1
|
Wang K, Nilsson M, Angelini M, Olcese R, Elinder F, Pantazis A. A rich conformational palette underlies human Ca V2.1-channel availability. Nat Commun 2025; 16:3815. [PMID: 40268901 PMCID: PMC12019389 DOI: 10.1038/s41467-025-58884-2] [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: 09/17/2024] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
Depolarization-evoked opening of CaV2.1 (P/Q-type) Ca2+-channels triggers neurotransmitter release, while voltage-dependent inactivation (VDI) limits channel availability to open, contributing to synaptic plasticity. The mechanism of CaV2.1 response to voltage is unclear. Using voltage-clamp fluorometry and kinetic modeling, we optically track and physically characterize the structural dynamics of the four CaV2.1 voltage-sensor domains (VSDs). The VSDs are differentially sensitive to voltage changes, both brief and long-lived. VSD-I seems to directly drive opening and convert between two modes of function, associated with VDI. VSD-II is apparently voltage-insensitive. VSD-III and VSD-IV sense more negative voltages and undergo voltage-dependent conversion uncorrelated with VDI. Auxiliary β-subunits regulate VSD-I-to-pore coupling and VSD conversion kinetics. Hence, the central role of CaV2.1 channels in synaptic release, and their contribution to plasticity, memory formation and learning, can arise from the voltage-dependent conformational changes of VSD-I.
Collapse
Affiliation(s)
- Kaiqian Wang
- Division of Cell and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Michelle Nilsson
- Division of Cell and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Marina Angelini
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Riccardo Olcese
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Fredrik Elinder
- Division of Cell and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Science for Life Laboratory, Linköping University, Linköping, Sweden
| | - Antonios Pantazis
- Division of Cell and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
- Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden.
| |
Collapse
|
2
|
Chabriat H. Is migraine a common manifestation of CADASIL? Arguments Pros. J Headache Pain 2025; 26:64. [PMID: 40169951 PMCID: PMC11963349 DOI: 10.1186/s10194-025-01980-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/14/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Migraine with aura (MA) is a hallmark feature of CADASIL, a hereditary small-vessel disease caused by NOTCH3 mutations. While MA is prevalent in CADASIL, its underlying mechanisms remain unclear, and the links observed can be questioned or debated. This study examined the prevalence, clinical characteristics, and pathophysiology of MA in patients with CADASIL. METHODS Clinical and experimental data were reviewed to assess MA prevalence, aura characteristics, sex differences, and pathophysiological insights from CADASIL models to confirm the indisputable pathophysiological links between migraine and aura and this unique genetic model of cerebral small vessel disease. RESULTS MA was 10-20 times more prevalent in patients with CADASIL than in the general population, with frequent atypical and prolonged auras. The altered sex distribution and delayed onset suggest disease-specific mechanisms. Experimental data also revealed heightened susceptibility to cortical spreading depression (CSD) in preclinical CADASIL models, linked to greater fragility in maintaining cortical ionic homeostasis. CONCLUSION The high prevalence and distinct MA features, as well as the data obtained at the preclinical level, strongly support a causal relationship mediated by neurovascular dysfunction in CADASIL. Accumulating data in this condition sheds new light on the much-debated relationship between migraine and cerebrovascular diseases.
Collapse
Affiliation(s)
- Hugues Chabriat
- Centre Neurovasculaire Translationnel-CERVCO - Département de Neurologie, APHP and Université Paris- Cité, Paris, France.
- INSERM U1127, Paris Brain Institute, 75013, Paris, France.
| |
Collapse
|
3
|
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
|
4
|
Hussey JW, DeMarco E, DiSilvestre D, Brohus M, Busuioc AO, Iversen ED, Jensen HH, Nyegaard M, Overgaard MT, Ben-Johny M, Dick IE. Voltage Gated Calcium Channel Dysregulation May Contribute to Neurological Symptoms in Calmodulinopathies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626503. [PMID: 39677635 PMCID: PMC11642847 DOI: 10.1101/2024.12.02.626503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Calmodulinopathies are caused by mutations in calmodulin (CaM), and result in debilitating cardiac arrythmias such as long-QT syndrome (LQTS) and catecholaminergic polymorphic ventricular tachycardia (CPVT). In addition, many patients exhibit neurological comorbidities, including developmental delay and autism spectrum disorder. Until now, most work into these mutations has focused on cardiac effects, identifying impairment of Ca 2+ /CaM-dependent inactivation (CDI) of Ca V 1.2 channels as a major pathogenic mechanism. However, the impact of these mutations on neurological function has yet to be fully explored. CaM regulation of voltage-gated calcium channels (VGCCs) is a critical element of neuronal function, implicating multiple VGCC subtypes in the neurological pathogenesis of calmodulinopathies. Here, we explore the potential for pathological CaM variants to impair the Ca 2+ /CaM-dependent regulation of Ca V 1.3 and Ca V 2.1, both essential for neuronal function. We find that mutations in CaM can impair the CDI of Ca V 1.3 and reduce the Ca 2+ -dependent facilitation (CDF) of Ca V 2.1 channels. We find that mutations associated with significant neurological symptoms exhibit marked effects on Ca V 1.3 CDI, with overlapping but distinct impacts on Ca V 2.1 CDF. Moreover, while the majority of CaM variants demonstrated the ability to bind the IQ region of each channel, distinct differences were noted between Ca V 1.3 and Ca V 2.1, demonstrating distinct CaM interactions across the two channel subtypes. Further, C-domain CaM variants display a reduced ability to sense Ca 2+ when in complex with the Ca V IQ domains, explaining the Ca 2+ /CaM regulation deficits. Overall, these results support the possibility that disrupted Ca 2+ /CaM regulation of VGCCs may contribute to neurological pathogenesis of calmodulinopathies.
Collapse
|
5
|
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
|
6
|
Wang K, Nilsson M, Angelini M, Olcese R, Elinder F, Pantazis A. A Rich Conformational Palette Underlies Human Ca V2.1-Channel Availability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615501. [PMID: 39464068 PMCID: PMC11507735 DOI: 10.1101/2024.09.27.615501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Depolarization-evoked opening of CaV2.1 (P/Q-type) Ca2+-channels triggers neurotransmitter release, while voltage-dependent inactivation (VDI) limits channel availability to open, contributing to synaptic plasticity. The mechanism of CaV2.1 response to voltage is unclear. Using voltage-clamp fluorometry and kinetic modeling, we optically tracked and physically characterized the structural dynamics of the four CaV2.1 voltage-sensor domains (VSDs). VSD-I seems to directly drive opening and convert between two modes of function, associated with VDI. VSD-II is apparently voltage-insensitive. VSD-III and VSD-IV sense more negative voltages and undergo voltage-dependent conversion uncorrelated with VDI. Auxiliary β -subunits regulate VSD-I-to-pore coupling and VSD conversion kinetics. CaV2.1 VSDs are differentially sensitive to voltage changes brief and long-lived. Specifically the voltage-dependent conformational changes of VSD-I are linked to synaptic release and plasticity.
Collapse
Affiliation(s)
- Kaiqian Wang
- Division of Cell and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University; SE-581 85 Linköping, Sweden
| | - Michelle Nilsson
- Division of Cell and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University; SE-581 85 Linköping, Sweden
| | - Marina Angelini
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Riccardo Olcese
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Fredrik Elinder
- Division of Cell and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University; SE-581 85 Linköping, Sweden
- Science for Life Laboratory, Linköping University; SE-581 85 Linköping, Sweden
| | - Antonios Pantazis
- Division of Cell and Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University; SE-581 85 Linköping, Sweden
- Wallenberg Center for Molecular Medicine, Linköping University; SE-581 85 Linköping, Sweden
| |
Collapse
|
7
|
Turner C, Campbell L, Fung R, Desai S, Oyenubi A, Cayabyab F, Huntsman RJ. Treatment of CACNA1A Encephalopathy and Cerebral Edema with Magnesium and Dexamethasone. Can J Neurol Sci 2024:1-4. [PMID: 39157864 DOI: 10.1017/cjn.2024.52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Pathogenic CACNA1A mutations can result in paroxysmal attacks of encephalopathy, hemiplegia and cerebral edema. We report two patients with CACNA1A-associated encephalopathy, hemiplegia and contralateral hemispheric cerebral edema treated successfully with intravenous magnesium sulfate and dexamethasone. One patient met the clinical criteria for familial hemiplegic migraine. There is a paucity of guidance in the literature on how to manage these patients. Despite some discrepancies in the treatment protocols in our two cases, they indicate that magnesium and dexamethasone could be part of the treatment algorithm for these patients. Further research to delineate appropriate dosing and duration of therapy is needed.
