1
|
Pikor D, Hurła M, Słowikowski B, Szymanowicz O, Poszwa J, Banaszek N, Drelichowska A, Jagodziński PP, Kozubski W, Dorszewska J. Calcium Ions in the Physiology and Pathology of the Central Nervous System. Int J Mol Sci 2024; 25:13133. [PMID: 39684844 DOI: 10.3390/ijms252313133] [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: 11/09/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Calcium ions play a key role in the physiological processes of the central nervous system. The intracellular calcium signal, in nerve cells, is part of the neurotransmission mechanism. They are responsible for stabilizing membrane potential and controlling the excitability of neurons. Calcium ions are a universal second messenger that participates in depolarizing signal transduction and contributes to synaptic activity. These ions take an active part in the mechanisms related to memory and learning. As a result of depolarization of the plasma membrane or stimulation of receptors, there is an extracellular influx of calcium ions into the cytosol or mobilization of these cations inside the cell, which increases the concentration of these ions in neurons. The influx of calcium ions into neurons occurs via plasma membrane receptors and voltage-dependent ion channels. Calcium channels play a key role in the functioning of the nervous system, regulating, among others, neuronal depolarization and neurotransmitter release. Channelopathies are groups of diseases resulting from mutations in genes encoding ion channel subunits, observed including the pathophysiology of neurological diseases such as migraine. A disturbed ability of neurons to maintain an appropriate level of calcium ions is also observed in such neurodegenerative processes as Alzheimer's disease, Parkinson's disease, Huntington's disease, and epilepsy. This review focuses on the involvement of calcium ions in physiological and pathological processes of the central nervous system. We also consider the use of calcium ions as a target for pharmacotherapy in the future.
Collapse
Affiliation(s)
- Damian Pikor
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Mikołaj Hurła
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Bartosz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Oliwia Szymanowicz
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Joanna Poszwa
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Natalia Banaszek
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Alicja Drelichowska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Paweł P Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| |
Collapse
|
2
|
Lauerer RJ, Lerche H. Voltage-gated calcium channels in genetic epilepsies. J Neurochem 2024; 168:3853-3871. [PMID: 37822150 PMCID: PMC11591408 DOI: 10.1111/jnc.15983] [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: 07/08/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
Voltage-gated calcium channels (VGCC) are abundant in the central nervous system and serve a broad spectrum of functions, either directly in cellular excitability or indirectly to regulate Ca2+ homeostasis. Ca2+ ions act as one of the main connections in excitation-transcription coupling, muscle contraction and excitation-exocytosis coupling, including synaptic transmission. In recent years, many genes encoding VGCCs main α or additional auxiliary subunits have been associated with epilepsy. This review sums up the current state of knowledge on disease mechanisms and provides guidance on disease-specific therapies where applicable.
Collapse
Affiliation(s)
- Robert J. Lauerer
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain ResearchUniversity and University Hospital TuebingenTuebingenGermany
| |
Collapse
|
3
|
Fu SJ, Cheng KM, Hsiao CT, Fang YC, Jeng CJ, Tang CY. Pin1 promotes human Ca V2.1 channel polyubiquitination by RNF138: pathophysiological implication for episodic ataxia type 2. Cell Commun Signal 2024; 22:571. [PMID: 39609819 PMCID: PMC11603662 DOI: 10.1186/s12964-024-01960-9] [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: 10/10/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024] Open
Abstract
Loss-of-function mutations in the human gene encoding the neuron-specific Ca2+ channel CaV2.1 are linked to the neurological disease episodic ataxia type 2 (EA2), as well as neurodevelopmental disorders such as developmental delay and developmental epileptic encephalopathy. Disease-associated CaV2.1 mutants may exhibit defective proteostasis and promote endoplasmic reticulum (ER)-associated degradation of their wild-type (WT) counterpart in a dominant-negative manner. The E3 ubiquitin ligase RNF138 was previously shown to mediate EA2-related aberrant degradation of CaV2.1 at the ER. Herein we aimed to elucidate the ER proteostasis mechanism of CaV2.1. The peptidyl-prolyl cis/trans isomerase, NIMA-interacting 1 (Pin1) was identified as a novel neuronal CaV2.1 binding partner that promoted polyubiquitination and proteasomal degradation of CaV2.1. Suppression of endogenous Pin1 level with either shRNA knockdown or the Pin1 inhibitor all-trans retinoic acid (ATRA) enhanced endogenous CaV2.1 protein level in neurons, and attenuated ER-associated degradation of CaV2.1 WT and EA2-causing mutants. Detailed mutation analyses suggested that Pin1 interacted with specific phosphorylated serine/threonine-proline motifs in the intracellular II-III loop and the distal carboxy-terminal region of human CaV2.1. We further generated Pin1-insensitive CaV2.1 constructs and demonstrated that, during ER quality control, Pin1 served as an upstream regulator of CaV2.1 polyubiquitination and degradation by RNF138. Pin1 regulation was required for the dominant-negative effect of EA2 missense mutants, but not nonsense mutants, on CaV2.1 WT protein expression. Our data are consistent with the idea that CaV2.1 proteostasis at the ER, as well as dominant-negative suppression of disease-causing loss-of-function mutants on CaV2.1 WT, entail both Pin1/RNF138-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Ssu-Ju Fu
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Kai-Min Cheng
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Cheng-Tsung Hsiao
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
- Department of Neurology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Department of Neurology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Ya-Ching Fang
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Chung-Jiuan Jeng
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| | - Chih-Yung Tang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
| |
Collapse
|
4
|
Kim S, Kim JS, Lee SH, Kim JM, Na S, Choi JH, Kim HJ. Intellectual Disability in Episodic Ataxia Type 2: Beyond Paroxysmal Vertigo and Ataxia. J Clin Neurol 2024; 20:563-570. [PMID: 39505308 PMCID: PMC11543395 DOI: 10.3988/jcn.2024.0274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND AND PURPOSE Episodic ataxia type 2 (EA2) is characterized by recurrent vertigo and ataxia due to mutations in CACNA1A that encodes the α1A-subunit of the P/Q-type voltage-gated calcium channel. This study aimed to determine intellectual function in EA2. METHODS During 2019-2023, 13 patients (6 males, age range=10-52 years, median age=29 years) with a genetically confirmed diagnosis of EA2 had their intellectual function evaluated using the Korean versions of the Wechsler Intelligence Scales (version IV) for adults or children in 3 referral-based university hospitals in South Korea. RESULTS The full-scale intelligence quotients (FSIQs) among the 13 patients were below the average (90-109) in 11, low average (80-89) in 5 (38.5%), borderline (70-79) in 1 (7.7%), and indicated intellectual disability (≤69) in 5 (38.5%). These patterns of cognitive impairments were observed in all four of the following subtests: verbal comprehension, perceptual reasoning, working memory, and processing speed. The FSIQ was not correlated with the ages at onset for vertigo and ataxia (Pearson correlation: p=0.40). CONCLUSIONS Patients with EA2 may have hidden intellectual disabilities even without a history of epilepsy or administration of antiepileptic drugs, and should be considered for genetic counseling and therapeutic interventions. Given the availability of medication to control episodic vertigo and ataxia, early diagnosis and management are important in preventing irreversible brain dysfunction in EA2.
Collapse
Affiliation(s)
- Seoyeon Kim
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - Ji-Soo Kim
- Department of Neurology, College of Medicine, Seoul National University, Seoul, Korea
- Department of Neurology, Clinical Neuroscience Center, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seung-Han Lee
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
| | - Jae-Myung Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
| | - Seunghee Na
- Department of Neurology, Incheon St. Mary's Hospital, The Catholic University of Korea, Incheon, Korea
| | - Jae-Hwan Choi
- Department of Neurology, Pusan National University School of Medicine, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Hyo-Jung Kim
- Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, Korea.
| |
Collapse
|
5
|
Mammadova D, Kraus C, Leis T, Popp B, Zweier C, Reis A, Trollmann R. Intrafamilial neurological phenotypic variability due to either biallelic or monoallelic pathogenic variants in CACNA1A. Front Neurol 2024; 15:1458109. [PMID: 39416668 PMCID: PMC11479977 DOI: 10.3389/fneur.2024.1458109] [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: 07/01/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Pathogenic heterozygous variants in CACNA1A are associated with familial hemiplegic migraine, episodic ataxia type 2 and spinocerebellar ataxia type 6, and more recently, neurodevelopmental disorders. We describe a severe, early-onset phenotype including severe muscular hypotonia, early-onset epileptic seizures, apnoea, optic atrophy and dysphagia in three siblings carrying compound heterozygous frameshift variants in CACNA1A. Two male patients died at the age of 5 or 14 months of suspected SIDS or severe developmental epileptic encephalopathy (DEE) with refractory seizures and apnoea. A male child (index patient) developed severe early-onset DEE including seizures and ictal apnoea at the age of 4 weeks. Another male child developed generalized epilepsy and mild intellectual impairment in late infancy, and his mother and his maternal uncle were identified as carriers of a known CACNA1A pathogenic variant [c.2602delG heterozygous, p. (Ala868Profs*24)] with a diagnosis of episodic ataxia type 2. This maternal pathogenic variant c.2602delG was detected in the index patient and child 2. Trio-Exome sequencing identified an additional heterozygous pathogenic variant in the CACNA1A gene, c.5476delC, p.(His1826Thrfs*30) in the index patient and child 2, which was inherited from the asymptomatic father. In conclusion, the novel compound heterozygosity for two frameshift pathogenic variants, maternally [c.2602delG, p.(Ala868Profs*24)] and paternally [c.5476delC, p.(His1826Thrfs*3)] is associated with a severe phenotype of early-onset DEE. This observation highlights the necessity of additional analyses to clarify unusual phenotypes even if a pathogenic variant has already been identified, and expands the clinical spectrum of CACNA1A-related disorders.
Collapse
Affiliation(s)
- Dilbar Mammadova
- Department of Pediatrics, Pediatric Neurology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Cornelia Kraus
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Leis
- Department of Pediatrics, Pediatric Neurology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bernt Popp
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Center of Functional Genomics, Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Christiane Zweier
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Human Genetics, Inselspital, University of Bern, Bern, Switzerland
| | - Andre Reis
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Centre for Rare Disorders Erlangen, University Hospital Erlangen, Erlangen, Germany
| | - Regina Trollmann
- Department of Pediatrics, Pediatric Neurology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Centre for Rare Disorders Erlangen, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
6
|
Leitch B. Molecular Mechanisms Underlying the Generation of Absence Seizures: Identification of Potential Targets for Therapeutic Intervention. Int J Mol Sci 2024; 25:9821. [PMID: 39337309 PMCID: PMC11432152 DOI: 10.3390/ijms25189821] [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: 08/08/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Understanding the molecular mechanisms underlying the generation of absence seizures is crucial for developing effective, patient-specific treatments for childhood absence epilepsy (CAE). Currently, one-third of patients remain refractive to the antiseizure medications (ASMs), previously called antiepileptic drugs (AEDs), available to treat CAE. Additionally, these ASMs often produce serious side effects and can even exacerbate symptoms in some patients. Determining the precise cellular and molecular mechanisms directly responsible for causing this type of epilepsy has proven challenging as they appear to be complex and multifactorial in patients with different genetic backgrounds. Aberrant neuronal activity in CAE may be caused by several mechanisms that are not fully understood. Thus, dissecting the causal factors that could be targeted in the development of precision medicines without side effects remains a high priority and the ultimate goal in this field of epilepsy research. The aim of this review is to highlight our current understanding of potential causative mechanisms for absence seizure generation, based on the latest research using cutting-edge technologies. This information will be important for identifying potential targets for future therapeutic intervention.
Collapse
Affiliation(s)
- Beulah Leitch
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
7
|
Maity S, Huang Y, Kilgore MD, Thurmon AN, Vaasjo LO, Galazo MJ, Xu X, Cao J, Wang X, Ning B, Liu N, Fan J. Mapping dynamic molecular changes in hippocampal subregions after traumatic brain injury through spatial proteomics. Clin Proteomics 2024; 21:32. [PMID: 38735925 PMCID: PMC11089002 DOI: 10.1186/s12014-024-09485-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/24/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) often results in diverse molecular responses, challenging traditional proteomic studies that measure average changes at tissue levels and fail to capture the complexity and heterogeneity of the affected tissues. Spatial proteomics offers a solution by providing insights into sub-region-specific alterations within tissues. This study focuses on the hippocampal sub-regions, analyzing proteomic expression profiles in mice at the acute (1 day) and subacute (7 days) phases of post-TBI to understand subregion-specific vulnerabilities and long-term consequences. METHODS Three mice brains were collected from each group, including Sham, 1-day post-TBI and 7-day post-TBI. Hippocampal subregions were extracted using Laser Microdissection (LMD) and subsequently analyzed by label-free quantitative proteomics. RESULTS The spatial analysis reveals region-specific protein abundance changes, highlighting the elevation of FN1, LGALS3BP, HP, and MUG-1 in the stratum moleculare (SM), suggesting potential immune cell enrichment post-TBI. Notably, established markers of chronic traumatic encephalopathy, IGHM and B2M, exhibit specific upregulation in the dentate gyrus bottom (DG2) independent of direct mechanical injury. Metabolic pathway analysis identifies disturbances in glucose and lipid metabolism, coupled with activated cholesterol synthesis pathways enriched in SM at 7-Day post-TBI and subsequently in deeper DG1 and DG2 suggesting a role in neurogenesis and the onset of recovery. Coordinated activation of neuroglia and microtubule dynamics in DG2 suggest recovery mechanisms in less affected regions. Cluster analysis revealed spatial variations post-TBI, indicative of dysregulated neuronal plasticity and neurogenesis and further predisposition to neurological disorders. TBI-induced protein upregulation (MUG-1, PZP, GFAP, TJP, STAT-1, and CD44) across hippocampal sub-regions indicates shared molecular responses and links to neurological disorders. Spatial variations were demonstrated by proteins dysregulated in both or either of the time-points exclusively in each subregion (ELAVL2, CLIC1 in PL, CD44 and MUG-1 in SM, and SHOC2, LGALS3 in DG). CONCLUSIONS Utilizing advanced spatial proteomics techniques, the study unveils the dynamic molecular responses in distinct hippocampal subregions post-TBI. It uncovers region-specific vulnerabilities and dysregulated neuronal processes, and potential recovery-related pathways that contribute to our understanding of TBI's neurological consequences and provides valuable insights for biomarker discovery and therapeutic targets.
