetic cause of the myoclonus-dystonia phenotype. GRIN2A should be considered a part of the differential diagnosis of myoclonus-dystonia in individuals with developmental delay without epilepsy.
Collapse
Affiliation(s)
- Xena Al Qahtani
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Trisha Multhaupt-Buell
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Nutan Sharma
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Marisela E. Dy-Hollins
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
43
|
Li QQ, Chen J, Hu P, Jia M, Sun JH, Feng HY, Qiao FC, Zang YY, Shi YY, Chen G, Sheng N, Xu Y, Yang JJ, Xu Z, Shi YS. Enhancing GluN2A-type NMDA receptors impairs long-term synaptic plasticity and learning and memory. Mol Psychiatry 2022; 27:3468-3478. [PMID: 35484243 DOI: 10.1038/s41380-022-01579-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022]
Abstract
N-methyl-D-aspartic acid type glutamate receptors (NMDARs) play critical roles in synaptic transmission and plasticity, the dysregulation of which leads to cognitive defects. Here, we identified a rare variant in the NMDAR subunit GluN2A (K879R) in a patient with intellectual disability. The K879R mutation enhanced receptor expression on the cell surface by disrupting a KKK motif that we demonstrated to be an endoplasmic reticulum retention signal. Expression of GluN2A_K879R in mouse hippocampal CA1 neurons enhanced the excitatory postsynaptic currents mediated by GluN2A-NMDAR but suppressed those mediated by GluN2B-NMDAR and the AMPA receptor. GluN2A_K879R knock-in mice showed similar defects in synaptic transmission and exhibited impaired learning and memory. Furthermore, both LTP and LTD were severely impaired in the KI mice, likely explaining their learning and memory defects. Therefore, our study reveals a new mechanism by which elevated synaptic GluN2A-NMDAR impairs long-term synaptic plasticity as well as learning and memory.
Collapse
Affiliation(s)
- Qing-Qing Li
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Neurology, Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210032, China
| | - Jiang Chen
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Neurology, Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210032, China
| | - Ping Hu
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Min Jia
- Department of Anesthesiology, Pain and Perioperative Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jia-Hui Sun
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Neurology, Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210032, China.,State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210032, China
| | - Hao-Yang Feng
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Feng-Chang Qiao
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Yan-Yu Zang
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Neurology, Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210032, China.,State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210032, China
| | - Yong-Yun Shi
- Department of Orthopaedics, Luhe People's Hospital Affiliated to Yangzhou University, Nanjing, 211500, China
| | - Guiquan Chen
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Neurology, Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210032, China.,State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210032, China
| | - Nengyin Sheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Yun Xu
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Neurology, Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210032, China.,State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210032, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Zhengfeng Xu
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China.
| | - Yun Stone Shi
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Neurology, Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210032, China. .,State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210032, China. .,Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210032, China. .,Guangdong Institute of Intelligence Science and Technology, Zhuhai, 519031, China.
| |
Collapse
|
44
|
Benske TM, Mu TW, Wang YJ. Protein quality control of N-methyl-D-aspartate receptors. Front Cell Neurosci 2022; 16:907560. [PMID: 35936491 PMCID: PMC9352929 DOI: 10.3389/fncel.2022.907560] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/29/2022] [Indexed: 12/23/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are glutamate-gated cation channels that mediate excitatory neurotransmission and are critical for synaptic development and plasticity in the mammalian central nervous system (CNS). Functional NMDARs typically form via the heterotetrameric assembly of GluN1 and GluN2 subunits. Variants within GRIN genes are implicated in various neurodevelopmental and neuropsychiatric disorders. Due to the significance of NMDAR subunit composition for regional and developmental signaling at synapses, properly folded receptors must reach the plasma membrane for their function. This review focuses on the protein quality control of NMDARs. Specifically, we review the quality control mechanisms that ensure receptors are correctly folded and assembled within the endoplasmic reticulum (ER) and trafficked to the plasma membrane. Further, we discuss disease-associated variants that have shown disrupted NMDAR surface expression and function. Finally, we discuss potential targeted pharmacological and therapeutic approaches to ameliorate disease phenotypes by enhancing the expression and surface trafficking of subunits harboring disease-associated variants, thereby increasing their incorporation into functional receptors.
Collapse
Affiliation(s)
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Ya-Juan Wang
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
45
|
Liu R, Rizzo S, Waliany S, Garmhausen MR, Pal N, Huang Z, Chaudhary N, Wang L, Harbron C, Neal J, Copping R, Zou J. Systematic pan-cancer analysis of mutation-treatment interactions using large real-world clinicogenomics data. Nat Med 2022; 28:1656-1661. [PMID: 35773542 DOI: 10.1038/s41591-022-01873-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/16/2022] [Indexed: 11/10/2022]
Abstract
Quantifying the effectiveness of different cancer therapies in patients with specific tumor mutations is critical for improving patient outcomes and advancing precision medicine. Here we perform a large-scale computational analysis of 40,903 US patients with cancer who have detailed mutation profiles, treatment sequences and outcomes derived from electronic health records. We systematically identify 458 mutations that predict the survival of patients on specific immunotherapies, chemotherapy agents or targeted therapies across eight common cancer types. We further characterize mutation-mutation interactions that impact the outcomes of targeted therapies. This work demonstrates how computational analysis of large real-world data generates insights, hypotheses and resources to enable precision oncology.