Collapse
Affiliation(s)
- Cailey Turner
- Postgraduate Medical Education Program, Department of Pediatrics, University of Saskatchewan, Saskatoon, SK, Canada
| | - Lauren Campbell
- Postgraduate Medical Education Program, Department of Pediatrics, University of Saskatchewan, Regina, SK, Canada
| | - Ryan Fung
- Department of Pediatric Pharmacy, Saskatchewan Health Authority, Saskatoon, SK, Canada
| | - Srirupa Desai
- Department of Radiology, University of Saskatchewan, Regina, SK, Canada
| | - Abimbola Oyenubi
- Department of Pediatrics, University of Saskatchewan, Regina, SK, Canada
| | - Francisco Cayabyab
- Department of Surgery, University of Saskatchewan, Saskatoon, SK, Canada
| | - Richard James Huntsman
- Division of Pediatric Neurology, Department of Pediatrics, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
8
|
Li Z, Cong Y, Wu T, Wang T, Lou X, Yang X, Yan N. Structural basis for different ω-agatoxin IVA sensitivities of the P-type and Q-type Ca v2.1 channels. Cell Res 2024; 34:455-457. [PMID: 38443561 PMCID: PMC11143261 DOI: 10.1038/s41422-024-00940-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/14/2024] [Indexed: 03/07/2024] Open
Affiliation(s)
- Zhangqiang Li
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| | - Ye Cong
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Tong Wu
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Tongtong Wang
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xinyao Lou
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xinyu Yang
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
- Institute of Bio-Architecture and Bio-Interactions, Shenzhen Medical Academy of Research and Translation, Shenzhen, Guangdong, China.
| |
Collapse
|
9
|
Li S, Zhao S, Sinson JC, Bajic A, Rosenfeld JA, Neeley MB, Pena M, Worley KC, Burrage LC, Weisz-Hubshman M, Ketkar S, Craigen WJ, Clark GD, Lalani S, Bacino CA, Machol K, Chao HT, Potocki L, Emrick L, Sheppard J, Nguyen MTT, Khoramnia A, Hernandez PP, Nagamani SC, Liu Z, Eng CM, Lee B, Liu P. The clinical utility and diagnostic implementation of human subject cell transdifferentiation followed by RNA sequencing. Am J Hum Genet 2024; 111:841-862. [PMID: 38593811 PMCID: PMC11080285 DOI: 10.1016/j.ajhg.2024.03.007] [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: 10/04/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
RNA sequencing (RNA-seq) has recently been used in translational research settings to facilitate diagnoses of Mendelian disorders. A significant obstacle for clinical laboratories in adopting RNA-seq is the low or absent expression of a significant number of disease-associated genes/transcripts in clinically accessible samples. As this is especially problematic in neurological diseases, we developed a clinical diagnostic approach that enhanced the detection and evaluation of tissue-specific genes/transcripts through fibroblast-to-neuron cell transdifferentiation. The approach is designed specifically to suit clinical implementation, emphasizing simplicity, cost effectiveness, turnaround time, and reproducibility. For clinical validation, we generated induced neurons (iNeurons) from 71 individuals with primary neurological phenotypes recruited to the Undiagnosed Diseases Network. The overall diagnostic yield was 25.4%. Over a quarter of the diagnostic findings benefited from transdifferentiation and could not be achieved by fibroblast RNA-seq alone. This iNeuron transcriptomic approach can be effectively integrated into diagnostic whole-transcriptome evaluation of individuals with genetic disorders.
Collapse
Affiliation(s)
- Shenglan Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Sen Zhao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jefferson C Sinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Aleksandar Bajic
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Advanced Technology Cores, Baylor College of Medicine, Houston, TX, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Matthew B Neeley
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA
| | - Mezthly Pena
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Kim C Worley
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Lindsay C Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Monika Weisz-Hubshman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Shamika Ketkar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - William J Craigen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Gary D Clark
- Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Seema Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Carlos A Bacino
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Keren Machol
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Hsiao-Tuan Chao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Cain Pediatric Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, TX, USA
| | - Lorraine Potocki
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Lisa Emrick
- Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Jennifer Sheppard
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - My T T Nguyen
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Anahita Khoramnia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | - Sandesh Cs Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Christine M Eng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Baylor Genetics, Houston, TX, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Pengfei Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Baylor Genetics, Houston, TX, USA.
| |
Collapse
|
10
|
Mascherpa M, Fichera A, Orabona R, Recupero D, Borroni B, Odicino FE, Prefumo F. Inherited episodic ataxia type 2 in pregnancy: A case report. Int J Gynaecol Obstet 2024; 165:387-389. [PMID: 38059694 DOI: 10.1002/ijgo.15292] [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: 09/11/2023] [Revised: 11/15/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023]
Abstract
SynopsisAn optimal materno‐neonatal outcome was obtained in a pregnancy with inherited episodic ataxia through a multidisciplinary team handling timing and mode of delivery.
Collapse
Affiliation(s)
- Margaret Mascherpa
- Department of Obstetrics and Gynecology, ASST-Spedali Civili, Brescia, Italy
| | - Anna Fichera
- Department of Obstetrics and Gynecology, ASST-Spedali Civili, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Rossana Orabona
- Department of Obstetrics and Gynecology, ASST-Spedali Civili, Brescia, Italy
| | - Daniela Recupero
- Department of Anesthesiology 1, Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Franco Edoardo Odicino
- Department of Obstetrics and Gynecology, ASST-Spedali Civili, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Federico Prefumo
- Obstetrics and Gynecology Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| |
Collapse
|
11
|
Mueller A, Lam I, Kishor K, Lee RK, Bhattacharya S. Secondary glaucoma: Toward interventions based on molecular underpinnings. WIREs Mech Dis 2024; 16:e1628. [PMID: 37669762 DOI: 10.1002/wsbm.1628] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 09/07/2023]
Abstract
Glaucoma is a heterogeneous group of progressive diseases that leads to irreversible blindness. Secondary glaucoma refers to glaucoma caused by a known underlying condition. Pseudoexfoliation and pigment dispersion syndromes are common causes of secondary glaucoma. Their respective deposits may obstruct the trabecular meshwork, leading to aqueous humor outflow resistance, ocular hypertension, and optic neuropathy. There are no disease-specific interventions available for either. Pseudoexfoliation syndrome is characterized by fibrillar deposits (pseudoexfoliative material) on anterior segment structures. Over a decade of multiomics analyses taken together with the current knowledge on pseudoexfoliative glaucoma warrant a re-think of mechanistic possibilities. We propose that the presence of nucleation centers (e.g., vitamin D binding protein), crosslinking enzymes (e.g., transglutaminase 2), aberrant extracellular matrix, flawed endocytosis, and abnormal aqueous-blood barrier contribute to the formation of proteolytically resistant pseudoexfoliative material. Pigment dispersion syndrome is characterized by abnormal iridolenticular contact that disrupts iris pigment epithelium and liberates melanin granules. Iris melanogenesis is aberrant in this condition. Cytotoxic melanogenesis intermediates leak out of melanosomes and cause iris melanocyte and pigment epithelium cell death. Targeting melanogenesis can likely decrease the risk of pigmentary glaucoma. Skin and melanoma research provides insights into potential therapeutics. We propose that specific prostanoid agonists and fenofibrates may reduce melanogenesis by inhibiting cholesterol internalization and de novo synthesis. Additionally, melatonin is a potent melanogenesis suppressor, antioxidant, and hypotensive agent, rendering it a valuable agent for pigmentary glaucoma. In pseudoexfoliative glaucoma, where environmental insults drive pseudoexfoliative material formation, melatonin's antioxidant and hypotensive properties may offer adjunct therapeutic benefits. This article is categorized under: Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Anna Mueller
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Isabel Lam
- Idaho College of Osteopathic Medicine, Meridian, Idaho, USA
| | - Krishna Kishor
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
- Miami Integrative Metabolomics Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Richard K Lee
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