Collapse
Affiliation(s)
- Sudipa Maity
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Yuanyu Huang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Mitchell D Kilgore
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Abbigail N Thurmon
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
- Tulane Brain Institute, New Orleans, LA, USA
| | | | - Maria J Galazo
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
- Tulane Brain Institute, New Orleans, LA, USA
| | - Xiaojiang Xu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jing Cao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaoying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Bo Ning
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ning Liu
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane University Translational Sciences Institute, New Orleans, LA, USA.
| | - Jia Fan
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA.
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
8
|
Szymanowicz O, Drużdż A, Słowikowski B, Pawlak S, Potocka E, Goutor U, Konieczny M, Ciastoń M, Lewandowska A, Jagodziński PP, Kozubski W, Dorszewska J. A Review of the CACNA Gene Family: Its Role in Neurological Disorders. Diseases 2024; 12:90. [PMID: 38785745 PMCID: PMC11119137 DOI: 10.3390/diseases12050090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/25/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
Calcium channels are specialized ion channels exhibiting selective permeability to calcium ions. Calcium channels, comprising voltage-dependent and ligand-gated types, are pivotal in neuronal function, with their dysregulation is implicated in various neurological disorders. This review delves into the significance of the CACNA genes, including CACNA1A, CACNA1B, CACNA1C, CACNA1D, CACNA1E, CACNA1G, and CACNA1H, in the pathogenesis of conditions such as migraine, epilepsy, cerebellar ataxia, dystonia, and cerebellar atrophy. Specifically, variants in CACNA1A have been linked to familial hemiplegic migraine and epileptic seizures, underscoring its importance in neurological disease etiology. Furthermore, different genetic variants of CACNA1B have been associated with migraine susceptibility, further highlighting the role of CACNA genes in migraine pathology. The complex relationship between CACNA gene variants and neurological phenotypes, including focal seizures and ataxia, presents a variety of clinical manifestations of impaired calcium channel function. The aim of this article was to explore the role of CACNA genes in various neurological disorders, elucidating their significance in conditions such as migraine, epilepsy, and cerebellar ataxias. Further exploration of CACNA gene variants and their interactions with molecular factors, such as microRNAs, holds promise for advancing our understanding of genetic neurological disorders.
Collapse
Affiliation(s)
- Oliwia Szymanowicz
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Artur Drużdż
- Department of Neurology, Municipal Hospital in Poznan, 61-285 Poznan, Poland;
| | - Bartosz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Sandra Pawlak
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Ewelina Potocka
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Ulyana Goutor
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Mateusz Konieczny
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Małgorzata Ciastoń
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Aleksandra Lewandowska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| | - Paweł P. Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (O.S.); (S.P.); (E.P.); (U.G.); (M.K.); (M.C.); (A.L.)
| |
Collapse
|
9
|
Song N, Mei S, Wang X, Hu G, Lu M. Focusing on mitochondria in the brain: from biology to therapeutics. Transl Neurodegener 2024; 13:23. [PMID: 38632601 PMCID: PMC11022390 DOI: 10.1186/s40035-024-00409-w] [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: 12/10/2023] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Mitochondria have multiple functions such as supplying energy, regulating the redox status, and producing proteins encoded by an independent genome. They are closely related to the physiology and pathology of many organs and tissues, among which the brain is particularly prominent. The brain demands 20% of the resting metabolic rate and holds highly active mitochondrial activities. Considerable research shows that mitochondria are closely related to brain function, while mitochondrial defects induce or exacerbate pathology in the brain. In this review, we provide comprehensive research advances of mitochondrial biology involved in brain functions, as well as the mitochondria-dependent cellular events in brain physiology and pathology. Furthermore, various perspectives are explored to better identify the mitochondrial roles in neurological diseases and the neurophenotypes of mitochondrial diseases. Finally, mitochondrial therapies are discussed. Mitochondrial-targeting therapeutics are showing great potentials in the treatment of brain diseases.
Collapse
Affiliation(s)
- Nanshan Song
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shuyuan Mei
- The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166, China
| | - Xiangxu Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Gang Hu
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
- Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China.
| |
Collapse
|
10
|
Modarres Mousavi SM, Alipour F, Noorbakhsh F, Jafarian M, Ghadipasha M, Gharehdaghi J, Kellinghaus C, Speckmann EJ, Stummer W, Khaleghi Ghadiri M, Gorji A. Clinical Correlation of Altered Molecular Signatures in Epileptic Human Hippocampus and Amygdala. Mol Neurobiol 2024; 61:725-752. [PMID: 37658249 PMCID: PMC10861640 DOI: 10.1007/s12035-023-03583-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 08/14/2023] [Indexed: 09/03/2023]
Abstract
Widespread alterations in the expression of various genes could contribute to the pathogenesis of epilepsy. The expression levels of various genes, including major inhibitory and excitatory receptors, ion channels, cell type-specific markers, and excitatory amino acid transporters, were assessed and compared between the human epileptic hippocampus and amygdala, and findings from autopsy controls. Moreover, the potential correlation between molecular alterations in epileptic brain tissues and the clinical characteristics of patients undergoing epilepsy surgery was evaluated. Our findings revealed significant and complex changes in the expression of several key regulatory genes in both the hippocampus and amygdala of patients with intractable epilepsy. The expression changes in various genes differed considerably between the epileptic hippocampus and amygdala. Different correlation patterns were observed between changes in gene expression and clinical characteristics, depending on whether the patients were considered as a whole or were subdivided. Altered molecular signatures in different groups of epileptic patients, defined within a given category, could be viewed as diagnostic biomarkers. Distinct patterns of molecular changes that distinguish these groups from each other appear to be associated with epilepsy-specific functional consequences.
Collapse
Affiliation(s)
| | - Fatemeh Alipour
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Farshid Noorbakhsh
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Maryam Jafarian
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Masoud Ghadipasha
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Jaber Gharehdaghi
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | | | - Erwin-Josef Speckmann
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Walter Stummer
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Department of Neuroscience, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Kramer AA, Bennett DF, Barañano KW, Bannister RA. A neurodevelopmental disorder caused by a dysfunctional CACNA1A allele. eNeurologicalSci 2023; 31:100456. [PMID: 36938367 PMCID: PMC10020665 DOI: 10.1016/j.ensci.2023.100456] [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: 08/03/2022] [Revised: 11/05/2022] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
P/Q-type Ca2+ flux into nerve terminals via CaV2.1 channels is essential for neurotransmitter release at neuromuscular junctions and nearly all central synapses. Mutations in CACNA1A, the gene encoding CaV2.1, cause a spectrum of pediatric neurological disorders. We have identified a patient harboring an autosomal-dominant de novo frameshift-causing nucleotide duplication in CACNA1A (c.5018dupG). The duplicated guanine precipitated 43 residues of altered amino acid sequence beginning with a glutamine to serine substitution in CaV2.1 at position 1674 ending with a premature stop codon (CaV2.1 p.Gln1674Serfs*43). The patient presented with episodic downbeat vertical nystagmus, hypotonia, ataxia, developmental delay and febrile seizures. In patch-clamp experiments, no Ba2+ current was observed in tsA-201 cells expressing CaV2.1 p.Gln1674Serfs*43 with β4 and α2δ-1 auxiliary subunits. The ablation of divalent flux in response to depolarization was likely attributable to the inability of CaV2.1 p.Gln1674Serfs*43 to form a complete channel pore. Our results suggest that the pathology resulting from this frameshift-inducing nucleotide duplication is a consequence of an effective haploinsufficiency.
Collapse
Affiliation(s)
- Audra A. Kramer
- Department of Pathology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD 21201, USA
- Corresponding authors at: National Institutes of Health, Center for Scientific Review, Division of Neuroscience, Development and Aging, 6701 Rockledge Drive, Bethesda, MD 20892, USA.
| | - Daniel F. Bennett
- Department of Pathology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD 21201, USA
| | - Kristin W. Barañano
- Department of Neurology, Johns Hopkins University School of Medicine, 200 North Wolfe Street, Suite 2158, Baltimore, MD 21287, USA
| | - Roger A. Bannister
- Department of Pathology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD 21201, USA
- Corresponding authors at: National Institutes of Health, Center for Scientific Review, Division of Neuroscience, Development and Aging, 6701 Rockledge Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
12
|
Gray MM, Naik A, Ebner TJ, Carter RE. Altered brain state during episodic dystonia in tottering mice decouples primary motor cortex from limb kinematics. DYSTONIA 2023; 2:10974. [PMID: 37800168 PMCID: PMC10554815 DOI: 10.3389/dyst.2023.10974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Episodic Ataxia Type 2 (EA2) is a rare neurological disorder caused by a mutation in the CACNA1A gene, encoding the P/Q-type voltage-gated Ca2+ channel important for neurotransmitter release. Patients with this channelopathy exhibit both cerebellar and cerebral pathologies, suggesting the condition affects both regions. The tottering (tg/tg) mouse is the most commonly used EA2 model due to an orthologous mutation in the cacna1a gene. The tg/tg mouse has three prominent behavioral phenotypes: a dramatic episodic dystonia; absence seizures with generalized spike and wave discharges (GSWDs); and mild ataxia. We previously observed a novel brain state, transient low-frequency oscillations (LFOs) in the cerebellum and cerebral cortex under anesthesia. In this study, we examine the relationships among the dystonic attack, GSWDs, and LFOs in the cerebral cortex. Previous studies characterized LFOs in the motor cortex of anesthetized tg/tg mice using flavoprotein autofluorescence imaging testing the hypothesis that LFOs provide a mechanism for the paroxysmal dystonia. We sought to obtain a more direct understanding of motor cortex (M1) activity during the dystonic episodes. Using two-photon Ca2+ imaging to investigate neuronal activity in M1 before, during, and after the dystonic attack, we show that there is not a significant change in the activity of M1 neurons from baseline through the attack. We also conducted simultaneous, multi-electrode recordings to further understand how M1 cellular activity and local field potentials change throughout the progression of the dystonic attack. Neither putative pyramidal nor inhibitory interneuron firing rate changed during the dystonic attack. However, we did observe a near complete loss of GSWDs during the dystonic attack in M1. Finally, using spike triggered averaging to align simultaneously recorded limb kinematics to the peak Ca2+ response, and vice versa, revealed a reduction in the spike triggered average during the dystonic episodes. Both the loss of GSWDs and the reduction in the coupling suggest that, during the dystonic attack, M1 is effectively decoupled from other structures. Overall, these results indicate that the attack is not initiated or controlled in M1, but elsewhere in the motor circuitry. The findings also highlight that LFOs, GSWDs, and dystonic attacks represent three brain states in tg/tg mice.
Collapse
Affiliation(s)
- Madelyn M Gray
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Anant Naik
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Timothy J Ebner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Russell E Carter
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
13
|
Indelicato E, Boesch S. CACNA1A-Related Channelopathies: Clinical Manifestations and Treatment Options. Handb Exp Pharmacol 2023; 279:227-248. [PMID: 36592223 DOI: 10.1007/164_2022_625] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In the last decade, variants in the Ca2+ channel gene CACNA1A emerged as a frequent aetiology of rare neurological phenotypes sharing a common denominator of variable paroxysmal manifestations and chronic cerebellar dysfunction. The spectrum of paroxysmal manifestations encompasses migraine with hemiplegic aura, episodic ataxia, epilepsy and paroxysmal non-epileptic movement disorders. Additional chronic neurological symptoms range from severe developmental phenotypes in early-onset cases to neurobehavioural disorders and chronic cerebellar ataxia in older children and adults.In the present review we systematically approach the clinical manifestations of CACNA1A variants, delineate genotype-phenotype correlations and elaborate on the emerging concept of an age-dependent phenotypic spectrum in CACNA1A disease. We furthermore reflect on different therapy options available for paroxysmal symptoms in CACNA1A and address open issues to prioritize in the future clinical research.