Collapse
Affiliation(s)
- Ruishan Liu
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA.,Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | | | - Sarah Waliany
- School of Medicine, Stanford University, Stanford, CA, USA
| | | | | | - Zhi Huang
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | | | - Lisa Wang
- Genentech, South San Francisco, CA, USA
| | | | - Joel Neal
- School of Medicine, Stanford University, Stanford, CA, USA
| | | | - James Zou
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA. .,Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
| |
Collapse
|
46
|
Lin JJ, Meletti S, Vaudano AE, Lin KL. Developmental and epileptic encephalopathies: Is prognosis related to different epileptic network dysfunctions? Epilepsy Behav 2022; 131:107654. [PMID: 33349540 DOI: 10.1016/j.yebeh.2020.107654] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 11/19/2022]
Abstract
Developmental and epileptic encephalopathies are a group of rare, severe epilepsies, which are characterized by refractory seizures starting in infancy or childhood and developmental delay or regression. Developmental changes might be independent of epilepsy. However, interictal epileptic activity and seizures can further deteriorate cognition and behavior. Recently, the concept of developmental and epileptic encephalopathies has moved from the lesions associated with epileptic encephalopathies toward the epileptic network dysfunctions on the functioning of the brain. Early recognition and differentiation of patients with developmental and epileptic encephalopathies is important, as precision therapies need to be holistic to address the often devastating symptoms. In this review, we discuss the evolution of the concept of developmental and epileptic encephalopathies in recent years, as well as the current understanding of the genetic basis of developmental and epileptic encephalopathies. Finally, we will discuss the role of epileptic network dysfunctions on prognosis for these severe conditions.
Collapse
Affiliation(s)
- Jainn-Jim Lin
- Division of Pediatric Critical Care and Pediatric Neurocritical Care Center, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University, College of Medicine, Taoyuan, Taiwan; Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Department of Respiratory Therapy, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Study Group for Intensive and Integrated Care of Pediatric Central Nervous System (iCNS Group), Chang Gung Children's Hospital, Taoyuan, Taiwan
| | - Stefano Meletti
- Division of Neurology, University Hospital of Modena, Modena, Italy; Department of Biomedical, Metabolic and Neural Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Elisabetta Vaudano
- Division of Neurology, University Hospital of Modena, Modena, Italy; Department of Biomedical, Metabolic and Neural Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Kuang-Lin Lin
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Study Group for Intensive and Integrated Care of Pediatric Central Nervous System (iCNS Group), Chang Gung Children's Hospital, Taoyuan, Taiwan.
| |
Collapse
|
47
|
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
|
48
|
Bavan S, Goodkin HP, Papazian DM. Altered Closed State Inactivation Gating in Kv4.2 Channels Results in Developmental and Epileptic Encephalopathies in Human Patients. Hum Mutat 2022; 43:1286-1298. [PMID: 35510384 DOI: 10.1002/humu.24396] [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: 09/21/2021] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 11/06/2022]
Abstract
Kv4.2 subunits, encoded by KCND2, serve as the pore-forming components of voltage-gated, inactivating ISA K+ channels expressed in the brain. ISA channels inactivate without opening in response to subthreshold excitatory input, temporarily increasing neuronal excitability, the back propagation of action potentials, and Ca2+ influx into dendrites, thereby regulating mechanisms of spike timing-dependent synaptic plasticity. As previously described, a de novo variant in Kv4.2, p.Val404Met, is associated with an infant-onset developmental and epileptic encephalopathy (DEE) in monozygotic twin boys. The p.Val404Met variant enhances inactivation directly from closed states, but dramatically impairs inactivation after channel opening. We now report the identification of a closely related, novel, de novo variant in Kv4.2, p.Val402Leu, in a boy with an early-onset pharmacoresistant epilepsy that evolved to an epileptic aphasia syndrome (Continuous Spike Wave during Sleep Syndrome). Like p.Val404Met, the p.Val402Leu variant increases the rate of inactivation from closed states, but significantly slows inactivation after the pore opens. Although quantitatively the p.Val402Leu mutation alters channel kinetics less dramatically than p.Val404Met, our results strongly support the conclusion that p.Val402Leu and p.Val404Met cause the clinical features seen in the affected individuals and underscore the importance of closed state inactivation in ISA channels in normal brain development and function. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Selvan Bavan
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095-1571.,Labcorp Drug Development, Huntingdon, PE28 4HS, UK
| | - Howard P Goodkin
- Departments of Neurology and Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, 22903
| | - Diane M Papazian
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095-1571
| |
Collapse
|
49
|