- Miami Integrative Metabolomics Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sanjoy Bhattacharya
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
- Miami Integrative Metabolomics Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
12
|
Lyu H, Boßelmann CM, Johannesen KM, Koko M, Ortigoza-Escobar JD, Aguilera-Albesa S, Garcia-Navas Núñez D, Linnankivi T, Gaily E, van Ruiten HJA, Richardson R, Betzler C, Horvath G, Brilstra E, Geerdink N, Orsucci D, Tessa A, Gardella E, Fleszar Z, Schöls L, Lerche H, Møller RS, Liu Y. Clinical and electrophysiological features of SCN8A variants causing episodic or chronic ataxia. EBioMedicine 2023; 98:104855. [PMID: 38251463 PMCID: PMC10628346 DOI: 10.1016/j.ebiom.2023.104855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Variants in SCN8A are associated with a spectrum of epilepsies and neurodevelopmental disorders. Ataxia as a predominant symptom of SCN8A variation has not been well studied. We set out to investigate disease mechanisms and genotype-phenotype correlations of SCN8A-related ataxia. METHODS We collected genetic and electro-clinical data of ten individuals from nine unrelated families carrying novel SCN8A variants associated with chronic progressive or episodic ataxia. Electrophysiological characterizations of these variants were performed in ND7/23 cells and cultured neurons. FINDINGS Variants associated with chronic progressive ataxia either decreased Na+ current densities and shifted activation curves towards more depolarized potentials (p.Asn995Asp, p.Lys1498Glu and p.Trp1266Cys) or resulted in a premature stop codon (p.Trp937Ter). Three variants (p.Arg847Gln and biallelic p.Arg191Trp/p.Asp1525Tyr) were associated with episodic ataxia causing loss-of-function by decreasing Na+ current densities or a hyperpolarizing shift of the inactivation curve. Two additional episodic ataxia-associated variants caused mixed gain- and loss-of function effects in ND7/23 cells and were further examined in primary murine hippocampal neuronal cultures. Neuronal firing in excitatory neurons was increased by p.Arg1629His, but decreased by p.Glu1201Lys. Neuronal firing in inhibitory neurons was decreased for both variants. No functional effect was observed for p.Arg1913Trp. In four individuals, treatment with sodium channel blockers exacerbated symptoms. INTERPRETATION We identified episodic or chronic ataxia as predominant phenotypes caused by variants in SCN8A. Genotype-phenotype correlations revealed a more pronounced loss-of-function effect for variants causing chronic ataxia. Sodium channel blockers should be avoided under these conditions. FUNDING BMBF, DFG, the Italian Ministry of Health, University of Tuebingen.
Collapse
Affiliation(s)
- Hang Lyu
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Christian M Boßelmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Katrine M Johannesen
- Department of Clinical Genetics, University Hospital of Copenhagen, Rigshospitalet, Copenhagen, Denmark; Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre (Member of the ERN EpiCARE), Dianalund, Denmark
| | - Mahmoud Koko
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Juan Dario Ortigoza-Escobar
- Movement Disorders Unit, Institut de Recerca Sant Joan de Déu, CIBERER-ISCIII and European Reference Network for Rare Neurological Diseases (ERN-RND), Barcelona, Spain
| | - Sergio Aguilera-Albesa
- Pediatric Neurology Unit, Department of Pediatrics, Hospital Universitario de Navarra, Pamplona, Spain; Navarrabiomed-Fundación Miguel Servet, Pamplona, Spain
| | | | - Tarja Linnankivi
- Department of Pediatric Neurology, New Children's Hospital and Pediatric Research Center, Epilepsia Helsinki, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Eija Gaily
- Department of Pediatric Neurology, New Children's Hospital and Pediatric Research Center, Epilepsia Helsinki, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Henriette J A van Ruiten
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, Great North Children's Hospital, Newcastle upon Tyne, UK
| | - Ruth Richardson
- Northern Genetics Service, The Newcastle Upon Tyne Hospitals NHS Foundation Trust, UK
| | - Cornelia Betzler
- Institute for Rehabilitation, Transition and Palliation, Paracelsus Medical University, Salzburg, Austria; Specialist Center for Paediatric Neurology, Neuro-Rehabilitation and Epileptology, Schön Klinik Vogtareuth, Germany
| | - Gabriella Horvath
- Adult Metabolic Diseases Clinic, BC Children's Hospital, Vancouver, Canada
| | - Eva Brilstra
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Niels Geerdink
- Department of Pediatrics, Rijnstate Hospital, Arnhem, the Netherlands
| | | | | | - Elena Gardella
- Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre (Member of the ERN EpiCARE), Dianalund, Denmark; Department of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Denmark
| | - Zofia Fleszar
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Ludger Schöls
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Centre (Member of the ERN EpiCARE), Dianalund, Denmark; Department of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Denmark
| | - Yuanyuan Liu
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
13
|
Kapuganti RS, Alone DP. Current understanding of genetics and epigenetics in pseudoexfoliation syndrome and glaucoma. Mol Aspects Med 2023; 94:101214. [PMID: 37729850 DOI: 10.1016/j.mam.2023.101214] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/22/2023]
Abstract
Pseudoexfoliation is a complex, progressive, and systemic age-related disorder. The early stage of deposition of extracellular fibrillar material on ocular and extraocular tissues is termed as pseudoexfoliation syndrome (PEXS). The severe advanced stage is known as pseudoexfoliation glaucoma (PEXG), which involves increased intraocular pressure and optic nerve damage. Through genome-wide association and candidate gene studies, PEX has been associated with numerous genetic risk variants in various gene loci. However, the genetic basis of the disease fails to explain certain features of PEX pathology, such as the progressive nature of the disease, asymmetric ocular manifestation, age-related onset, and only a subset of PEXS individuals developing PEXG. Increasing evidence shows an interplay of genetic and epigenetic factors in the pathology of complex, multifactorial diseases. In this review, we have discussed the genetic basis of the disease and the emerging contribution of epigenetic regulations in PEX pathogenesis, focusing on DNA methylation and non-coding RNAs. Aberrant methylation patterns, histone modifications, and post-transcriptional regulation by microRNAs lead to aberrant gene expression changes. We have reviewed these aberrant epigenetic changes in PEX pathology and their effect on molecular pathways associated with PEX. We have further discussed some possible genetic/epigenetic-based diagnoses and therapeutics for PEX. Although studies to understand the role of epigenetic regulations in PEX are just emerging, epigenetic modifications contribute significantly to PEX pathogenesis and may pave the way for better and targeted therapeutics.
Collapse
Affiliation(s)
- Ramani Shyam Kapuganti
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha, 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai, 400094, India
| | - Debasmita Pankaj Alone
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, P.O. Bhimpur-Padanpur, Jatni, Khurda, Odisha, 752050, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai, 400094, India.
| |
Collapse
|
14
|
Dahimene S, Page KM, Nieto-Rostro M, Pratt WS, Dolphin AC. The Interplay Between Splicing of Two Exon Combinations Differentially Affects Membrane Targeting and Function of Human Ca V2.2. FUNCTION 2023; 5:zqad060. [PMID: 38020068 PMCID: PMC10666670 DOI: 10.1093/function/zqad060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
N-type calcium channels (CaV2.2) are predominantly localized in presynaptic terminals, and are particularly important for pain transmission in the spinal cord. Furthermore, they have multiple isoforms, conferred by alternatively spliced or cassette exons, which are differentially expressed. Here, we have examined alternatively spliced exon47 variants that encode a long or short C-terminus in human CaV2.2. In the Ensembl database, all short exon47-containing transcripts were associated with the absence of exon18a, therefore, we also examined the effect of inclusion or absence of exon18a, combinatorially with the exon47 splice variants. We found that long exon47, only in the additional presence of exon18a, results in CaV2.2 currents that have a 3.6-fold greater maximum conductance than the other three combinations. In contrast, cell-surface expression of CaV2.2 in both tsA-201 cells and hippocampal neurons is increased ∼4-fold by long exon47, relative to short exon47, in either the presence or the absence of exon18a. This surprising discrepancy between trafficking and function indicates that cell-surface expression is enhanced by long exon47, independently of exon18a. However, in the presence of long exon47, exon18a mediates an additional permissive effect on CaV2.2 gating. We also investigated the single-nucleotide polymorphism in exon47 that has been linked to schizophrenia and Parkinson's disease, which we found is only non-synonymous in the short exon47 C-terminal isoform, resulting in two minor alleles. This study highlights the importance of investigating the combinatorial effects of exon inclusion, rather than each in isolation, in order to increase our understanding of calcium channel function.