Collapse
Affiliation(s)
- Elisabetta Indelicato
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Sylvia Boesch
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
14
|
Vithayathil J, Freeman C, Jacobwitz M, Schwartz ES, Agarwal S. Prolonged neurologic deficits with brain MRI changes following ECT in an adolescent with a CACNA1a-related disorder; a case report. BMC Neurol 2022; 22:466. [PMID: 36494636 PMCID: PMC9743018 DOI: 10.1186/s12883-022-02994-7] [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: 08/17/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Electroconvulsive therapy is used to treat depression and schizophrenia with infrequent use in pediatric patients. We report a case of an adolescent with autism spectrum disorder and acute catatonia that presented with status epilepticus (SE) and prolonged neurologic deficits with unilateral left cerebral edema on imaging following unilateral electroconvulsive therapy (ECT) on the right side, subsequently found to have a CACNA1a pathogenic variant. This case highlights a potential adverse effect of ECT in patients with CACNA1a related disorders. CASE The patient received unilateral ECT to the right side and subsequently had an episode of SE with right-sided hemiplegia for 72 h prior to regaining some function with persistent mild right-hand weakness that persisted for at least 1-2 weeks. A brain MRI 2 days after ECT was unremarkable, but a repeat MRI on day four of admission showed left hemisphere cortical diffusion restriction, increased perfusion and T2 prolongation suggestive of cortical edema. They had whole exome genetic testing sent after discharge that showed a known pathogenic CACNA1a variant (p.I1709T). CACNA1a encodes the P/Q type calcium channels and deleterious variants in this gene result in a channelopathy associated with a spectrum of neurodevelopmental disorders that include autism spectrum disorder, hemiplegic migraine with unilateral cerebral edema, epileptic encephalopathies, or episodic ataxia syndromes. CONCLUSION A literature review of ECT and neurologic deficits showed that most neurologic deficits resolve within 30 min of ECT. Case reports of prolonged deficits are rare and there are no prior reports of acute MRI changes related to ECT. Thus, the acute deterioration and MRI findings in this patient are likely related to the underlying CACNA1a channelopathy disorder with ECT as a precipitating event. This case report suggests care should be taken when using ECT in patients with pathogenic variants in CACNA1a. Furthermore, it reinforces the utility and importance of expanded genetic testing in patients with neurodevelopmental disorders as findings can provide valuable information that can guide treatment decisions.
Collapse
Affiliation(s)
- Joseph Vithayathil
- grid.239552.a0000 0001 0680 8770Division of Neurology, Children’s Hospital of Philadelphia, 3401 Civic Center Drive, Philadelphia, PA 19104 USA
| | - Colbey Freeman
- grid.411115.10000 0004 0435 0884Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - Marin Jacobwitz
- grid.239552.a0000 0001 0680 8770Division of Neurology, Children’s Hospital of Philadelphia, 3401 Civic Center Drive, Philadelphia, PA 19104 USA
| | - Erin Simon Schwartz
- grid.411115.10000 0004 0435 0884Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, USA ,grid.239552.a0000 0001 0680 8770Division of Neuroradiology, Children’s Hospital of Philadelphia, Philadelphia, USA
| | - Sonika Agarwal
- grid.239552.a0000 0001 0680 8770Division of Neurology, Children’s Hospital of Philadelphia, 3401 Civic Center Drive, Philadelphia, PA 19104 USA
| |
Collapse
|
15
|
Zhou X, Chen Z, Xiao L, Zhong Y, Liu Y, Wu J, Tao H. Intracellular calcium homeostasis and its dysregulation underlying epileptic seizures. Seizure 2022; 103:126-136. [DOI: 10.1016/j.seizure.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/25/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
|
16
|
Nakao A, Hayashida K, Ogura H, Mori Y, Imoto K. Hippocampus-related cognitive disorders develop in the absence of epilepsy and ataxia in the heterozygous Cacna1a mutant mice tottering. Channels (Austin) 2022; 16:113-126. [PMID: 35548926 PMCID: PMC9103357 DOI: 10.1080/19336950.2022.2072449] [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] [Indexed: 11/17/2022] Open
Abstract
CACNA1A-associated epilepsy and ataxia frequently accompany cognitive impairments as devastating co-morbidities. However, it is unclear whether the cognitive deficits are consequences secondary to the neurological symptoms elicited by CACNA1A mutations. To address this issue, Cacna1a mutant mice tottering (tg), and in particular tg/+ heterozygotes, serve as a suitable model system, given that tg/+ heterozygotes fail to display spontaneous absence epilepsy and ataxia typically observed in tg/tg homozygotes. Here, we examined hippocampus-dependent behaviors and hippocampal learning-related synaptic plasticity in tg mice. In behavioral analyses of tg/+ and tg/tg, acquisition and retention of spatial reference memory were characteristically impaired in the Morris water maze task, while working memory was intact in the eight-arm radial maze and T-maze tasks. tg/+ heterozygotes showed normal motor function in contrast to tg/tg homozygotes. In electrophysiological analyses, Schaffer collateral–CA1 synapses showed a deficit in the maintenance of long-term potentiation in tg/+ and tg/tg mice and an increased paired-pulse facilitation induced by paired pulses with 100 ms in tg/tg mice. Our results indicate that the tg mutation causes a dominant disorder of the hippocampus-related memory and synaptic plasticity, raising the possibility that in CACNA1A-associated human diseases, functionally aberrant CaV2.1 Ca2+ channels actively induce the observed cognitive deficits independently of the neurological symptoms.
Collapse
Affiliation(s)
- Akito Nakao
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Katsumi Hayashida
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Hiroo Ogura
- Product Creation Headquarters, Eisai Corporate, Limited, Tokyo, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Keiji Imoto
- Division of Neural Signaling, Department of Information Physiology, National Institute for Physiological Sciences, Okazaki, Japan
| |
Collapse
|
17
|
Genetic paroxysmal neurological disorders featuring episodic ataxia and epilepsy. Eur J Med Genet 2022; 65:104450. [DOI: 10.1016/j.ejmg.2022.104450] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 02/02/2022] [Accepted: 02/06/2022] [Indexed: 01/25/2023]
|
18
|
Niu X, Yang Y, Chen Y, Cheng M, Liu M, Ding C, Tian X, Yang Z, Jiang Y, Zhang Y. Genotype-phenotype correlation of CACNA1A variants in children with epilepsy. Dev Med Child Neurol 2022; 64:105-111. [PMID: 34263451 DOI: 10.1111/dmcn.14985] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2021] [Indexed: 01/12/2023]
Abstract
AIM To explore the genotypes and phenotypes of CACNA1A variants in children with epilepsy. METHOD Eighteen children (six males, 12 females) with CACNA1A variants were identified using next-generation sequencing. RESULTS There were 14 missense variants, two nonsense variants, one frameshift variant, and one splice site variant. Sixteen variants were de novo. Age at seizure onset ranged from 1 day to 8 years; median age was 8 months. Multiple seizure types were observed, including focal, generalized tonic-clonic, myoclonic, and absence seizures, as well as epileptic spasms and tonic seizures. Focal motor status epilepticus occurred in 10 individuals and generalized motor status epilepticus occurred in two individuals. All 18 children showed developmental delay. Focal motor status epilepticus resulted in cerebral atrophy in five individuals, mainly on the contralateral side. Interictal electroencephalogram showed focal discharges in 12 individuals, whereas five individuals had generalized discharges. Three individuals were seizure-free, whereas 15 still had seizures and five had recurrent status epilepticus at last follow-up. INTERPRETATION Most children with epilepsy and CACNA1A variants had early seizure onset and developmental delay. Focal seizure was the most common seizure type. Most patients experienced status epilepticus. Unilateral cerebral atrophy could occur after focal motor status epilepticus. Patients with CACNA1A variants located in the transmembrane region may be at high risk of status epilepticus.
Collapse
Affiliation(s)
- Xueyang Niu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Ying Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yi Chen
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Miaomiao Cheng
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Ming Liu
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Changhong Ding
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xiaojuan Tian
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Zhixian Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yuehua Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
19
|
Faheem M, Ameer S, Khan AW, Haseeb M, Raza Q, Ali Shah F, Khusro A, Aarti C, Umar Khayam Sahibzada M, El-Saber Batiha G, Koirala N, Adnan M, Alghamdi S, Assaggaf H, Alsiwiehri NO. A comprehensive review on antiepileptic properties of medicinal plants. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2021.103478] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|
20
|
Alehabib E, Kokotović T, Ranji-Burachaloo S, Tafakhori A, Ramshe SM, Esmaeilizadeh Z, Darvish H, Movafagh A, Nagy V. Leu226Trp CACNA1A variant associated with juvenile myoclonic epilepsy with and without intellectual disability. Clin Neurol Neurosurg 2021; 213:107108. [PMID: 34995834 DOI: 10.1016/j.clineuro.2021.107108] [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: 07/27/2021] [Revised: 11/28/2021] [Accepted: 12/25/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Epilepsy is a disease of Central Nervous System (CNS) characterized by abnormal brain activity and recurrent seizures and is considered a clinically and genetically heterogeneous disease. Here, we investigated pathogenic genetic alteration and described the clinical characteristics of three Iranian family members affected by Idiopathic Generalized Epilepsy (IGE) with and without intellectual disability. METHODS A non-consanguineous Iranian family with juvenile myoclonic epilepsy was enrolled in the study. The comprehensive neurological evaluation included motor and sensory skills, vision, hearing, speech, coordination, and mood. Whole-exome Sequencing (WES) was performed on the proband to detect probable pathogenic variant, and after the filtering process, probable variants were evaluated with familial segregation analysis using Sanger sequencing. RESULTS Using WES, we identified a heterozygous missense substitution (NM_023035.3:c.T677G:p.Leu226Trp) in CACNA1A gene in the studied family with juvenile myoclonic epilepsy with and without intellectual disability and psychiatric phenotype. Considering the patients' clinical synopsis, familial segregation analysis, and literature review, we postulated this variant to be causative of the disease. Indeed, the resulting missense mutation of Leu226Trp affects a highly conserved residue supporting our hypothesis that this mutation is potentially pathogenic. CONCLUSION To the best of our knowledge, this is the first report of juvenile myoclonic epilepsy related to CACNA1A gene. Our results provide evidence for expanding the clinical and molecular findings related to the CACNA1A gene.
Collapse
Affiliation(s)
- Elham Alehabib
- Student Research Committee, Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, 1090 Vienna, Austria
| | - Tomislav Kokotović
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, 1090 Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Sakineh Ranji-Burachaloo
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Molaei Ramshe
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Esmaeilizadeh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Darvish
- Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abolfazl Movafagh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Vanja Nagy
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, 1090 Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
21
|
Alehabib E, Esmaeilizadeh Z, Ranji-Burachaloo S, Tafakhori A, Darvish H, Movafagh A. Clinical and molecular spectrum of P/Q type calcium channel Cav2.1 in epileptic patients. Orphanet J Rare Dis 2021; 16:461. [PMID: 34727962 PMCID: PMC8562004 DOI: 10.1186/s13023-021-02101-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 10/24/2021] [Indexed: 11/24/2022] Open
Abstract
Background Epilepsy is a neurological disorder characterized by the potential to induce seizure and accompanied by cognitive, psychological, and social consequences. CACNA1A gene is a voltage-gated P/Q-type Cav2.1 channel that is broadly expressed in the central nervous system, and the pathogenic variants within this gene may be associated with the epileptic phenotype. In the present study, we collected clinical and molecular data related to epileptic patients with CACNA1A pathogenic variants and investigated possible meaningful relationship between age at onset, neurodevelopmental disorders, type of seizures, brain imaging abnormalities, genotype, and protein domains. Results In our retrospective literature studies, from among 890 articles reviewed, a total of 90 individuals were related to epilepsy phenotype. Our findings showed that about 90 percent of patients have shown the first symptoms in childhood and teenage years and different types of neurodevelopmental disorders, such as intellectual disability, developmental arrest, and behavioral disorders, have been common findings for these patients. Further, a wide range of abnormalities have been observed in their brain imaging, and generalized seizures have been the most type of seizures in these patients. However, our data showed no specific genotype–phenotype correlation in epileptic patients with CACNA1A pathogenic alterations. Conclusions Our study focused on epileptic phenotype in patients with CACNA1A pathogenic variants and showed a wide range of clinical and molecular heterogeneity with no specific genotype–phenotype correlation. It seems that incomplete penetrance, de-novo variants, and modifier genes are obstacles in predicting the clinical outcome. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-02101-y.
Collapse
Affiliation(s)
- Elham Alehabib
- Student Research Committee, Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Esmaeilizadeh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sakineh Ranji-Burachaloo
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Darvish
- Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abolfazl Movafagh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Le Roux M, Barth M, Gueden S, Desbordes de Cepoy P, Aeby A, Vilain C, Hirsch E, de Saint Martin A, Portes VD, Lesca G, Riquet A, Chaton L, Villeneuve N, Villard L, Cances C, Valton L, Renaldo F, Vermersch AI, Altuzarra C, Nguyen-Morel MA, Van Gils J, Angelini C, Biraben A, Arnaud L, Riant F, Van Bogaert P. CACNA1A-associated epilepsy: Electroclinical findings and treatment response on seizures in 18 patients. Eur J Paediatr Neurol 2021; 33:75-85. [PMID: 34102571 DOI: 10.1016/j.ejpn.2021.05.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/25/2021] [Accepted: 05/19/2021] [Indexed: 02/02/2023]
Abstract
CACNA1A pathogenic mutations are involved in various neurological phenotypes including episodic ataxia (EA2), spinocerebellar ataxia (SCA6), and familial hemiplegic migraine (FHM1). Epilepsy is poorly documented. We studied 18 patients (10 males) carrying de novo or inherited CACNA1A mutations, with median age of 2,5 years at epilepsy onset. Eight mutations were novel. Two variants known leading to gain of function (GOF) were found in 5 patients. Five other patients had non-sense variants leading to loss of function (LOF). Seizures were most often revealed by either status epilepticus (SE) (n = 8), eventually triggered by fever (n = 5), or absences/behavioural arrests (n = 7). Non-epileptic paroxysmal events were frequent and consisted in recurrent hemiplegic accesses (n = 9), jitteriness in the neonatal period (n = 6), and ocular paroxysmal events (n = 9). Most of the patients had early permanent cerebellar dysfunction (n = 16) and early moderate to severe global developmental delay (GDD)/intellectual deficiency (ID) (n = 17). MRI was often abnormal, with cerebellar (n = 8) and/or cerebral (n = 6) atrophy. Stroke-like occurred in 2 cases. Some antiepileptic drugs including topiramate, levetiracetam, lamotrigine and valproate were effective on seizures. Acetazolamide and calcium channel blockers were often effective when used. More than half of the patients had refractory epilepsy. CACNA1A mutation should be evoked in front of 2 main electro-clinical phenotypes that are associated with permanent cerebellar dysfunction and moderate to severe GDD/ID. The first one, found in all 5 patients with GOF variants, is characterized by intractable seizures, early and recurrent SE and hemiplegic accesses. The second, less severe, found in 5 patients with LOF variants, is characterized by refractory early onset absence seizures.