Specchio N, Wirrell EC, Scheffer IE, Nabbout R, Riney K, Samia P, Guerreiro M, Gwer S, Zuberi SM, Wilmshurst JM, Yozawitz E, Pressler R, Hirsch E, Wiebe S, Cross HJ, Perucca E, Moshé SL, Tinuper P, Auvin S. International League Against Epilepsy classification and definition of epilepsy syndromes with onset in childhood: Position paper by the ILAE Task Force on Nosology and Definitions. Epilepsia 2022; 63:1398-1442. [PMID: 35503717 DOI: 10.1111/epi.17241] [Citation(s) in RCA: 320] [Impact Index Per Article: 106.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/30/2022]
Abstract
The 2017 International League Against Epilepsy classification has defined a three-tier system with epilepsy syndrome identification at the third level. Although a syndrome cannot be determined in all children with epilepsy, identification of a specific syndrome provides guidance on management and prognosis. In this paper, we describe the childhood onset epilepsy syndromes, most of which have both mandatory seizure type(s) and interictal electroencephalographic (EEG) features. Based on the 2017 Classification of Seizures and Epilepsies, some syndrome names have been updated using terms directly describing the seizure semiology. Epilepsy syndromes beginning in childhood have been divided into three categories: (1) self-limited focal epilepsies, comprising four syndromes: self-limited epilepsy with centrotemporal spikes, self-limited epilepsy with autonomic seizures, childhood occipital visual epilepsy, and photosensitive occipital lobe epilepsy; (2) generalized epilepsies, comprising three syndromes: childhood absence epilepsy, epilepsy with myoclonic absence, and epilepsy with eyelid myoclonia; and (3) developmental and/or epileptic encephalopathies, comprising five syndromes: epilepsy with myoclonic-atonic seizures, Lennox-Gastaut syndrome, developmental and/or epileptic encephalopathy with spike-and-wave activation in sleep, hemiconvulsion-hemiplegia-epilepsy syndrome, and febrile infection-related epilepsy syndrome. We define each, highlighting the mandatory seizure(s), EEG features, phenotypic variations, and findings from key investigations.
Collapse
Affiliation(s)
- Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, Scientific Institute for Research and Health Care, Full Member of EpiCARE, Rome, Italy
| | - Elaine C Wirrell
- Divisions of Child and Adolescent Neurology and Epilepsy, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ingrid E Scheffer
- Austin Health and Royal Children's Hospital, Florey Institute, Murdoch Children's Research Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Rima Nabbout
- Reference Center for Rare Epilepsies, Department of Pediatric Neurology, Necker-Sick Children Hospital, Public Hospital Network of Paris, member of EpiCARE, Imagine Institute, National Institute of Health and Medical Research, Mixed Unit of Research 1163, University of Paris, Paris, France
| | - Kate Riney
- Neurosciences Unit, Queensland Children's Hospital, South Brisbane, Queensland, Australia.,Faculty of Medicine, University of Queensland, South Brisbane, Queensland, Australia
| | - Pauline Samia
- Department of Pediatrics and Child Health, Aga Khan University, Nairobi, Kenya
| | | | - Sam Gwer
- School of Medicine, Kenyatta University, and Afya Research Africa, Nairobi, Kenya
| | - Sameer M Zuberi
- Paediatric Neurosciences Research Group, Royal Hospital for Children and Institute of Health & Wellbeing, member of EpiCARE, University of Glasgow, Glasgow, UK
| | - Jo M Wilmshurst
- Department of Paediatric Neurology, Red Cross War Memorial Children's Hospital, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Elissa Yozawitz
- Isabelle Rapin Division of Child Neurology of the Saul R. Korey Department of Neurology, Montefiore Medical Center, Bronx, New York, USA
| | - Ronit Pressler
- Programme of Developmental Neurosciences, University College London National Institute for Health Research Biomedical Research Centre Great Ormond Street Institute of Child Health, Department of Clinical Neurophysiology, Great Ormond Street Hospital for Children, London, UK
| | - Edouard Hirsch
- Neurology Epilepsy Units "Francis Rohmer", INSERM 1258, FMTS, Strasbourg University, Strasbourg, France
| | - Sam Wiebe
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Helen J Cross
- Programme of Developmental Neurosciences, University College London National Institute for Health Research Biomedical Research Centre Great Ormond Street Institute of Child Health, Great Ormond Street Hospital for Children, and Young Epilepsy Lingfield, London, UK
| | - Emilio Perucca
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Solomon L Moshé
- Isabelle Rapin Division of Child Neurology, Saul R. Korey Department of Neurology, and Departments of Neuroscience and Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, USA
| | - Paolo Tinuper
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Institute of Neurological Sciences, Scientific Institute for Research and Health Care, Bologna, Italy
| | - Stéphane Auvin
- Robert Debré Hospital, Public Hospital Network of Paris, NeuroDiderot, National Institute of Health and Medical Research, Department Medico-Universitaire Innovation Robert-Debré, Pediatric Neurology, University of Paris, Paris, France
| |
Collapse
|
50
|
Mild neurological phenotype in a family carrying a novel N-terminal null GRIN2A variant. Eur J Med Genet 2022; 65:104500. [DOI: 10.1016/j.ejmg.2022.104500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/06/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022]
|