Collapse
Affiliation(s)
- Shehrazade Dahimene
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Karen M Page
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Manuela Nieto-Rostro
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Wendy S Pratt
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| |
Collapse
|
15
|
Pilotto F, Douthwaite C, Diab R, Ye X, Al Qassab Z, Tietje C, Mounassir M, Odriozola A, Thapa A, Buijsen RAM, Lagache S, Uldry AC, Heller M, Müller S, van Roon-Mom WMC, Zuber B, Liebscher S, Saxena S. Early molecular layer interneuron hyperactivity triggers Purkinje neuron degeneration in SCA1. Neuron 2023; 111:2523-2543.e10. [PMID: 37321222 PMCID: PMC10431915 DOI: 10.1016/j.neuron.2023.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/17/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Toxic proteinaceous deposits and alterations in excitability and activity levels characterize vulnerable neuronal populations in neurodegenerative diseases. Using in vivo two-photon imaging in behaving spinocerebellar ataxia type 1 (Sca1) mice, wherein Purkinje neurons (PNs) degenerate, we identify an inhibitory circuit element (molecular layer interneurons [MLINs]) that becomes prematurely hyperexcitable, compromising sensorimotor signals in the cerebellum at early stages. Mutant MLINs express abnormally elevated parvalbumin, harbor high excitatory-to-inhibitory synaptic density, and display more numerous synaptic connections on PNs, indicating an excitation/inhibition imbalance. Chemogenetic inhibition of hyperexcitable MLINs normalizes parvalbumin expression and restores calcium signaling in Sca1 PNs. Chronic inhibition of mutant MLINs delayed PN degeneration, reduced pathology, and ameliorated motor deficits in Sca1 mice. Conserved proteomic signature of Sca1 MLINs, shared with human SCA1 interneurons, involved the higher expression of FRRS1L, implicated in AMPA receptor trafficking. We thus propose that circuit-level deficits upstream of PNs are one of the main disease triggers in SCA1.
Collapse
Affiliation(s)
- Federica Pilotto
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Christopher Douthwaite
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Rim Diab
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - XiaoQian Ye
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Zahraa Al Qassab
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Christoph Tietje
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Martinsried, Germany
| | - Meriem Mounassir
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Martinsried, Germany
| | | | - Aishwarya Thapa
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Ronald A M Buijsen
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Sophie Lagache
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Anne-Christine Uldry
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Stefan Müller
- Flow Cytometry and Cell sorting, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Benoît Zuber
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Sabine Liebscher
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians University Munich, Martinsried, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; University Hospital Cologne, Deptartment of Neurology, Cologne, Germany.
| | - Smita Saxena
- Department of Neurology, Inselspital University Hospital, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
16
|
Garrido JJ. Contribution of Axon Initial Segment Structure and Channels to Brain Pathology. Cells 2023; 12:cells12081210. [PMID: 37190119 DOI: 10.3390/cells12081210] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Brain channelopathies are a group of neurological disorders that result from genetic mutations affecting ion channels in the brain. Ion channels are specialized proteins that play a crucial role in the electrical activity of nerve cells by controlling the flow of ions such as sodium, potassium, and calcium. When these channels are not functioning properly, they can cause a wide range of neurological symptoms such as seizures, movement disorders, and cognitive impairment. In this context, the axon initial segment (AIS) is the site of action potential initiation in most neurons. This region is characterized by a high density of voltage-gated sodium channels (VGSCs), which are responsible for the rapid depolarization that occurs when the neuron is stimulated. The AIS is also enriched in other ion channels, such as potassium channels, that play a role in shaping the action potential waveform and determining the firing frequency of the neuron. In addition to ion channels, the AIS contains a complex cytoskeletal structure that helps to anchor the channels in place and regulate their function. Therefore, alterations in this complex structure of ion channels, scaffold proteins, and specialized cytoskeleton may also cause brain channelopathies not necessarily associated with ion channel mutations. This review will focus on how the AISs structure, plasticity, and composition alterations may generate changes in action potentials and neuronal dysfunction leading to brain diseases. AIS function alterations may be the consequence of voltage-gated ion channel mutations, but also may be due to ligand-activated channels and receptors and AIS structural and membrane proteins that support the function of voltage-gated ion channels.
Collapse
Affiliation(s)
- Juan José Garrido
- Instituto Cajal, CSIC, 28002 Madrid, Spain
- Alzheimer's Disease and Other Degenerative Dementias, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28002 Madrid, Spain
| |
Collapse
|
17
|
Leyva-Leyva M, Sandoval A, Morales-Lázaro SL, Corzo-López A, Felix R, González-Ramírez R. Identification of Dp140 and α1-syntrophin as novel molecular interactors of the neuronal Ca V2.1 channel. Pflugers Arch 2023; 475:595-606. [PMID: 36964781 DOI: 10.1007/s00424-023-02803-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/21/2023] [Accepted: 03/06/2023] [Indexed: 03/26/2023]
Abstract
The primary function of dystrophin is to form a link between the cytoskeleton and the extracellular matrix. In addition to this crucial structural function, dystrophin also plays an essential role in clustering and organizing several signaling proteins, including ion channels. Proteomic analysis of the whole rodent brain has stressed the role of some components of the dystrophin-associated glycoprotein complex (DGC) as potential interacting proteins of the voltage-gated Ca2+ channels of the CaV2 subfamily. The interaction of CaV2 with signaling and scaffolding proteins, such as the DGC components, may influence their function, stability, and location in neurons. This work aims to study the interaction between dystrophin and CaV2.1. Our immunoprecipitation data showed the presence of a complex formed by CaV2.1, CaVα2δ-1, CaVβ4e, Dp140, and α1-syntrophin in the brain. Furthermore, proximity ligation assays (PLA) showed that CaV2.1 and CaVα2δ-1 interact with dystrophin in the hippocampus and cerebellum. Notably, Dp140 and α1-syntrophin increase CaV2.1 protein stability, half-life, permanence in the plasma membrane, and current density through recombinant CaV2.1 channels. Therefore, we have identified the Dp140 and α1-syntrophin as novel interaction partners of CaV2.1 channels in the mammalian brain. Consistent with previous findings, our work provides evidence of the role of DGC in anchoring and clustering CaV channels in a macromolecular complex.
Collapse
Affiliation(s)
- Margarita Leyva-Leyva
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Universidad Nacional Autónoma de México (UNAM), Ciudad de Mexico, México
| | - Alejandro Sandoval
- School of Medicine FES Iztacala, National Autonomous University of México (UNAM), Tlalnepantla, Mexico
| | - Sara Luz Morales-Lázaro
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - Alejandra Corzo-López
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Ricardo Felix
- Department of Cell Biology, Centre for Research and Advanced Studies (Cinvestav), Mexico City, Mexico.
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, "Dr. Manuel Gea González" General Hospital, Mexico City, Mexico.
| |
Collapse
|
18
|
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
|
19
|
Folacci M, Estaran S, Ménard C, Bertaud A, Rousset M, Roussel J, Thibaud JB, Vignes M, Chavanieu A, Charnet P, Cens T. Functional Characterization of Four Known Cav2.1 Variants Associated with Neurodevelopmental Disorders. MEMBRANES 2023; 13:96. [PMID: 36676903 PMCID: PMC9864995 DOI: 10.3390/membranes13010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 06/17/2023]
Abstract
Cav2.1 channels are expressed throughout the brain and are the predominant Ca2+ channels in the Purkinje cells. These cerebellar neurons fire spontaneously, and Cav2.1 channels are involved in the regular pacemaking activity. The loss of precision of the firing pattern of Purkinje cells leads to ataxia, a disorder characterized by poor balance and difficulties in performing coordinated movements. In this study, we aimed at characterizing functional and structural consequences of four variations (p.A405T in I-II loop and p.R1359W, p.R1667W and p.S1799L in IIIS4, IVS4, and IVS6 helices, respectively) identified in patients exhibiting a wide spectrum of disorders including ataxia symptoms. Functional analysis using two major Cav2.1 splice variants (Cav2.1+e47 and Cav2.1-e47) in Xenopus laevis oocytes, revealed a lack of effect upon A405T substitution and a significant loss-of-function caused by R1359W, whereas R1667W and S1799L caused both channel gain-of-function and loss-of-function, in a splice variant-dependent manner. Structural analysis revealed the loss of interactions with S1, S2, and S3 helices upon R1359W and R1667W substitutions, but a lack of obvious structural changes with S1799L. Computational modeling suggests that biophysical changes induced by Cav2.1 pathogenic mutations might affect action potential frequency in Purkinje cells.