Collapse
Affiliation(s)
- Marie Le Roux
- Department of Pediatric Neurology and Neurosurgery, CHU Angers, France.
| | | | - Sophie Gueden
- Department of Pediatric Neurology and Neurosurgery, CHU Angers, France
| | | | - Alec Aeby
- Department of Pediatric Neurology, HUDERF, Bruxelles, Belgium
| | - Catheline Vilain
- Department of Medical Genetics, Erasme Hospital, Bruxelles, Belgium
| | | | | | - Vincent des Portes
- Department of Pediatric Neurology, Hospices civils de Lyon, Bron, France
| | - Gaëtan Lesca
- Department of Genetics, Hospices civils de Lyon, Bron, France
| | - Audrey Riquet
- Department of Pediatric Neurology, CHRU Lille, France
| | | | - Nathalie Villeneuve
- Department of Pediatric Neurology, Hôpital de La Timone, AP-HM, Marseille, France
| | - Laurent Villard
- Department of Medical Genetics, Hôpital de La Timone, AP-HM, Marseille, France; Aix Marseille Univ, Inserm, Marseille Medical Genetics, U1251, Marseille, France
| | - Claude Cances
- Department of Pediatric Neurology, CHU Purpan, Toulouse, France
| | - Luc Valton
- Explorations Neurophysiologiques, CHU Purpan, Toulouse, France; Centre de Recherche Cerveau et Cognition (CerCo), University of Toulouse, Toulouse F, 31300, France
| | - Florence Renaldo
- Department of Pediatric Neurology, Hôpital Trousseau, Assistance publique-Hôpitaux de Paris, France
| | - Anne-Isabelle Vermersch
- Department of Neurophysiology, Hôpital Trousseau, Assistance publique-Hôpitaux de Paris, France
| | | | | | - Julien Van Gils
- Department of Medical Genetics, CHU Bordeaux Pellegrin, Bordeaux, France
| | - Chloé Angelini
- Department of Medical Genetics, CHU Bordeaux Pellegrin, Bordeaux, France
| | - Arnaud Biraben
- Department of Neurology, CHU Rennes Pontchaillou, Rennes, France
| | - Lionel Arnaud
- Department of Genetics, Hôpital de la Pitie Salpetrière, Assistance publique-Hôpitaux de Paris, France
| | - Florence Riant
- Department of Genetics, Groupe hospitalier Saint Louis-Lariboisière, Assistance publique-Hôpitaux de Paris, France
| | - Patrick Van Bogaert
- Department of Pediatric Neurology and Neurosurgery, CHU Angers, France; Laboratoire Angevin de Recherche en Ingénierie des Systèmes (LARIS), Université d'Angers, France
| |
Collapse
|
23
|
Classifying epilepsy pragmatically: Past, present, and future. J Neurol Sci 2021; 427:117515. [PMID: 34174531 PMCID: PMC7613525 DOI: 10.1016/j.jns.2021.117515] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 01/31/2023]
Abstract
The classification of epilepsy is essential for people with epilepsy and their families, healthcare providers, physicians and researchers. The International League Against Epilepsy proposed updated seizure and epilepsy classifications in 2017, while another four-dimensional epilepsy classification was updated in 2019. An Integrated Epilepsy Classification system was proposed in 2020. Existing classifications, however, lack consideration of important pragmatic factors relevant to the day-to-day life of people with epilepsy and stakeholders. Despite promising developments, consideration of comorbidities in brain development, genetic causes, and environmental triggers of epilepsy remains largely user-dependent in existing classifications. Demographics of epilepsy have changed over time, while existing classification schemes exhibit caveats. A pragmatic classification scheme should incorporate these factors to provide a nuanced classification. Validation across disparate contexts will ensure widespread applicability and ease of use. A team-based approach may simplify communication between healthcare personnel, while an individual-centred perspective may empower people with epilepsy. Together, incorporating these elements into a modern but pragmatic classification scheme may ensure optimal care for people with epilepsy by emphasising cohesiveness among its myriad users. Technological advancements such as 7T MRI, next-generation sequencing, and artificial intelligence may affect future classification efforts.
Collapse
|
24
|
Pulvirenti G, Caccamo M, Lo Bianco M, Mazzurco M, Praticò ER, Giallongo A, Gangi G, Zanghì A, Falsaperla R. Calcium Channels Genes and Their Epilepsy Phenotypes. JOURNAL OF PEDIATRIC NEUROLOGY 2021. [DOI: 10.1055/s-0041-1728684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractCalcium (Ca2+) channel gene mutations play an important role in the pathogenesis of neurological episodic disorders like epilepsy. CACNA1A and CACNA1H genes are involved in the synthesis of calcium channels. Mutations in the α1A subunit of the P/Q type voltage-gated calcium channel gene (CACNA1A) located in 19p13.13, which encodes for the transmembrane pore-forming subunit of CAV2.1 voltage-dependent calcium channel, have been correlated to a large clinical spectrum of epilepsy such as idiopathic genetic epilepsy, early infantile epilepsy, and febrile seizures. Moreover, CACNA1A mutations have been demonstrated to be involved in spinocerebellar ataxia type 6, familiar hemiplegic migraine, episodic ataxia type 2, early-onset encephalopathy, and hemiconvulsion–hemiplegia epilepsy syndrome. This wide phenotype heterogeneity associated with CACNA1A mutations is correlated to different clinical and electrophysiological manifestations. CACNA1H gene, located in 16p13.3, encodes the α1H subunit of T-type calcium channel, expressing the transmembrane pore-forming subunit Cav3.2. Despite data still remain controversial, it has been identified as an important gene whose mutations seem strictly related to the pathogenesis of childhood absence epilepsy and other generalized epilepsies. The studied variants are mainly gain-of-function, hence responsible for an increase in neuronal susceptibility to seizures. CACNA1H mutations have also been associated with autism spectrum disorder and other behavior disorders. More recently, also amyotrophic lateral sclerosis has been related to CACNA1H alterations. The aim of this review, other than describe the CACNA1A and CACNA1H gene functions, is to identify mutations reported in literature and to analyze their possible correlations with specific epileptic disorders, purposing to guide an appropriate medical treatment recommendation.
Collapse
Affiliation(s)
- Giulio Pulvirenti
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Martina Caccamo
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Manuela Lo Bianco
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | | | - Alessandro Giallongo
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Gloria Gangi
- Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Antonio Zanghì
- Department of Medical and Surgical Sciences and Advanced Technology “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Raffaele Falsaperla
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
- Unit of Neonatal Intensive Care and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
| |
Collapse
|
25
|
Verriello L, Pauletto G, Nilo A, Lonigro I, Betto E, Valente M, Curcio F, Gigli GL. Epilepsy and episodic ataxia type 2: family study and review of the literature. J Neurol 2021; 268:4296-4302. [PMID: 33983550 DOI: 10.1007/s00415-021-10555-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 10/21/2022]
Abstract
Episodic ataxia type 2 (EA2) is a hereditary disorder characterized by paroxysmal attacks of ataxia, vertigo and nausea, due to mutations in the CACNA1A gene, which encodes for α1 subunit of the P/Q-type voltage-gated Ca2+ channel (CaV2.1). Other manifestations may be associated to CACNA1A mutations, such as migraine and epilepsy. The correlation between episodic ataxia and epilepsy is often underestimated and misdiagnosed. Clinical presentation of EA2 varies among patients and within the same family, and the same genetic mutation can lead to different clinical phenotypes. We herewith describe an Italian family presenting with typical EA2 and, in two of the family members (patients II.3 and III.1), epileptic seizures. The sequencing revealed a heterozygous deletion of 6 nucleotides in exon 28 of CACNA1A gene, present in all affected patients. Evidence suggests that mutations of CACNA1A, conferring a loss/reduction of CaV2.1 function, lead to an increase of thalamocortical excitation that contributes to epileptiform discharges. Our description highlights intra-family variability of EA2 phenotype and suggests that mutations in the CACNA1A gene should be suspected in individuals with focal or generalized epilepsy, associated with a family history of episodic ataxia.
Collapse
Affiliation(s)
- Lorenzo Verriello
- Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, ASUFC, Piazzale Santa Maria della Misericordia 15, 33100, Udine, Italy.
| | - Giada Pauletto
- Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, ASUFC, Piazzale Santa Maria della Misericordia 15, 33100, Udine, Italy
| | - Annacarmen Nilo
- Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Incoronata Lonigro
- Department of Medicine (DAME), University of Udine, Udine, Italy.,Department of Laboratory Medicine, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Elena Betto
- Department of Laboratory Medicine, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Mariarosaria Valente
- Department of Medicine (DAME), University of Udine, Udine, Italy.,Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Francesco Curcio
- Department of Medicine (DAME), University of Udine, Udine, Italy.,Department of Laboratory Medicine, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy
| | - Gian Luigi Gigli
- Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, ASUFC, Udine, Italy.,Department of Mathematics, Informatics and Physics (DMIF), University of Udine, Udine, Italy
| |
Collapse
|
26
|
Ferron L, Koshti S, Zamponi GW. The life cycle of voltage-gated Ca 2+ channels in neurons: an update on the trafficking of neuronal calcium channels. Neuronal Signal 2021; 5:NS20200095. [PMID: 33664982 PMCID: PMC7905535 DOI: 10.1042/ns20200095] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/15/2021] [Indexed: 01/26/2023] Open
Abstract
Neuronal voltage-gated Ca2+ (CaV) channels play a critical role in cellular excitability, synaptic transmission, excitation-transcription coupling and activation of intracellular signaling pathways. CaV channels are multiprotein complexes and their functional expression in the plasma membrane involves finely tuned mechanisms, including forward trafficking from the endoplasmic reticulum (ER) to the plasma membrane, endocytosis and recycling. Whether genetic or acquired, alterations and defects in the trafficking of neuronal CaV channels can have severe physiological consequences. In this review, we address the current evidence concerning the regulatory mechanisms which underlie precise control of neuronal CaV channel trafficking and we discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Laurent Ferron
- Department of Physiology and Pharmacology, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Saloni Koshti
- Department of Physiology and Pharmacology, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Gerald W. Zamponi
- Department of Physiology and Pharmacology, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
27
|
Indelicato E, Boesch S. From Genotype to Phenotype: Expanding the Clinical Spectrum of CACNA1A Variants in the Era of Next Generation Sequencing. Front Neurol 2021; 12:639994. [PMID: 33737904 PMCID: PMC7960780 DOI: 10.3389/fneur.2021.639994] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
Ion channel dysfunction is a key pathological substrate of episodic neurological disorders. A classical gene associated to paroxysmal movement disorders is CACNA1A, which codes for the pore-forming subunit of the neuronal calcium channel P/Q. Non-polyglutamine CACNA1A variants underlie familial hemiplegic ataxia type 1 (FHM1) and episodic ataxia type 2 (EA2). Classical paroxysmal manifestations of FHM1 are migraine attacks preceded by motor aura consisting of hemiparesis, aphasia, and disturbances of consciousness until coma. Patients with EA2 suffer of recurrent episodes of vertigo, unbalance, diplopia, and vomiting. Beyond these typical presentations, several reports highlighted manifold clinical features associated with P/Q channelopathies, from chronic progressive cerebellar ataxia to epilepsy and psychiatric disturbances. These manifestations may often outlast the burden of classical episodic symptoms leading to pitfalls in the diagnostic work-up. Lately, the spreading of next generation sequencing techniques linked de novo CACNA1A variants to an even broader phenotypic spectrum including early developmental delay, autism spectrum disorders, epileptic encephalopathy, and early onset paroxysmal dystonia. The age-dependency represents a striking new aspect of these phenotypes und highlights a pivotal role for P/Q channels in the development of the central nervous system in a defined time window. While several reviews addressed the clinical presentation and treatment of FHM1 and EA2, an overview of the newly described age-dependent manifestations is lacking. In this Mini-Review we present a clinical update, delineate genotype-phenotype correlations as well as summarize evidence on the pathophysiological mechanisms underlying the expanded phenotype associated with CACNA1A variants.
Collapse
Affiliation(s)
| | - Sylvia Boesch
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Li Y, Chang C, Zhu Z, Sun L, Fan C. Terahertz Wave Enhances Permeability of the Voltage-Gated Calcium Channel. J Am Chem Soc 2021; 143:4311-4318. [PMID: 33625851 DOI: 10.1021/jacs.0c09401] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A deficiency of Ca2+ fluxes arising from dysfunctional voltage-gated calcium channels has been associated with a list of calcium channelopathies such as epilepsy, hypokalemic periodic paralysis, episodic ataxia, etc. Apart from analyzing the pathogenic channel mutations, understanding how the channel regulates the ion conduction would be instructive to the treatment as well. In the present work, in relating the free energetics of Ca2+ transport to the calcium channel, we demonstrate the importance of bridging Ca2+ hydration waters, which form hydrogen bonds with channel -COO- and -C═O groups and enable a long-distance effect on the Ca2+ permeation. By firing a terahertz wave which resonates with the stretching mode of either the -COO- or the -C═O group, we obtain significantly enhanced selectivity and conductance of Ca2+. The Ca2+ free energy negatively grows nearly 5-fold. The direct evidence is the reinforced hydrogen bonds. In addition, thanks to forced vibrations, -COO- contributes to raised permeation as well even under a field in resonance with -C═O, and vice versa. Since the resonant terahertz field could manipulate the conduction of calcium channels, it has potential applications in therapeutic intervention such as rectifying a Ca2+ deficiency in degraded calcium channels, inducing apoptosis of tumor cells with overloaded calcium etc.
Collapse
Affiliation(s)
- Yangmei Li
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing 100071, People's Republic of China
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing 100071, People's Republic of China.,Key Laboratory of Physical Electronics and Devices of the Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Zhi Zhu
- School of Optical-Electrical Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, People's Republic of China
| | - Lan Sun
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing 100071, People's Republic of China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, and Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| |
Collapse
|
29
|
Indelicato E, Unterberger I, Nachbauer W, Eigentler A, Amprosi M, Zeiner F, Haberlandt E, Kaml M, Gizewski E, Boesch S. The electrophysiological footprint of CACNA1A disorders. J Neurol 2021; 268:2493-2505. [PMID: 33544220 PMCID: PMC8217028 DOI: 10.1007/s00415-021-10415-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 12/14/2022]
Abstract
Objectives CACNA1A variants underlie three neurological disorders: familial hemiplegic migraine type 1 (FHM1), episodic ataxia type 2 (EA2) and spinocerebellar ataxia type 6 (SCA6). EEG is applied to study their episodic manifestations, but findings in the intervals did not gain attention up to date. Methods We analyzed repeated EEG recordings performed between 1994 and 2019 in a large cohort of genetically confirmed CACNA1A patients. EEG findings were compared with those of CACNA1A-negative phenocopies. A review of the related literature was performed. Results 85 EEG recordings from 38 patients (19 EA2, 14 FHM1, 5 SCA6) were analyzed. Baseline EEG was abnormal in 55% of cases (12 EA2, 9 FHM1). The most common finding was a lateralized intermittent slowing, mainly affecting the temporal region. Slowing was more pronounced after a recent attack but was consistently detected in the majority of patients also during the follow-up. Interictal epileptic discharges (IEDs) were detected in eight patients (7 EA2,1 FHM1). EEG abnormalities and especially IEDs were significantly associated with younger age at examination (16 ± 9 vs 43 ± 21 years in those without epileptic changes, p = 0.003) and with earlier onset of disease (1 (1–2) vs 12 (5–45) years, p = 0.0009). EEG findings in CACNA1A-negative phenocopies (n = 15) were largely unremarkable (p = 0.03 in the comparison with CACNA1A patients). Conclusions EEG abnormalities between attacks are highly prevalent in episodic CACNA1A disorders and especially associated with younger age at examination and earlier disease onset. Our findings underpin an age-dependent effect of CACNA1A variants, with a more severe impairment when P/Q channel dysfunction manifests early in life.
Collapse
Affiliation(s)
- Elisabetta Indelicato
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Iris Unterberger
- Epileptology Division, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Wolfgang Nachbauer
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Andreas Eigentler
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Matthias Amprosi
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Fiona Zeiner
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Edda Haberlandt
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
- Department of Pediatrics, City Hospital, Dornbirn, Austria
| | - Manuela Kaml
- Epileptology Division, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elke Gizewski
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sylvia Boesch
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
30
|
Myers KA, Bennett MF, Grinton BE, Dabscheck G, Chan EK, Bello-Espinosa LE, Sadleir LG, D'Alfonso S, Schneider AL, Damiano JA, Hildebrand MS, Bahlo M, Berkovic SF, Buchhalter J, Scheffer IE. Contribution of rare genetic variants to drug response in absence epilepsy. Epilepsy Res 2021; 170:106537. [PMID: 33421703 DOI: 10.1016/j.eplepsyres.2020.106537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 01/02/2023]
Abstract
OBJECTIVE We investigated the possible significance of rare genetic variants to response to valproic acid (VPA) and ethosuximide (ETX) in patients with absence epilepsy. Our primary hypothesis was that rare CACNA1H variants are more frequent in ETX-non-responsive patients compared to ETX-responsive. Our secondary hypothesis was that rare variants in GABA-receptor genes are more frequent in VPA-non-responsive patients compared to VPA-responsive. METHODS We recruited patients with absence epilepsy treated with both VPA and ETX, and performed whole exome sequencing in order to investigate the potential role of rare variants in CACNA1H, other voltage-gated calcium channel (VGCC) genes, or GABA-receptor genes in predicting response to ETX or VPA. RESULTS Sixty-two patients were included; 12 were ETX-responsive, 14 VPA-responsive, and 36 did not have a clear positive response to either medication. We did not find significant enrichment inCACNA1H rare variants in ETX-responsive patients (odds ratio 3.43; 0.43-27.65; p = 0.20), nor was there enrichment for other VGCC genes. No significant enrichment of GABA-receptor gene rare variants was seen for VPA-non-responsive patients versus VPA-responsive. We found enrichment of rare GABA-receptor variants in our absence cohort compared to controls (odds ratio 3.82; 1.68-8.69). There was no difference in frequency of CACNA1H rs61734410 and CACNA1I rs3747178 polymorphisms between ETX-responsive and ETX-non-responsive groups; these polymorphisms have previously been reported to predict lack of response to ETX in absence epilepsy. SIGNIFICANCE We conclude that if CACNA1H rare variants predict lack of response to ETX, a larger sample is necessary to test this with sufficient power. Increased GABA-receptor gene rare variant frequency in absence epilepsy patients who fail initial anti-seizure therapy suggests subtle GABA receptor dysfunction may contribute to the underlying pathophysiology.
Collapse
Affiliation(s)
- Kenneth A Myers
- Department of Pediatrics, Division of Child Neurology, Montreal Children's Hospital, McGill University Health Centre, 1001 Décarie Blvd, Montreal, PQ, H4A 3J1, Canada; Department of Neurology & Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, 1001 Décarie Blvd, Montreal, PQ, H4A 3J1, Canada; Research Institute of the McGill University Health Center, 1001 Décarie Blvd, Montreal, PQ, H4A 3J1, Canada; Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, 245 Burgundy St, Heidelberg, VIC, 3084, Australia.
| | - Mark F Bennett
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, 245 Burgundy St, Heidelberg, VIC, 3084, Australia; Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, 1G, Royal Parade, Parkville, VIC, 3052, Australia; Department of Medical Biology, The University of Melbourne, 1G, Royal Parade, Parkville, VIC, 3052, Australia
| | - Bronwyn E Grinton
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, 245 Burgundy St, Heidelberg, VIC, 3084, Australia
| | - Gabriel Dabscheck
- Department of Neurology, Royal Children's Hospital, The University of Melbourne, 50 Flemington Rd, Parkville, VIC, 3052, Australia
| | - Eunice K Chan
- Department of Neurology, Royal Children's Hospital, The University of Melbourne, 50 Flemington Rd, Parkville, VIC, 3052, Australia
| | - Luis E Bello-Espinosa
- Division of Pediatric Neurology, Department of Pediatrics, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
| | - Lynette G Sadleir
- Department of Paediatrics and Child Health, University of Otago, Wellington, 23 Mein Street, Newtown, Wellington, 6021, New Zealand
| | - Sabrina D'Alfonso
- Division of Pediatric Neurology, Department of Pediatrics, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
| | - Amy L Schneider
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, 245 Burgundy St, Heidelberg, VIC, 3084, Australia
| | - John A Damiano
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, 245 Burgundy St, Heidelberg, VIC, 3084, Australia
| | - Michael S Hildebrand
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, 245 Burgundy St, Heidelberg, VIC, 3084, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville, VIC, 3052, Australia
| | - Melanie Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, 1G, Royal Parade, Parkville, VIC, 3052, Australia; Department of Medical Biology, The University of Melbourne, 1G, Royal Parade, Parkville, VIC, 3052, Australia
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, 245 Burgundy St, Heidelberg, VIC, 3084, Australia
| | - Jeffrey Buchhalter
- Division of Pediatric Neurology, Department of Pediatrics, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, 28 Oki Dr NW, Calgary, AB, T3B 6A8, Canada
| | - Ingrid E Scheffer
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, 245 Burgundy St, Heidelberg, VIC, 3084, Australia; Department of Neurology, Royal Children's Hospital, The University of Melbourne, 50 Flemington Rd, Parkville, VIC, 3052, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville, VIC, 3052, Australia; Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, Parkville, VIC, 3052, Australia
| |
Collapse
|
31
|
Gur-Hartman T, Berkowitz O, Yosovich K, Roubertie A, Zanni G, Macaya A, Heimer G, Dueñas BP, Sival DA, Pode-Shakked B, López-Laso E, Humbertclaude V, Riant F, Bosco L, Cayron LB, Nissenkorn A, Nicita F, Bertini E, Hassin S, Ben Zeev B, Zerem A, Libzon S, Lev D, Linder I, Lerman-Sagie T, Blumkin L. Clinical phenotypes of infantile onset CACNA1A-related disorder. Eur J Paediatr Neurol 2021; 30:144-154. [PMID: 33349592 DOI: 10.1016/j.ejpn.2020.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND CACNA1A-related disorders present with persistent progressive and non-progressive cerebellar ataxia and paroxysmal events: epileptic seizures and non-epileptic attacks. These phenotypes overlap and co-exist in the majority of patients. OBJECTIVE To describe phenotypes in infantile onset CACNA1A-related disorder and to explore intra-familial variations and genotype-phenotype correlations. MATERIAL AND METHODS This study was a multicenter international collaboration. A retrospective chart review of CACNA1A patients was performed. Clinical, radiological, and genetic data were collected and analyzed in 47 patients with infantile-onset disorder. RESULTS Paroxysmal non-epileptic events (PNEE) were observed in 68% of infants, with paroxysmal tonic upward gaze (PTU) noticed in 47% of infants. Congenital cerebellar ataxia (CCA) was diagnosed in 51% of patients including four patients with developmental delay and only one neurological sign. PNEEs were found in 63% of patients at follow-up, with episodic ataxia (EA) in 40% of the sample. Cerebellar ataxia was found in 58% of the patients at follow-up. Four patients had epilepsy in infancy and nine in childhood. Seven infants had febrile convulsions, three of which developed epilepsy later; all three patients had CCA. Cognitive difficulties were demonstrated in 70% of the children. Cerebellar atrophy was found in only one infant but was depicted in 64% of MRIs after age two. CONCLUSIONS Nearly all of the infants had CCA, PNEE or both. Cognitive difficulties were frequent and appeared to be associated with CCA. Epilepsy was more frequent after age two. Febrile convulsions in association with CCA may indicate risk of epilepsy in later childhood. Brain MRI was normal in infancy. There were no genotype-phenotype correlations found.
Collapse
Affiliation(s)
- Tamar Gur-Hartman
- Pediatric Neurology Unit, Wolfson Medical Center, Holon, Israel; Pediatric Movement Disorders Service, Wolfson Medical Center, Holon, Israel; School of Psychological Sciences, Tel-Aviv University, Israel
| | - Oren Berkowitz
- Department of Health Systems Management, Ariel University, Ariel, Israel
| | - Keren Yosovich
- Molecular Genetics Laboratory, Wolfson Medical Center, Holon, Israel
| | - Agathe Roubertie
- Departement de Neuropediatrie, CHU Gui de Chauliac, Institut des Neurosciences de Montpellier, Montpellier, France
| | - Ginevra Zanni
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, IRCCS Bambino Gesu' Children's Hospital, Rome, Italy
| | - Alfons Macaya
- Vall d'Hebron Research Institute, Pediatric Neurology Research Group, Autonomous University of Barcelona, Barcelona, Spain
| | - Gali Heimer
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Belén Pérez Dueñas
- Vall d'Hebron Research Institute, Pediatric Neurology Research Group, Autonomous University of Barcelona, Barcelona, Spain
| | - Deborah A Sival
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ben Pode-Shakked
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; The Institute for Rare Diseases, Edmond and Lily Safra Children's Hospital; Talpiot Medical Leadership Program, Sheba Medical Center, Ramat Gan, Israel
| | - Eduardo López-Laso
- University Hospital Reina Sofía, Pediatric Neurology Unit, IMIBIC and CIBERER, Córdoba, Spain
| | - Véronique Humbertclaude
- Service de Médecine Psychologique Enfants et Adolescents, CHU Saint Eloi, Montpellier, France
| | - Florence Riant
- AP-HP, GH Saint Louis-Lariboisière-Fernand Widal, Service de Génétique Moléculaire Neurovasculaire, Paris, France
| | - Luca Bosco
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, IRCCS Bambino Gesu' Children's Hospital, Rome, Italy
| | | | - Andreea Nissenkorn
- Pediatric Neurology Unit, Wolfson Medical Center, Holon, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Francesco Nicita
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, IRCCS Bambino Gesu' Children's Hospital, Rome, Italy
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, IRCCS Bambino Gesu' Children's Hospital, Rome, Italy
| | - Sharon Hassin
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Movement Disorders Institute and Department of Neurology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Bruria Ben Zeev
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Ayelet Zerem
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Pediatric Neurology Unit TASMC, Tel-Aviv University, Israel
| | | | - Dorit Lev
- Molecular Genetics Laboratory, Wolfson Medical Center, Holon, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Medical Genetics, Wolfson Medical Center, Holon, Israel
| | - Ilan Linder
- Pediatric Epilepsy & Neurology Service, Barzilay Medical Center, Ashkelon, Israel
| | - Tally Lerman-Sagie
- Pediatric Neurology Unit, Wolfson Medical Center, Holon, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Lubov Blumkin
- Pediatric Neurology Unit, Wolfson Medical Center, Holon, Israel; Pediatric Movement Disorders Service, Wolfson Medical Center, Holon, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
32
|
Jaudon F, Baldassari S, Musante I, Thalhammer A, Zara F, Cingolani LA. Targeting Alternative Splicing as a Potential Therapy for Episodic Ataxia Type 2. Biomedicines 2020; 8:E332. [PMID: 32899500 PMCID: PMC7555146 DOI: 10.3390/biomedicines8090332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 12/26/2022] Open
Abstract
Episodic ataxia type 2 (EA2) is an autosomal dominant neurological disorder characterized by paroxysmal attacks of ataxia, vertigo, and nausea that usually last hours to days. It is caused by loss-of-function mutations in CACNA1A, the gene encoding the pore-forming α1 subunit of P/Q-type voltage-gated Ca2+ channels. Although pharmacological treatments, such as acetazolamide and 4-aminopyridine, exist for EA2, they do not reduce or control the symptoms in all patients. CACNA1A is heavily spliced and some of the identified EA2 mutations are predicted to disrupt selective isoforms of this gene. Modulating splicing of CACNA1A may therefore represent a promising new strategy to develop improved EA2 therapies. Because RNA splicing is dysregulated in many other genetic diseases, several tools, such as antisense oligonucleotides, trans-splicing, and CRISPR-based strategies, have been developed for medical purposes. Here, we review splicing-based strategies used for genetic disorders, including those for Duchenne muscular dystrophy, spinal muscular dystrophy, and frontotemporal dementia with Parkinsonism linked to chromosome 17, and discuss their potential applicability to EA2.