Collapse
|
20
|
Zong P, Yue L. Regulation of Presynaptic Calcium Channels. ADVANCES IN NEUROBIOLOGY 2023; 33:171-202. [PMID: 37615867 DOI: 10.1007/978-3-031-34229-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Voltage-gated calcium channels (VGCCs), especially Cav2.1 and Cav2.2, are the major mediators of Ca2+ influx at the presynaptic membrane in response to neuron excitation, thereby exerting a predominant control on synaptic transmission. To guarantee the timely and precise release of neurotransmitters at synapses, the activity of presynaptic VGCCs is tightly regulated by a variety of factors, including auxiliary subunits, membrane potential, G protein-coupled receptors (GPCRs), calmodulin (CaM), Ca2+-binding proteins (CaBP), protein kinases, various interacting proteins, alternative splicing events, and genetic variations.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA.
| |
Collapse
|
21
|
Morgenstern TJ, Nirwan N, Hernández-Ochoa EO, Bibollet H, Choudhury P, Laloudakis YD, Ben Johny M, Bannister RA, Schneider MF, Minor DL, Colecraft HM. Selective posttranslational inhibition of Ca Vβ 1-associated voltage-dependent calcium channels with a functionalized nanobody. Nat Commun 2022; 13:7556. [PMID: 36494348 PMCID: PMC9734117 DOI: 10.1038/s41467-022-35025-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
Ca2+ influx through high-voltage-activated calcium channels (HVACCs) controls diverse cellular functions. A critical feature enabling a singular signal, Ca2+ influx, to mediate disparate functions is diversity of HVACC pore-forming α1 and auxiliary CaVβ1-CaVβ4 subunits. Selective CaVα1 blockers have enabled deciphering their unique physiological roles. By contrast, the capacity to post-translationally inhibit HVACCs based on CaVβ isoform is non-existent. Conventional gene knockout/shRNA approaches do not adequately address this deficit owing to subunit reshuffling and partially overlapping functions of CaVβ isoforms. Here, we identify a nanobody (nb.E8) that selectively binds CaVβ1 SH3 domain and inhibits CaVβ1-associated HVACCs by reducing channel surface density, decreasing open probability, and speeding inactivation. Functionalizing nb.E8 with Nedd4L HECT domain yielded Chisel-1 which eliminated current through CaVβ1-reconstituted CaV1/CaV2 and native CaV1.1 channels in skeletal muscle, strongly suppressed depolarization-evoked Ca2+ influx and excitation-transcription coupling in hippocampal neurons, but was inert against CaVβ2-associated CaV1.2 in cardiomyocytes. The results introduce an original method for probing distinctive functions of ion channel auxiliary subunit isoforms, reveal additional dimensions of CaVβ1 signaling in neurons, and describe a genetically-encoded HVACC inhibitor with unique properties.
Collapse
Affiliation(s)
- Travis J. Morgenstern
- grid.239585.00000 0001 2285 2675Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY USA
| | - Neha Nirwan
- grid.266102.10000 0001 2297 6811Cardiovascular Research Institute, University of California, San Francisco, CA USA
| | - Erick O. Hernández-Ochoa
- grid.411024.20000 0001 2175 4264Department of Biochemistry and Biology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Hugo Bibollet
- grid.411024.20000 0001 2175 4264Department of Biochemistry and Biology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Papiya Choudhury
- grid.239585.00000 0001 2285 2675Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY USA
| | - Yianni D. Laloudakis
- grid.239585.00000 0001 2285 2675Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY USA
| | - Manu Ben Johny
- grid.239585.00000 0001 2285 2675Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY USA
| | - Roger A. Bannister
- grid.411024.20000 0001 2175 4264Department of Biochemistry and Biology, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Department of Pathology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Martin F. Schneider
- grid.411024.20000 0001 2175 4264Department of Biochemistry and Biology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Daniel L. Minor
- grid.266102.10000 0001 2297 6811Cardiovascular Research Institute, University of California, San Francisco, CA USA ,grid.266102.10000 0001 2297 6811Department of Biochemistry and Biophysics, University of California, San Francisco, CA USA ,grid.266102.10000 0001 2297 6811Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA USA ,grid.266102.10000 0001 2297 6811California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA USA ,grid.266102.10000 0001 2297 6811Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA USA ,grid.184769.50000 0001 2231 4551Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| | - Henry M. Colecraft
- grid.239585.00000 0001 2285 2675Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, NY USA ,grid.239585.00000 0001 2285 2675Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY USA
| |
Collapse
|
22
|
Johnson SL, Tsou WL, Prifti MV, Harris AL, Todi SV. A survey of protein interactions and posttranslational modifications that influence the polyglutamine diseases. Front Mol Neurosci 2022; 15:974167. [PMID: 36187346 PMCID: PMC9515312 DOI: 10.3389/fnmol.2022.974167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/27/2022] [Indexed: 01/20/2023] Open
Abstract
The presence and aggregation of misfolded proteins has deleterious effects in the nervous system. Among the various diseases caused by misfolded proteins is the family of the polyglutamine (polyQ) disorders. This family comprises nine members, all stemming from the same mutation—the abnormal elongation of a polyQ repeat in nine different proteins—which causes protein misfolding and aggregation, cellular dysfunction and disease. While it is the same type of mutation that causes them, each disease is distinct: it is influenced by regions and domains that surround the polyQ repeat; by proteins with which they interact; and by posttranslational modifications they receive. Here, we overview the role of non-polyQ regions that control the pathogenicity of the expanded polyQ repeat. We begin by introducing each polyQ disease, the genes affected, and the symptoms experienced by patients. Subsequently, we provide a survey of protein-protein interactions and posttranslational modifications that regulate polyQ toxicity. We conclude by discussing shared processes and pathways that bring some of the polyQ diseases together and may serve as common therapeutic entry points for this family of incurable disorders.
Collapse
Affiliation(s)
- Sean L. Johnson
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Matthew V. Prifti
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Autumn L. Harris
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
- Maximizing Access to Research Careers (MARC) Program, Wayne State University, Detroit, MI, United States
| | - Sokol V. Todi
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
- Maximizing Access to Research Careers (MARC) Program, Wayne State University, Detroit, MI, United States
- Department of Neurology, Wayne State University, Detroit, MI, United States
- *Correspondence: Sokol V. Todi,
| |
Collapse
|
23
|
Yuan X, Zheng Y, Gao F, Sun W, Wang Z, Zhao G. Case Report: A Novel CACNA1A Mutation Caused Flunarizine-Responsive Type 2 Episodic Ataxia and Hemiplegic Migraine With Abnormal MRI of Cerebral White Matter. Front Neurol 2022; 13:899813. [PMID: 35677330 PMCID: PMC9168224 DOI: 10.3389/fneur.2022.899813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/21/2022] [Indexed: 11/23/2022] Open
Abstract
Episodic ataxia type 2 (EA2) is one autosomal-dominant neurological disorder characterized by debilitating attacks of ataxia. It is mainly caused by loss-of-function mutations of the CACNA1A gene, which encodes the pore-forming α1A subunit of Cav2.1 (P/Q type voltage-gated calcium channel). Sporadic hemiplegic migraine (SHM) is another rare disease involving CACNA1A variants, which seldom coexists with EA2. Here we report a novel pathogenic mutation in CACNA1A (c.3836dupA, exon 23, p.Y1279X) of a 16-year-old female, who complained about paroxysmal dizziness, headache, and unsteady gait. Her brain MRI revealed a slightly atrophic cerebellum and numerous asymptomatic hyperintense lesions of the cerebral white matter. The diagnosis of EA2 combined with SHM was made. Administration of 5-mg flunarizine once daily at night effectively reduced the attacks and attenuated her symptoms for a month.
Collapse
Affiliation(s)
| | | | | | | | | | - Guiping Zhao
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
24
|
Generation of induced pluripotent stem cell lines carrying monoallelic (UCSFi001-A-60) or biallelic (UCSFi001-A-61; UCSFi001-A-62) frameshift variants in CACNA1A using CRISPR/Cas9. Stem Cell Res 2022; 61:102730. [DOI: 10.1016/j.scr.2022.102730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/25/2022] [Indexed: 11/20/2022] Open
|
25
|
Luan H, Zhang L, Zhang S, Zhang M. Next-generation sequencing identified a novel CACNA1A I1379F variant in a familial hemiplegic migraine type 1 pedigree: A case report. Medicine (Baltimore) 2021; 100:e28141. [PMID: 34941060 PMCID: PMC8702007 DOI: 10.1097/md.0000000000028141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/17/2021] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Familial hemiplegic migraine (FHM) is a rare, autosomal dominant migraine with aura. CACNA1A encodes the α1A subunit of P/Q-type voltage-gated calcium channels, and its mutations have been associated with a wide spectrum of episodic and chronic neurological disorders, including FHM type 1 (FHM1). PATIENT CONCERNS A Chinese girl and some of her relatives who presented with hemiplegia with or without migraine were found to carry a novel heterozygous missense variant, I1379F, in CACNA1A by whole-exome sequencing. The variant consegregated with the disease and was predicted to be pathogenic. DIAGNOSIS The patient was diagnosed with FHM1 clinically and genetically. INTERVENTIONS Prophylactic therapy with flunarizine 5 mg daily was prescribed to the patient. OUTCOMES Therapy with flunarizine was terminated after a few weeks. The intensity of the attacks was the same as before. LESSONS This case indicates that FHM should be considered when a patient manifests with episodic hemiplegia without migraine. In addition, genetic testing is an indispensable method to identify atypical attacks of hemiplegic migraine.
Collapse
Affiliation(s)
- Huiyan Luan
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Lei Zhang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Sijin Zhang
- Department of Pediatrics, the Second Hospital of Jilin University, Changchun, Jilin, China
| | - Meng Zhang
- Department of Pediatrics, the Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
26
|
Hofer NT, Pinggera A, Nikonishyna YV, Tuluc P, Fritz EM, Obermair GJ, Striessnig J. Stabilization of negative activation voltages of Cav1.3 L-Type Ca 2+-channels by alternative splicing. Channels (Austin) 2021; 15:38-52. [PMID: 33380256 PMCID: PMC7781618 DOI: 10.1080/19336950.2020.1859260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 11/30/2020] [Indexed: 11/30/2022] Open
Abstract
-->Low voltage-activated Cav1.3 L-type Ca2+-channels are key regulators of neuronal excitability controlling neuronal development and different types of learning and memory. Their physiological functions are enabled by their negative activation voltage-range, which allows Cav1.3 to be active at subthreshold voltages. Alternative splicing in the C-terminus of their pore-forming α1-subunits gives rise to C-terminal long (Cav1.3L) and short (Cav1.3S) splice variants allowing Cav1.3S to activate at even more negative voltages than Cav1.3L. We discovered that inclusion of exons 8b, 11, and 32 in Cav1.3S further shifts activation (-3 to -4 mV) and inactivation (-4 to -6 mV) to more negative voltages as revealed by functional characterization in tsA-201 cells. We found transcripts of these exons in mouse chromaffin cells, the cochlea, and the brain. Our data further suggest that Cav1.3-containing exons 11 and 32 constitute a significant part of native channels in the brain. We therefore investigated the effect of these splice variants on human disease variants. Splicing did not prevent the gating defects of the previously reported human pathogenic variant S652L, which further shifted the voltage-dependence of activation of exon 11-containing channels by more than -12 mV. In contrast, we found no evidence for gating changes of the CACNA1D missense variant R498L, located in exon 11, which has recently been identified in a patient with an epileptic syndrome. Our data demonstrate that alternative splicing outside the C-terminus involving exons 11 and 32 contributes to channel fine-tuning by stabilizing negative activation and inactivation gating properties of wild-type and mutant Cav1.3 channels.
Collapse
Affiliation(s)
- Nadja T. Hofer
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Alexandra Pinggera
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Yuliia V. Nikonishyna
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Eva M. Fritz
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Gerald J. Obermair
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
- Division Physiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| |
Collapse
|
27
|
The complexities of CACNA1A in clinical neurogenetics. J Neurol 2021; 269:3094-3108. [PMID: 34806130 DOI: 10.1007/s00415-021-10897-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/25/2022]
Abstract
Variants in CACNA1A are classically related to episodic ataxia type 2, familial hemiplegic migraine type 1, and spinocerebellar ataxia type 6. Over the years, CACNA1A has been associated with a broader spectrum of phenotypes. Targeted analysis and unbiased sequencing of CACNA1A result not only in clear molecular diagnoses, but also in large numbers of variants of uncertain significance (VUS), or likely pathogenic variants with a phenotype that does not directly match the CACNA1A spectrum. Over the last years, targeted and clinical exome sequencing in our center has identified 41 CACNA1A variants. Ultimately, variants were considered pathogenic or likely pathogenic in 23 cases, with most phenotypes ranging from episodic or progressive ataxia to more complex ataxia syndromes, as well as intellectual disability and epilepsy. In two cases, the causality of the variant was discarded based on non-segregation or an alternative diagnosis. In the remaining 16 cases, the variant was classified as uncertain, due to lack of opportunities for segregation analysis or uncertain association with a non-classic phenotype. Phenotypic variability and the large number of VUS make CACNA1A a challenging gene for neurogenetic diagnostics. Accessible functional read-outs are clearly needed, especially in cases with a non-classic phenotype.
Collapse
|
28
|
Novel Mutation in CACNA1A Associated with Activity-Induced Dystonia, Cervical Dystonia, and Mild Ataxia. Case Rep Neurol Med 2021; 2021:7797770. [PMID: 34395002 PMCID: PMC8356012 DOI: 10.1155/2021/7797770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/15/2021] [Accepted: 07/28/2021] [Indexed: 12/18/2022] Open
Abstract
CACNA1A encodes the pore-forming α1 subunit of the neuronal voltage-gated Cav2.1 (P/Q-type) channels, which are predominantly localized at the presynaptic terminals of the brain and cerebellar neurons and play an important role in controlling neurotransmitter release. Mutations in CACNA1A have been associated with several autosomal dominant neurologic disorders, including familial hemiplegic migraine type 1, episodic ataxia type 2 (EA2), and spinocerebellar ataxia type 6. A 37-year-old woman presented with a history of slowly progressive, activity-induced stiffness, and pain in her right leg since age 15 and cervical dystonia since age 20. She denied any right leg stiffness or pain at rest, but when she began to walk, her right foot turned in and her right leg stiffened up. She also had neck pain, stiffness, and spams. There was no family history of similar symptoms. On physical exam, her strength, tone, and reflexes were normal in all extremities at rest. There was mild head titubation and very mild past pointing on finger-to-nose testing. MRI of the brain and spinal cord was unremarkable. This patient's clinical picture was felt to be most consistent with paroxysmal kinesigenic dyskinesia, as she has attacks of dystonia that are triggered by voluntary movement, last from a few seconds to a minute, and are relieved with rest. She was trialed on carbidopa/levodopa without improvement. A dystonia panel showed two potentially pathologic mutations, one in CACNA1A and the other in PNKP, along with a variant of unknown significance in ATP7B. The mutation in CACNA1A is C2324 G < A. It is heterozygous, autosomal dominant, and computer modeling suggests pathogenicity. This mutation has not been reported previously and is likely the cause of her paroxysmal dystonia; dystonia is sometimes seen during episodes of ataxia in EA2, and CACNA1A knockout mice exhibit dystonia and cerebellar atrophy. After receiving her genetic diagnosis, the patient was trialed on acetazolamide without improvement in her dystonia symptoms. This is the second case report of a patient with cervical dystonia and cerebellar ataxia associated with a mutation in CACNA1A.