Collapse
Affiliation(s)
- Fanny Jaudon
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Simona Baldassari
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (S.B.); (I.M.); (F.Z.)
| | - Ilaria Musante
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (S.B.); (I.M.); (F.Z.)
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16126 Genoa, Italy
| | - Agnes Thalhammer
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Federico Zara
- Unit of Medical Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (S.B.); (I.M.); (F.Z.)
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16126 Genoa, Italy
| | - Lorenzo A. Cingolani
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), 16132 Genoa, Italy;
| |
Collapse
|
33
|
Kumar P, Sharma D. Ameliorative effect of curcumin on altered expression of CACNA1A and GABRD in the pathogenesis of FeCl 3-induced epilepsy. Mol Biol Rep 2020; 47:5699-5710. [PMID: 32803504 DOI: 10.1007/s11033-020-05538-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/17/2020] [Indexed: 11/28/2022]
Abstract
The pivotal role played by ion-channel dysregulations in the pathogenesis of epilepsy has always garnered much attention. Since mutation of ion-channel proteins CACNA1A and GABRD have been associated with epilepsy, it is important to determine the post-traumatic epilepsy-associated changes in expression levels of these ion channel proteins. Additionally, curcumin is already known for its antiepileptic and neuroprotective potential in FeCl3-induced model of post-traumatic epilepsy. Thus, we investigated FeCl3-induced epilepsy mediated differential expression of CACNA1A and GABRD in the cortical region of the rat brain. Furthermore, we investigated the effect of curcumin on the expression of both proteins. For this, epilepsy was induced by intracortical FeCl3 injection (5 μl of 100 mM). Additionally, curcumin (conc. 1000 ppm; 75 mg/kg of b.wt.; for 14 and 28 days) was administered, mixed with normal food pellets. Results obtained from EEG-MUA and Morris water maze assay demonstrate the progression of epilepsy after FeCl3 injection. Additionally, western blotting and histological studies show the downregulation of CACNA1A and GABRD during epileptogenesis. It was observed that epilepsy-associated decline in learning and memory of animals might be linked with the dysregulation of both proteins. Results also demonstrated that curcumin administration ameliorated epilepsy-associated change in expression of both CACNA1A and GABRD proteins. In conclusion, the neuroprotective effect of curcumin against iron-induced epilepsy might be accompanied by the alleviated upregulation of these channel proteins.
Collapse
Affiliation(s)
- Prince Kumar
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Deepak Sharma
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
34
|
Gawel K, Langlois M, Martins T, van der Ent W, Tiraboschi E, Jacmin M, Crawford AD, Esguerra CV. Seizing the moment: Zebrafish epilepsy models. Neurosci Biobehav Rev 2020; 116:1-20. [PMID: 32544542 DOI: 10.1016/j.neubiorev.2020.06.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/20/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
Zebrafish are now widely accepted as a valuable animal model for a number of different central nervous system (CNS) diseases. They are suitable both for elucidating the origin of these disorders and the sequence of events culminating in their onset, and for use as a high-throughput in vivo drug screening platform. The availability of powerful and effective techniques for genome manipulation allows the rapid modelling of different genetic epilepsies and of conditions with seizures as a core symptom. With this review, we seek to summarize the current knowledge about existing epilepsy/seizures models in zebrafish (both pharmacological and genetic) and compare them with equivalent rodent and human studies. New findings obtained from the zebrafish models are highlighted. We believe that this comprehensive review will highlight the value of zebrafish as a model for investigating different aspects of epilepsy and will help researchers to use these models to their full extent.
Collapse
Affiliation(s)
- Kinga Gawel
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway; Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego St. 8b, 20-090, Lublin, Poland
| | | | - Teresa Martins
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg
| | - Wietske van der Ent
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway
| | - Ettore Tiraboschi
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway; Neurophysics Group, Center for Mind/Brain Sciences, University of Trento, Piazza Manifattura 1, Building 14, 38068, Rovereto, TN, Italy
| | - Maxime Jacmin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg
| | - Alexander D Crawford
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg; Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Oslo, Norway
| | - Camila V Esguerra
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway.
| |
Collapse
|
35
|
Chow CY, Absalom N, Biggs K, King GF, Ma L. Venom-derived modulators of epilepsy-related ion channels. Biochem Pharmacol 2020; 181:114043. [PMID: 32445870 DOI: 10.1016/j.bcp.2020.114043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/18/2020] [Indexed: 12/18/2022]
Abstract
Epilepsy is characterised by spontaneous recurrent seizures that are caused by an imbalance between neuronal excitability and inhibition. Since ion channels play fundamental roles in the generation and propagation of action potentials as well as neurotransmitter release at a subset of excitatory and inhibitory synapses, their dysfunction has been linked to a wide variety of epilepsies. Indeed, these unique proteins are the major biological targets for antiepileptic drugs. Selective targeting of a specific ion channel subtype remains challenging for small molecules, due to the high level of homology among members of the same channel family. As a consequence, there is a growing trend to target ion channels with biologics. Venoms are the best known natural source of ion channel modulators, and venom peptides are increasingly recognised as potential therapeutics due to their high selectivity and potency gained through millions of years of evolutionary selection pressure. Here we describe the major ion channel families involved in the pathogenesis of various types of epilepsy, including voltage-gated Na+, K+, Ca2+ channels, Cys-loop receptors, ionotropic glutamate receptors and P2X receptors, and currently available venom-derived peptides that target these channel proteins. Although only a small number of venom peptides have successfully progressed to the clinic, there is reason to be optimistic about their development as antiepileptic drugs, notwithstanding the challenges associated with development of any class of peptide drug.
Collapse
Affiliation(s)
- Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nathan Absalom
- Brain and Mind Centre, School of Pharmacy, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW 2050, Australia
| | - Kimberley Biggs
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Linlin Ma
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia.
| |
Collapse
|
36
|
Jaworski T. Control of neuronal excitability by GSK-3beta: Epilepsy and beyond. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118745. [PMID: 32450268 DOI: 10.1016/j.bbamcr.2020.118745] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 12/22/2022]
Abstract
Glycogen synthase kinase 3beta (GSK-3β) is an enzyme with a variety of cellular functions in addition to the regulation of glycogen metabolism. In the central nervous system, different intracellular signaling pathways converge on GSK-3β through a cascade of phosphorylation events that ultimately control a broad range of neuronal functions in the development and adulthood. In mice, genetically removing or increasing GSK-3β cause distinct functional and structural neuronal phenotypes and consequently affect cognition. Precise control of GSK-3β activity is important for such processes as neuronal migration, development of neuronal morphology, synaptic plasticity, excitability, and gene expression. Altered GSK-3β activity contributes to aberrant plasticity within neuronal circuits leading to neurological, psychiatric disorders, and neurodegenerative diseases. Therapeutically targeting GSK-3β can restore the aberrant plasticity of neuronal networks at least in animal models of these diseases. Although the complete repertoire of GSK-3β neuronal substrates has not been defined, emerging evidence shows that different ion channels and their accessory proteins controlling excitability, neurotransmitter release, and synaptic transmission are regulated by GSK-3β, thereby supporting mechanisms of synaptic plasticity in cognition. Dysregulation of ion channel function by defective GSK-3β activity sustains abnormal excitability in the development of epilepsy and other GSK-3β-linked human diseases.
Collapse
Affiliation(s)
- Tomasz Jaworski
- Laboratory of Animal Models, Nencki Institute of Experimental Biology, Warsaw, Poland.
| |
Collapse
|
37
|
GABRG2 Deletion Linked to Genetic Epilepsy with Febrile Seizures Plus Affects the Expression of GABA A Receptor Subunits and Other Genes at Different Temperatures. Neuroscience 2020; 438:116-136. [PMID: 32418750 DOI: 10.1016/j.neuroscience.2020.04.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022]
Abstract
Mutations in γ-aminobutyric acid A receptor (GABAA) subunits and sodium channel genes, especially GABRG2 and SCN1A, have been reported to be associated with febrile seizures (FS) and genetic epilepsy with febrile seizures plus (GEFS+). GEFS+ is a well-known family of epileptic syndrome with autosomal dominant inheritance in children. Its most common phenotypes are febrile seizures often with accessory afebrile generalized tonic-clonic seizures, febrile seizures plus (FS+), severe epileptic encephalopathy, as well as other types of generalized or localization-related seizures. However, the pathogenesis of febrile seizures remains largely unknown. Here, we generated a GABRG2 gene knockout cell line (HT22GABRG2KO) by applying the CRISPR/Cas9-mediated genomic deletion in HT-22 mouse hippocampal neuronal cell line to explore the function of GABRG2 in vitro. With mRNA-seq, we found significant changes in the expression profiles of several epilepsy-related genes when GABRG2 was knockout, some of them showing temperature-induced changes as well. Kyoto Encyclopedia Gene and Genomic (KEGG) analysis revealed a significant alteration in the MAPK and PI3K-Akt signaling pathways. We also observed an up-regulation of the matrix metalloproteinases (MMPs) family after GABRG2 knockout. Furthermore, the significant decrease in expression of GABRA1 and CACNA1A (but not others) with an increase in temperature is a novel finding. In summary, mutations in the GABAA receptor can lead to a decrease in numbers of receptors, which may cause the impairment of GABAergic pathway signaling. This data has been the first time to reveal that GABRG2 mutations would affect the function of other genes, and based on this finding we hope this work would also provide a new direction for the research of GABRG2 in GEFS+. It also may provide a molecular basis for the severity of epilepsy, and guide the clinical medication for the treatment of the epilepsy focused on the function on GABAA receptors, which, might be a new strategy for genetic diagnosis and targeted treatment of epilepsy.
Collapse
|
38
|
Nardello R, Plicato G, Mangano GD, Gennaro E, Mangano S, Brighina F, Raieli V, Fontana A. Two distinct phenotypes, hemiplegic migraine and episodic Ataxia type 2, caused by a novel common CACNA1A variant. BMC Neurol 2020; 20:155. [PMID: 32336275 PMCID: PMC7183684 DOI: 10.1186/s12883-020-01704-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 03/27/2020] [Indexed: 12/18/2022] Open
Abstract
Background To investigate the genetic and environmental factors responsible for phenotype variability in a family carrying a novel CACNA1A missense mutation. Mutations in the CACNA1A gene were identified as responsible for at least three autosomal dominant disorders: FHM1 (Familial Hemiplegic Migraine), EA2 (Episodic Ataxia type 2), and SCA6 (Spinocerebellar Ataxia type 6). Overlapping clinical features within individuals of some families sharing the same CACNA1A mutation are not infrequent. Conversely, reports with distinct phenotypes within the same family associated with a common CACNA1A mutation are very rare. Case presentation A clinical, molecular, neuroradiological, neuropsychological, and neurophysiological study was carried out in proband and his carrier mother. The new heterozygous missense variant c.4262G > A (p.Arg1421Gln) in the CACNA1A gene was detected in the two affected family members. The proband showed a complex clinical presentation characterized by developmental delay, poor motor coordination, hemiplegic migraine attacks, behavioral dysregulation, and EEG abnormalities. The mother showed typical episodic ataxia attacks during infancy with no other comorbidities and mild cerebellar signs at present neurological evaluation. Conclusions The proband and his mother exhibit two distinct clinical phenotypes. It can be hypothesized that other unknown modifying genes and/or environmental factors may cooperate to generate the wide intrafamilial variability.