Collapse
|
29
|
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: 36] [Impact Index Per Article: 9.0] [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
|
30
|
Boscia F, Elkjaer ML, Illes Z, Kukley M. Altered Expression of Ion Channels in White Matter Lesions of Progressive Multiple Sclerosis: What Do We Know About Their Function? Front Cell Neurosci 2021; 15:685703. [PMID: 34276310 PMCID: PMC8282214 DOI: 10.3389/fncel.2021.685703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022] Open
Abstract
Despite significant advances in our understanding of the pathophysiology of multiple sclerosis (MS), knowledge about contribution of individual ion channels to axonal impairment and remyelination failure in progressive MS remains incomplete. Ion channel families play a fundamental role in maintaining white matter (WM) integrity and in regulating WM activities in axons, interstitial neurons, glia, and vascular cells. Recently, transcriptomic studies have considerably increased insight into the gene expression changes that occur in diverse WM lesions and the gene expression fingerprint of specific WM cells associated with secondary progressive MS. Here, we review the ion channel genes encoding K+, Ca2+, Na+, and Cl- channels; ryanodine receptors; TRP channels; and others that are significantly and uniquely dysregulated in active, chronic active, inactive, remyelinating WM lesions, and normal-appearing WM of secondary progressive MS brain, based on recently published bulk and single-nuclei RNA-sequencing datasets. We discuss the current state of knowledge about the corresponding ion channels and their implication in the MS brain or in experimental models of MS. This comprehensive review suggests that the intense upregulation of voltage-gated Na+ channel genes in WM lesions with ongoing tissue damage may reflect the imbalance of Na+ homeostasis that is observed in progressive MS brain, while the upregulation of a large number of voltage-gated K+ channel genes may be linked to a protective response to limit neuronal excitability. In addition, the altered chloride homeostasis, revealed by the significant downregulation of voltage-gated Cl- channels in MS lesions, may contribute to an altered inhibitory neurotransmission and increased excitability.
Collapse
Affiliation(s)
- Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Maria Louise Elkjaer
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Zsolt Illes
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Maria Kukley
- Achucarro Basque Center for Neuroscience, Leioa, Spain.,Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
31
|
Pu S, Thomas PJ. Resolving molecular contributions of ion channel noise to interspike interval variability through stochastic shielding. BIOLOGICAL CYBERNETICS 2021; 115:267-302. [PMID: 34021802 DOI: 10.1007/s00422-021-00877-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 05/04/2021] [Indexed: 06/12/2023]
Abstract
Molecular fluctuations can lead to macroscopically observable effects. The random gating of ion channels in the membrane of a nerve cell provides an important example. The contributions of independent noise sources to the variability of action potential timing have not previously been studied at the level of molecular transitions within a conductance-based model ion-state graph. Here we study a stochastic Langevin model for the Hodgkin-Huxley (HH) system based on a detailed representation of the underlying channel state Markov process, the "[Formula: see text]D model" introduced in (Pu and Thomas in Neural Computation 32(10):1775-1835, 2020). We show how to resolve the individual contributions that each transition in the ion channel graph makes to the variance of the interspike interval (ISI). We extend the mean return time (MRT) phase reduction developed in (Cao et al. in SIAM J Appl Math 80(1):422-447, 2020) to the second moment of the return time from an MRT isochron to itself. Because fixed-voltage spike detection triggers do not correspond to MRT isochrons, the inter-phase interval (IPI) variance only approximates the ISI variance. We find the IPI variance and ISI variance agree to within a few percent when both can be computed. Moreover, we prove rigorously, and show numerically, that our expression for the IPI variance is accurate in the small noise (large system size) regime; our theory is exact in the limit of small noise. By selectively including the noise associated with only those few transitions responsible for most of the ISI variance, our analysis extends the stochastic shielding (SS) paradigm (Schmandt and Galán in Phys Rev Lett 109(11):118101, 2012) from the stationary voltage clamp case to the current clamp case. We show numerically that the SS approximation has a high degree of accuracy even for larger, physiologically relevant noise levels. Finally, we demonstrate that the ISI variance is not an unambiguously defined quantity, but depends on the choice of voltage level set as the spike detection threshold. We find a small but significant increase in ISI variance, the higher the spike detection voltage, both for simulated stochastic HH data and for voltage traces recorded in in vitro experiments. In contrast, the IPI variance is invariant with respect to the choice of isochron used as a trigger for counting "spikes."
Collapse
Affiliation(s)
- Shusen Pu
- Department of Mathematics, Applied Mathematics, and Statistics, Case Western Reserve University, Cleveland, OH, USA.
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| | - Peter J Thomas
- Department of Mathematics, Applied Mathematics, and Statistics, Case Western Reserve University, Cleveland, OH, USA
- Department of Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Cognitive Science, Case Western Reserve University, Cleveland, OH, USA
- Department of Data and Computer Science, Case Western Reserve University, Cleveland, OH, USA
- Department of Electrical, Control, and Systems Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
32
|
Martínez-Monseny AF, Edo A, Casas-Alba D, Izquierdo-Serra M, Bolasell M, Conejo D, Martorell L, Muchart J, Carrera L, Ortez CI, Nascimento A, Oliva B, Fernández-Fernández JM, Serrano M. CACNA1A Mutations Causing Early Onset Ataxia: Profiling Clinical, Dysmorphic and Structural-Functional Findings. Int J Mol Sci 2021; 22:ijms22105180. [PMID: 34068417 PMCID: PMC8153625 DOI: 10.3390/ijms22105180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 01/07/2023] Open
Abstract
The CACNA1A gene encodes the pore-forming α1A subunit of the voltage-gated CaV2.1 Ca2+ channel, essential in neurotransmission, especially in Purkinje cells. Mutations in CACNA1A result in great clinical heterogeneity with progressive symptoms, paroxysmal events or both. During infancy, clinical and neuroimaging findings may be unspecific, and no dysmorphic features have been reported. We present the clinical, radiological and evolutionary features of three patients with congenital ataxia, one of them carrying a new variant. We report the structural localization of variants and their expected functional consequences. There was an improvement in cerebellar syndrome over time despite a cerebellar atrophy progression, inconsistent response to acetazolamide and positive response to methylphenidate. The patients shared distinctive facial gestalt: oval face, prominent forehead, hypertelorism, downslanting palpebral fissures and narrow nasal bridge. The two α1A affected residues are fully conserved throughout evolution and among the whole human CaV channel family. They contribute to the channel pore and the voltage sensor segment. According to structural data analysis and available functional characterization, they are expected to exert gain- (F1394L) and loss-of-function (R1664Q/R1669Q) effect, respectively. Among the CACNA1A-related phenotypes, our results suggest that non-progressive congenital ataxia is associated with developmental delay and dysmorphic features, constituting a recognizable syndromic neurodevelopmental disorder.
Collapse
Affiliation(s)
- Antonio F. Martínez-Monseny
- Department of Genetic and Molecular Medicine, Institut de Recerca, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (A.F.M.-M.); (D.C.-A.); (M.B.); (L.M.)
| | - Albert Edo
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.E.); (M.I.-S.)
| | - Dídac Casas-Alba
- Department of Genetic and Molecular Medicine, Institut de Recerca, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (A.F.M.-M.); (D.C.-A.); (M.B.); (L.M.)
| | - Mercè Izquierdo-Serra
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.E.); (M.I.-S.)
| | - Mercè Bolasell
- Department of Genetic and Molecular Medicine, Institut de Recerca, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (A.F.M.-M.); (D.C.-A.); (M.B.); (L.M.)
| | - David Conejo
- Pediatric Department, Complejo Asistencial de Burgos, 09006 Burgos, Spain;
| | - Loreto Martorell
- Department of Genetic and Molecular Medicine, Institut de Recerca, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (A.F.M.-M.); (D.C.-A.); (M.B.); (L.M.)
- U-703 Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, 08002 Barcelona, Spain; (C.I.O.); (A.N.)
| | - Jordi Muchart
- Pediatric Radiology Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Laura Carrera
- Neuropediatric Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Carlos I. Ortez
- U-703 Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, 08002 Barcelona, Spain; (C.I.O.); (A.N.)
- Neuropediatric Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Andrés Nascimento
- U-703 Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, 08002 Barcelona, Spain; (C.I.O.); (A.N.)