Collapse
Affiliation(s)
- Rosaria Nardello
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, Specialities "G. D'Alessandro," University of Palermo, Palermo, Italy
| | - Giorgia Plicato
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, Specialities "G. D'Alessandro," University of Palermo, Palermo, Italy
| | - Giuseppe Donato Mangano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, Specialities "G. D'Alessandro," University of Palermo, Palermo, Italy
| | - Elena Gennaro
- UOC Laboratorio di Genetica Umana, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Salvatore Mangano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, Specialities "G. D'Alessandro," University of Palermo, Palermo, Italy.
| | - Filippo Brighina
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Vincenzo Raieli
- Child Neuropsychiatry Department, Di Cristina - ARNAS Civico Hospital, Palermo, Italy
| | - Antonina Fontana
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, Specialities "G. D'Alessandro," University of Palermo, Palermo, Italy
| |
Collapse
|
39
|
Amano Y, Honda H, Sawada R, Nukada Y, Yamane M, Ikeda N, Morita O, Yamanishi Y. In silico systems for predicting chemical-induced side effects using known and potential chemical protein interactions, enabling mechanism estimation. J Toxicol Sci 2020; 45:137-149. [PMID: 32147637 DOI: 10.2131/jts.45.137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
In silico models for predicting chemical-induced side effects have become increasingly important for the development of pharmaceuticals and functional food products. However, existing predictive models have difficulty in estimating the mechanisms of side effects in terms of molecular targets or they do not cover the wide range of pharmacological targets. In the present study, we constructed novel in silico models to predict chemical-induced side effects and estimate the underlying mechanisms with high general versatility by integrating the comprehensive prediction of potential chemical-protein interactions (CPIs) with machine learning. First, the potential CPIs were comprehensively estimated by chemometrics based on the known CPI data (1,179,848 interactions involving 3,905 proteins and 824,143 chemicals). Second, the predictive models for 61 side effects in the cardiovascular system (CVS), gastrointestinal system (GIS), and central nervous system (CNS) were constructed by sparsity-induced classifiers based on the known and potential CPI data. The cross validation experiments showed that the proposed CPI-based models had a higher or comparable performance than the traditional chemical structure-based models. Moreover, our enrichment analysis indicated that the highly weighted proteins derived from predictive models could be involved in the corresponding functions of the side effects. For example, in CVS, the carcinogenesis-related pathways (e.g., prostate cancer, PI3K-Akt signal pathway), which were recently reported to be involved in cardiovascular side effects, were enriched. Therefore, our predictive models are biologically valid and would be useful for predicting side effects and novel potential underlying mechanisms of chemical-induced side effects.
Collapse
Affiliation(s)
- Yuto Amano
- R&D Safety Science Research, Kao Corporation
| | | | - Ryusuke Sawada
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology
| | - Yuko Nukada
- R&D Safety Science Research, Kao Corporation
| | | | | | | | | |
Collapse
|
40
|
Zhang L, Wen Y, Zhang Q, Chen Y, Wang J, Shi K, Du L, Bao X. CACNA1A Gene Variants in Eight Chinese Patients With a Wide Range of Phenotypes. Front Pediatr 2020; 8:577544. [PMID: 33425808 PMCID: PMC7793878 DOI: 10.3389/fped.2020.577544] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/20/2020] [Indexed: 11/13/2022] Open
Abstract
Background: The CACNA1A gene encodes the voltage-dependent P/Q-type calcium channel subunit alpha-1A, which is widely expressed throughout the CNS. The biological roles of the P/Q channel are diverse and the phenotypic spectrum caused by CACNA1A mutations is wide. The aim of this study is to demonstrate its phenotypic diversity and analyze the genotype-phenotype correlations in a cohort of Chinese patients. Methods: Patients with hemiplegic migraine, cerebellar ataxia, developmental delay, or epilepsy without known causes were tested by trios whole-exome sequencing. Patients with pathogenic CACNA1A gene variants were recruited. The clinical information of the patients was collected, and the association between the genotype and the phenotype was investigated. Results: In total, eight patients (six females and two males) were found to have CACNA1A gene variants. All the variants were de novo including six missense variants and one frameshift variant. Four de novo missense variants were found in five patients located in the S4, S5, or S6 transmembrane segments of Domain II and III (p.R1352Q, p.G701V, p.A713T, p.V1393M). All of them were correlated with severe phenotypes, including three with sporadic hemiplegic migraine type 1 and epilepsy, and two with developmental and epileptic encephalopathy. The other two missense variants, p.Y62C and p.F1814L, located in the cytoplasmic side of the N-terminus and C-terminus, respectively. The variant p.Y62C was associated with severe hemiconvulsion-hemiplegia-epilepsy syndrome, and p.F1814L was associated with relatively mild phenotypes. All the missense variants were speculated as gain-of-function (GOF) mutations. The only frameshift variant, p.Q681Rfs*100, a lose-of-function (LOF) mutation, was found in a patient with episodic ataxia type 2. Meanwhile, all the patients had developmental delay ranging from mild to severe, as well as cerebellar ataxia including one with congenital ataxia, one with episodic ataxia, and six with non-progressive ataxia. Conclusions: CACNA1A variants could lead to a wide spectrum of neurological disorders including epileptic or non-epileptic paroxysmal events, cerebellar ataxia, and developmental delay. The variants could be both GOF and LOF mutations. There appeared to be some correlations between genotypes and phenotypes.
Collapse
Affiliation(s)
- Linxia Zhang
- Department of Pediatric, Peking University First Hospital, Beijing, China.,Department of Neurology, Children's Hospital of Shanxi, Taiyuan, China
| | - Yongxin Wen
- Department of Pediatric, Peking University First Hospital, Beijing, China
| | - Qingping Zhang
- Department of Pediatric, Peking University First Hospital, Beijing, China
| | - Yan Chen
- Department of Pediatric, Peking University First Hospital, Beijing, China
| | - Jiaping Wang
- Department of Pediatric, Peking University First Hospital, Beijing, China
| | - Kaili Shi
- Department of Neurology, Children's Hospital of Shanxi, Taiyuan, China
| | - Lijun Du
- Department of Neurology, Children's Hospital of Shanxi, Taiyuan, China
| | - Xinhua Bao
- Department of Pediatric, Peking University First Hospital, Beijing, China
| |
Collapse
|
41
|
Gawel K, Turski WA, van der Ent W, Mathai BJ, Kirstein-Smardzewska KJ, Simonsen A, Esguerra CV. Phenotypic Characterization of Larval Zebrafish (Danio rerio) with Partial Knockdown of the cacna1a Gene. Mol Neurobiol 2019; 57:1904-1916. [PMID: 31875924 PMCID: PMC7118054 DOI: 10.1007/s12035-019-01860-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/15/2019] [Indexed: 12/18/2022]
Abstract
The CACNA1A gene encodes the pore-forming α1 subunit of voltage-gated P/Q type Ca2+ channels (Cav2.1). Mutations in this gene, among others, have been described in patients and rodents suffering from absence seizures and episodic ataxia type 2 with/without concomitant seizures. In this study, we aimed for the first time to assess phenotypic and behavioral alterations in larval zebrafish with partial cacna1aa knockdown, placing special emphasis on changes in epileptiform-like electrographic discharges in larval brains. Whole-mount in situ hybridization analysis revealed expression of cacna1aa in the optic tectum and medulla oblongata of larval zebrafish at 4 and 5 days post-fertilization. Next, microinjection of two antisense morpholino oligomers (individually or in combination) targeting all splice variants of cacna1aa into fertilized zebrafish eggs resulted in dose-dependent mortality and decreased or absent touch response. Over 90% knockdown of cacna1aa on protein level induced epileptiform-like discharges in the optic tectum of larval zebrafish brains. Incubation of morphants with antiseizure drugs (sodium valproate, ethosuximide, lamotrigine, topiramate) significantly decreased the number and, in some cases, cumulative duration of epileptiform-like discharges. In this context, sodium valproate seemed to be the least effective. Carbamazepine did not affect the number and duration of epileptiform-like discharges. Altogether, our data indicate that cacna1aa loss-of-function zebrafish may be considered a new model of absence epilepsy and may prove useful both for the investigation of Cacna1a-mediated epileptogenesis and for in vivo drug screening.
Collapse
Affiliation(s)
- Kinga Gawel
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, Faculty of Medicine, University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway.,Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego St. 8b, 20-090, Lublin, Poland
| | - Waldemar A Turski
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego St. 8b, 20-090, Lublin, Poland
| | - Wietske van der Ent
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, Faculty of Medicine, University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway
| | - Benan J Mathai
- Faculty of Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway
| | - Karolina J Kirstein-Smardzewska
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, Faculty of Medicine, University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway
| | - Anne Simonsen
- Faculty of Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 1112 Blindern, 0317, Oslo, Norway
| | - Camila V Esguerra
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway, Faculty of Medicine, University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway. .,School of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Sælandsvei 24, 0371, Oslo, Norway.
| |
Collapse
|
42
|
Calcium signalling in mammalian cell lines expressing wild type and mutant human α1-Antitrypsin. Sci Rep 2019; 9:17293. [PMID: 31754242 PMCID: PMC6872872 DOI: 10.1038/s41598-019-53535-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/28/2019] [Indexed: 11/08/2022] Open
Abstract
A possible role for calcium signalling in the autosomal dominant form of dementia, familial encephalopathy with neuroserpin inclusion bodies (FENIB), has been proposed, which may point towards a mechanism by which cells could sense and respond to the accumulation of mutant serpin polymers in the endoplasmic reticulum (ER). We therefore explored possible defects in Ca2+-signalling, which may contribute to the pathology associated with another serpinopathy, α1-antitrypsin (AAT) deficiency. Using CHO K1 cell lines stably expressing a wild type human AAT (MAAT) and a disease-causing polymer-forming variant (ZAAT) and the truncated variant (NHK AAT), we measured basal intracellular free Ca2+, its responses to thapsigargin (TG), an ER Ca2+-ATPase blocker, and store-operated Ca2+-entry (SOCE). Our fura2 based Ca2+ measurements detected no differences between these 3 parameters in cell lines expressing MAAT and cell lines expressing ZAAT and NHK AAT mutants. Thus, in our cell-based models of α1-antitrypsin (AAT) deficiency, unlike the case for FENIB, we were unable to detect defects in calcium signalling.
Collapse
|
43
|
Hixson KM, Cogswell M, Brooks-Kayal AR, Russek SJ. Evidence for a non-canonical JAK/STAT signaling pathway in the synthesis of the brain's major ion channels and neurotransmitter receptors. BMC Genomics 2019; 20:677. [PMID: 31455240 PMCID: PMC6712773 DOI: 10.1186/s12864-019-6033-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/15/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is a major signaling molecule that the brain uses to control a vast network of intracellular cascades fundamental to properties of learning and memory, and cognition. While much is known about BDNF signaling in the healthy nervous system where it controls the mitogen activated protein kinase (MAPK) and cyclic-AMP pathways, less is known about its role in multiple brain disorders where it contributes to the dysregulated neuroplasticity seen in epilepsy and traumatic brain injury (TBI). We previously found that neurons respond to prolonged BDNF exposure (both in vivo (in models of epilepsy and TBI) and in vitro (in BDNF treated primary neuronal cultures)) by activating the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) signaling pathway. This pathway is best known for its association with inflammatory cytokines in non-neuronal cells. RESULTS Here, using deep RNA-sequencing of neurons exposed to BDNF in the presence and absence of well characterized JAK/STAT inhibitors, and without non-neuronal cells, we determine the BDNF transcriptome that is specifically regulated by agents that inhibit JAK/STAT signaling. Surprisingly, the BDNF-induced JAK/STAT transcriptome contains ion channels and neurotransmitter receptors coming from all the major classes expressed in the brain, along with key modulators of synaptic plasticity, neurogenesis, and axonal remodeling. Analysis of this dataset has revealed a unique non-canonical mechanism of JAK/STATs in neurons as differential gene expression mediated by STAT3 is not solely dependent upon phosphorylation at residue 705 and may involve a BDNF-induced interaction of STAT3 with Heterochromatin Protein 1 alpha (HP1α). CONCLUSIONS These findings suggest that the neuronal BDNF-induced JAK/STAT pathway involves more than STAT3 phosphorylation at 705, providing the first evidence for a non-canonical mechanism that may involve HP1α. Our analysis reveals that JAK/STAT signaling regulates many of the genes associated with epilepsy syndromes where BDNF levels are markedly elevated. Uncovering the mechanism of this novel form of BDNF signaling in the brain may provide a new direction for epilepsy therapeutics and open a window into the complex mechanisms of STAT3 transcriptional regulation in neurological disease.
Collapse
Affiliation(s)
- Kathryn M. Hixson
- Laboratory of Translational Epilepsy, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine (BUSM), Boston, USA
- Graduate Program for Neuroscience (GPN), Boston University (BU), Boston, USA
| | - Meaghan Cogswell
- Laboratory of Translational Epilepsy, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine (BUSM), Boston, USA
| | - Amy R. Brooks-Kayal
- Department of Pediatric Neurology, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Shelley J. Russek
- Laboratory of Translational Epilepsy, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine (BUSM), Boston, USA
- Graduate Program for Neuroscience (GPN), Boston University (BU), Boston, USA
- Department of Biology, Boston University (BU), Boston, USA
| |
Collapse
|
44
|
De Gusmao CM, Silveira-Moriyama L. Paroxysmal movement disorders - practical update on diagnosis and management. Expert Rev Neurother 2019; 19:807-822. [PMID: 31353980 DOI: 10.1080/14737175.2019.1648211] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Paroxysmal dyskinesias and episodic ataxias are often caused by mutations in genes related to cell membrane and synaptic function. Despite the exponential increase in publications of genetically confirmed cases, management remains largely clinical based on non-systematic evidence. Areas covered: The authors provide a historical and clinical review of the main types of paroxysmal dyskinesias and episodic ataxias, with recommendations for diagnosis and management of patients suffering from these conditions. Expert opinion: After secondary paroxysmal dyskinesias, the most common paroxysmal movement disorders are likely to be PRRT2-associated paroxysmal kinesigenic dyskinesias, which respond well to small doses of carbamazepine, and episodic ataxia type 2, which often responds to acetazolamide. Familial paroxysmal non-kinesigenic dyskinesias are largely caused by mutations in PNKD and have poor response to therapy but improve with age. Exercise-induced dyskinesias are genetically heterogeneous, caused by disorders of glucose transport, mitochondrial function, dopaminergic pathways or neurodegenerative conditions amongst others. GNAO1 and ADCY5 mutations can also cause paroxysmal movement disorders, often in the context of ongoing motor symptoms. Although a therapeutic trial is justified for classic cases and in limited resource settings, genetic testing may help direct initial or rescue therapy. Deep brain stimulation may be an option for severe cases.
Collapse
Affiliation(s)
- Claudio M De Gusmao
- Department of Neurology, Harvard Medical School, Boston Children's Hospital , Boston , MA , USA.,Department of Neurology, Universidade Estadual de Campinas (UNICAMP) , São Paulo , Brazil
| | - Laura Silveira-Moriyama
- Department of Neurology, Universidade Estadual de Campinas (UNICAMP) , São Paulo , Brazil.,Education Unit, UCL Institute of Neurology, University College London , London , UK.,Department of Neurology, Hospital Bairral, Fundação Espírita Américo Bairral , Itapira , Brazil
| |
Collapse
|
45
|
Pan R, Qi X, Wang F, Chong Y, Li X, Chen Q. Correlations of Calcium Voltage-Gated Channel Subunit Alpha1 A (CACNA1A) Gene Polymorphisms with Benign Paroxysmal Positional Vertigo. Med Sci Monit 2019; 25:946-951. [PMID: 30710491 PMCID: PMC6368824 DOI: 10.12659/msm.912359] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background The aim of this study was to investigate the correlations of calcium voltage-gated channel subunit alpha1 A (CACNA1A) gene polymorphisms with benign paroxysmal positional vertigo (BPPV). Material/Methods A total of 120 BPPV patients and 60 healthy controls were enrolled according to the diagnostic criteria in the Guideline of Diagnosis and Treatment of Benign Paroxysmal Positional Vertigo (2017). Clinical and biochemical data were collected, the rs2074880 (T/G) polymorphisms in the CACNA1A gene were detected using TaqMan-MGB probe method, and the correlations of BPPV with predisposing factors were analyzed through logistic analysis. Results The BPPV group had higher levels of cholesterol and uric acid than in the control group (p<0.05). The cholesterol and uric acid levels were positively correlated with BPPV (p<0.05) [odds ratio (OR)=2.298 (1.252–4.350), 95% confidence interval (95% CI)=1.123 (0.987–1.987)]. The distribution frequency of TT genotype was higher than that of GG genotype (χ2=9.907, p=0.002, OR=0.279, 95% CI=0.123–0.633). In the BPPV group, cholesterol and uric acid levels of TT genotype were elevated compared with those in GG genotype (p<0.05). Conclusions The onset of BPPV is related to the increased levels of cholesterol and uric acid, as well as the dominant homozygous mutation of rs2074880 (T/G) in the CACNA1A gene.
Collapse
Affiliation(s)
- Ruichun Pan
- Southern Medical University, Guangzhou, Guangdong, China (mainland).,Department of Neurology, Baotou Central Hospital, Baotou, Inner Mongolia, China (mainland)
| | - Xiaokun Qi
- Southern Medical University, Guangzhou, Guangdong, China (mainland).,Department of Neurology, Navy General Hospital, Beijing, China (mainland)
| | - Fei Wang
- Department of Neurology, First Affiliated Hospital of Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China (mainland)
| | - Yi Chong
- Department of Neurology, Baotou Central Hospital, Baotou, Inner Mongolia, China (mainland)
| | - Xia Li
- Department of Neurology, Baotou Central Hospital, Baotou, Inner Mongolia, China (mainland)
| | - Qiang Chen
- Department of Neurology, Baotou Central Hospital, Baotou, Inner Mongolia, China (mainland)
| |
Collapse
|
46
|
Voltage-Dependent Calcium Channels, Calcium Binding Proteins, and Their Interaction in the Pathological Process of Epilepsy. Int J Mol Sci 2018; 19:ijms19092735. [PMID: 30213136 PMCID: PMC6164075 DOI: 10.3390/ijms19092735] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 01/08/2023] Open
Abstract
As an important second messenger, the calcium ion (Ca2+) plays a vital role in normal brain function and in the pathophysiological process of different neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), and epilepsy. Ca2+ takes part in the regulation of neuronal excitability, and the imbalance of intracellular Ca2+ is a trigger factor for the occurrence of epilepsy. Several anti-epileptic drugs target voltage-dependent calcium channels (VDCCs). Intracellular Ca2+ levels are mainly controlled by VDCCs located in the plasma membrane, the calcium-binding proteins (CBPs) inside the cytoplasm, calcium channels located on the intracellular calcium store (particular the endoplasmic reticulum/sarcoplasmic reticulum), and the Ca2+-pumps located in the plasma membrane and intracellular calcium store. So far, while many studies have established the relationship between calcium control factors and epilepsy, the mechanism of various Ca2+ regulatory factors in epileptogenesis is still unknown. In this paper, we reviewed the function, distribution, and alteration of VDCCs and CBPs in the central nervous system in the pathological process of epilepsy. The interaction of VDCCs with CBPs in the pathological process of epilepsy was also summarized. We hope this review can provide some clues for better understanding the mechanism of epileptogenesis, and for the development of new anti-epileptic drugs targeting on VDCCs and CBPs.
Collapse
|
47
|
Indelicato E, Nachbauer W, Karner E, Eigentler A, Wagner M, Unterberger I, Poewe W, Delazer M, Boesch S. The neuropsychiatric phenotype in CACNA1A mutations: a retrospective single center study and review of the literature. Eur J Neurol 2018; 26:66-e7. [PMID: 30063100 DOI: 10.1111/ene.13765] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/24/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND PURPOSE CACNA1A encodes the α1 subunit of the neuronal calcium channel P/Q. CACNA1A mutations underlie three allelic disorders: familial hemiplegic migraine type 1 (FHM1), episodic ataxia type 2 (EA2) and spinocerebellar ataxia type 6 (SCA6). A clear-cut genotype-phenotype correlation is often lacking since clinical manifestations may overlap. Several case reports have described cognitive and behavioral features in CACNA1A disorders, but studies in larger case series are lacking. METHODS Genetically confirmed CACNA1A cases were retrieved from the database of the ataxia outpatient clinic of the Department of Neurology at Innsbruck Medical University. Clinical charts and neuropsychological test results were retrospectively analyzed. In addition, a review of the literature including only genetically confirmed cases was performed. RESULTS Forty-four CACNA1A cases were identified in our database. Delayed psychomotor milestones and poor school performance were described in seven (four FHM1, three EA2) and eight (three FHM1, five EA2) patients, respectively. Psychiatric comorbidities were diagnosed in eight patients (two FHM1, six EA2). Neuropsychological testing was available for 23 patients (11 FHM1, 10 EA2, two SCA6). Various cognitive deficits were documented in 21 cases (all patients except one SCA6). Impairments were predominantly seen in figural memory, visuoconstructive abilities and verbal fluency. In the literature, an early psychomotor delay is described in several children with EA2 and FHM1, whilst reports of cognitive and psychiatric findings from adult cases are scarce. CONCLUSIONS Neuropsychiatric manifestations are common in episodic CACNA1A disorders. In the case of otherwise unexplained developmental delay and a positive family history, CACNA1A mutations should be considered in the differential diagnosis.
Collapse
Affiliation(s)
- E Indelicato
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - W Nachbauer
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - E Karner
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - A Eigentler
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - M Wagner
- Department of Neuroradiology, Innsbruck Medical University, Innsbruck, Austria
| | - I Unterberger
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - W Poewe
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - M Delazer
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - S Boesch
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
48
|
Feng H, Khalil S, Neubig RR, Sidiropoulos C. A mechanistic review on GNAO1-associated movement disorder. Neurobiol Dis 2018; 116:131-141. [PMID: 29758257 DOI: 10.1016/j.nbd.2018.05.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/28/2018] [Accepted: 05/10/2018] [Indexed: 02/07/2023] Open
Abstract
Mutations in the GNAO1 gene cause a complex constellation of neurological disorders including epilepsy, developmental delay, and movement disorders. GNAO1 encodes Gαo, the α subunit of Go, a member of the Gi/o family of heterotrimeric G protein signal transducers. Go is the most abundant membrane protein in the mammalian central nervous system and plays major roles in synaptic neurotransmission and neurodevelopment. GNAO1 mutations were first reported in early infantile epileptic encephalopathy 17 (EIEE17) but are also associated with a more common syndrome termed neurodevelopmental disorder with involuntary movements (NEDIM). Here we review a mechanistic model in which loss-of-function (LOF) GNAO1 alleles cause epilepsy and gain-of-function (GOF) alleles are primarily associated with movement disorders. We also develop a signaling framework related to cyclic AMP (cAMP), synaptic vesicle release, and neural development and discuss gene mutations perturbing those mechanisms in a range of genetic movement disorders. Finally, we analyze clinical reports of patients carrying GNAO1 mutations with respect to their symptom onset and discuss pharmacological/surgical treatments in the context of our mechanistic model.
Collapse
Affiliation(s)
- Huijie Feng
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Suad Khalil
- Department of Neurology & Ophthalmology, Michigan State University, East Lansing, MI 48824, USA
| | - Richard R Neubig
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| | - Christos Sidiropoulos
- Department of Neurology & Ophthalmology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
49
|
Hesse AN, Bevilacqua J, Shankar K, Reddi HV. Retrospective genotype-phenotype analysis in a 305 patient cohort referred for testing of a targeted epilepsy panel. Epilepsy Res 2018; 144:53-61. [PMID: 29778030 DOI: 10.1016/j.eplepsyres.2018.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/09/2018] [Accepted: 05/12/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Epilepsy is a diverse neurological condition with extreme genetic and phenotypic heterogeneity. The introduction of next-generation sequencing into the clinical laboratory has made it possible to investigate hundreds of associated genes simultaneously for a patient, even in the absence of a clearly defined syndrome. This has resulted in the detection of rare and novel mutations at a rate well beyond our ability to characterize their effects. This retrospective study reviews genotype data in the context of available phenotypic information on 305 patients spanning the epileptic spectrum to identify established and novel patterns of correlation. METHODS Our epilepsy panel comprising 377 genes was used to sequence 305 patients referred for genetic testing. Qualifying variants were annotated with phenotypic data obtained from either the test requisition form or supporting clinical documentation. Observed phenotypes were compared with established phenotypes in OMIM, published literature and the ILAEs 2010 report on genetic testing to assess congruity with known gene aberrations. RESULTS We identified a number of novel and recognized genetic variants consistent with established epileptic phenotypes. Forty-one pathogenic or predicted deleterious variants were detected in 39 patients with accompanying clinical documentation. Twenty-five of these variants across 15 genes were novel. Furthermore, evaluation of phenotype data for 194 patients with variants of unknown significance in genes with autosomal dominant and X-linked disease inheritance elucidated potentially disease-causing variants that were not currently characterized in the literature. CONCLUSIONS Assessment of key genotype-phenotype correlations from our cohort provide insight into variant classification, as well as the importance of including ILAE recommended genes as part of minimum panel content for comprehensive epilepsy tests. Many of the reported VUSs are likely genuine pathogenic variants driving the observed phenotypes, but not enough evidence is available for assertive classifications. Similar studies will provide more utility via mounting independent genotype-phenotype data from unrelated patients. The possible outcome would be a better molecular diagnostic product, with fewer indeterminate reports containing only VUSs.
Collapse
Affiliation(s)
- Andrew N Hesse
- Transgenomic Inc, 5 Science Park, New Haven, CT, 06511, USA
| | | | - Kritika Shankar
- Transgenomic Inc, 5 Science Park, New Haven, CT, 06511, USA.
| | - Honey V Reddi
- Transgenomic Inc, 5 Science Park, New Haven, CT, 06511, USA.
| |
Collapse
|
50
|
Peng J, Wang Y, He F, Chen C, Wu LW, Yang LF, Ma YP, Zhang W, Shi ZQ, Chen C, Xia K, Guo H, Yin F, Pang N. Novel West syndrome candidate genes in a Chinese cohort. CNS Neurosci Ther 2018; 24:1196-1206. [PMID: 29667327 DOI: 10.1111/cns.12860] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/15/2018] [Accepted: 03/18/2018] [Indexed: 12/19/2022] Open
Abstract
AIMS West syndrome (WS) is a classic form of early infantile epileptic encephalopathy (EIEE) characterized by tonic spasms with clustering, arrest of psychomotor development, and hypsarrhythmia on electroencephalography. Genetic defects play a critical role in the pathology of WS, and 54 EIEE genes have been identified till date. This study was designed to uncover new candidate genes for West syndrome. METHODS In this study, we recruited 56 Chinese families with WS of unknown etiology. Whole exome sequencing (WES) was performed to identify Mendelian inheritance rare or novel variants. The association between candidate genes and WS was analyzed from many aspects, including recurrent genes in patients, predicted variant effect on genes, human tolerance to deficient genes, gene expression in the nervous system, coexpression with EIEE genes, mutual interaction with known EIEE proteins, genes related to ion channel or fragile X mental retardation protein function, and mouse models with manifestation of seizures. Genes with supporting evidence from those aspects were defined as highlight candidate genes. RESULTS Whole exome sequencing identified 112 candidate variants in 89 genes. Among the candidate genes, 33 were autosomal dominant, 22 were autosomal recessive, and 34 were X-linked. Complex bioinformatic analysis revealed 17 highlight candidate genes: ATP2A2, CD99L2, CLCN6, CYFIP1, CYFIP2, GNB1, GPT2, HUWE1, KMT2D, MYO18A, NOS3, RYR1, RYR2, RYR3, TAF1, TECTA, and UBA1. The majority of highlight candidate genes are calcium-signaling pathway and mental retardation genes. CONCLUSIONS This is the first WES study of Chinese WS patients with unknown etiology. This combination of phenotypic and genomic data will enable further testing to elucidate mechanisms underlying the pathogenesis of WS.
Collapse
Affiliation(s)
- Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Ying Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Chen Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Li-Wen Wu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Li-Fen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Ping Ma
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Wen Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Zi-Qing Shi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Chen
- Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Kun Xia
- Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Hui Guo
- Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Nan Pang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|