- Neuropediatric Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain;
| | - Baldo Oliva
- Structural Bioinformatics Lab, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain;
| | - José M. Fernández-Fernández
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.E.); (M.I.-S.)
- Correspondence: (J.M.F.-F.); (M.S.); Tel.: +34-93-3160854 (J.M.F.-F.); +34-93-253-2100 (M.S.)
| | - Mercedes Serrano
- U-703 Centre for Biomedical Research on Rare Diseases (CIBER-ER), Instituto de Salud Carlos III, 08002 Barcelona, Spain; (C.I.O.); (A.N.)
- Neuropediatric Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain;
- Correspondence: (J.M.F.-F.); (M.S.); Tel.: +34-93-3160854 (J.M.F.-F.); +34-93-253-2100 (M.S.)
| |
Collapse
|
33
|
Stubberud A, O'Connor E, Tronvik E, Houlden H, Matharu M. R1352Q CACNA1A Variant in a Patient with Sporadic Hemiplegic Migraine, Ataxia, Seizures and Cerebral Oedema: A Case Report. Case Rep Neurol 2021; 13:123-130. [PMID: 33790770 PMCID: PMC7989667 DOI: 10.1159/000512275] [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: 09/09/2020] [Accepted: 10/07/2020] [Indexed: 11/19/2022] Open
Abstract
Mutations in the CACNA1A gene show a wide range of neurological phenotypes including hemiplegic migraine, ataxia, mental retardation and epilepsy. In some cases, hemiplegic migraine attacks can be triggered by minor head trauma and culminate in encephalopathy and cerebral oedema. A 37-year-old male without a family history of complex migraine experienced hemiplegic migraine attacks from childhood. The attacks were usually triggered by minor head trauma, and on several occasions complicated with encephalopathy and cerebral oedema. Genetic testing of the proband and unaffected parents revealed a de novo heterozygous nucleotide missense mutation in exon 25 of the CACNA1A gene (c.4055G>A, p.R1352Q). The R1352Q CACNA1A variant shares the phenotype with other described CACNA1A mutations and highlights the interesting association of trauma as a precipitant for hemiplegic migraine. Subjects with early-onset sporadic hemiplegic migraine triggered by minor head injury or associated with seizures, ataxia or episodes of encephalopathy should be screened for mutations. These patients should also be advised to avoid activities that may result in head trauma, and anticonvulsants should be considered as prophylactic migraine therapy.
Collapse
Affiliation(s)
- Anker Stubberud
- Headache and Facial Pain Group, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, United Kingdom.,Department of Neuromedicine and Movement Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Emer O'Connor
- Headache and Facial Pain Group, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, United Kingdom.,Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, Queen Square, London, United Kingdom
| | - Erling Tronvik
- Department of Neuromedicine and Movement Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,Department of Neurology, St. Olavs Hospital, Trondheim, Norway
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, Queen Square, London, United Kingdom
| | - Manjit Matharu
- Headache and Facial Pain Group, UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, London, United Kingdom
| |
Collapse
|
34
|
Verriello L, Carrera P, Pauletto G, Bernardini A, Valente M, Gigli GL. Case report and ten-year follow-up of episodic ataxia type 2 due to a novel variant in CACNA1A. eNeurologicalSci 2021; 23:100334. [PMID: 33786385 PMCID: PMC7994720 DOI: 10.1016/j.ensci.2021.100334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/23/2021] [Accepted: 03/06/2021] [Indexed: 11/16/2022] Open
Affiliation(s)
- Lorenzo Verriello
- Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Paola Carrera
- Laboratory of Clinical and Molecular Biology and Unit of Genomics for Diagnosis of Genetic Diseases, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Giada Pauletto
- Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Andrea Bernardini
- Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Mariarosaria Valente
- Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy.,Department of Medicine (DAME), University of Udine, Italy
| | - Gian Luigi Gigli
- Clinical Neurology Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy.,Department of Mathematics, Informatics and Physics (DMIF), University of Udine, Italy
| |
Collapse
|
35
|
Albamonte E, Barp A, Duga V, Carraro E, Passarini A, Bergamoni S, Maggi L, Sansone VA. Sporadic Hemiplegic Migraine Type 1 and Congenital Ataxia due to a Single Amino Acid Deletion (ΔF1502) in CACNA1A: A Challenging Diagnosis. JOURNAL OF PEDIATRIC NEUROLOGY 2021. [DOI: 10.1055/s-0041-1725984] [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
AbstractMutations in the CACNA1A gene have been classically related to three neurologic disorders: hemiplegic migraine type 1 (both familiar and sporadic FHM1/SHM1), episodic ataxia type 2 (EA2), and spinocerebellar ataxia type 6 (SCA6). More recently, pathogenic variants in CACNA1A have been recognized as causative of an early-onset cerebellar syndrome consistent with the definition of congenital ataxia (CA), variably associated with paroxysmal symptoms. Early recognition of congenital ataxia is challenging because the presenting symptoms, such as hypotonia, weak deep tendon reflexes, and delayed motor milestones, are unspecific while clear signs of a cerebellar syndrome which are usually not seen before the second or third year. Here, we report on a case of nonepisodic ataxia of congenital onset and severe SHM1 where the diagnosis of congenital ataxia was made retrospectively after the identification of the ΔF1502 pathogenic variant in CACNA1A by an hemiplegic migraine multigene panel, conducted for the onset of hemiplegic migraine attacks associated with hemispheric swelling. A significant reduction in migraine attacks frequency was achieved with acetazolamide.
Collapse
Affiliation(s)
- Emilio Albamonte
- NEuroMuscular Omnicentre, The NeMO Clinical Center in Milan, University of Milan, Milan, Italy
| | - Andrea Barp
- NEuroMuscular Omnicentre, The NeMO Clinical Center in Milan, University of Milan, Milan, Italy
| | - Valentina Duga
- Child Neurology Unit, IRCCS Neurological Institute Foundation “Carlo Besta,” Milan, Italy
| | - Elena Carraro
- NEuroMuscular Omnicentre, The NeMO Clinical Center in Milan, University of Milan, Milan, Italy
| | - Alice Passarini
- Child Neurology Unit, Niguarda Ca' Granda Hospital, Milan, Italy
| | | | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, IRCCS Neurological Institute Foundation “Carlo Besta,” Milan, Italy
| | - Valeria Ada Sansone
- NEuroMuscular Omnicentre, The NeMO Clinical Center in Milan, University of Milan, Milan, Italy
| |
Collapse
|
36
|
Kowalska M, Prendecki M, Piekut T, Kozubski W, Dorszewska J. Migraine: Calcium Channels and Glia. Int J Mol Sci 2021; 22:2688. [PMID: 33799975 PMCID: PMC7962070 DOI: 10.3390/ijms22052688] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 01/03/2023] Open
Abstract
Migraine is a common neurological disease that affects about 11% of the adult population. The disease is divided into two main clinical subtypes: migraine with aura and migraine without aura. According to the neurovascular theory of migraine, the activation of the trigeminovascular system (TGVS) and the release of numerous neuropeptides, including calcitonin gene-related peptide (CGRP) are involved in headache pathogenesis. TGVS can be activated by cortical spreading depression (CSD), a phenomenon responsible for the aura. The mechanism of CSD, stemming in part from aberrant interactions between neurons and glia have been studied in models of familial hemiplegic migraine (FHM), a rare monogenic form of migraine with aura. The present review focuses on those interactions, especially as seen in FHM type 1, a variant of the disease caused by a mutation in CACNA1A, which encodes the α1A subunit of the P/Q-type voltage-gated calcium channel.
Collapse
Affiliation(s)
- Marta Kowalska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznan, Poland; (M.K.); (M.P.); (T.P.)
| | - Michał Prendecki
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznan, Poland; (M.K.); (M.P.); (T.P.)
| | - Thomas Piekut
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznan, Poland; (M.K.); (M.P.); (T.P.)
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznan, Poland;
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznan, Poland; (M.K.); (M.P.); (T.P.)
| |
Collapse
|
37
|
Young SM, Veeraraghavan P. Presynaptic voltage-gated calcium channels in the auditory brainstem. Mol Cell Neurosci 2021; 112:103609. [PMID: 33662542 DOI: 10.1016/j.mcn.2021.103609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/06/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022] Open
Abstract
Sound information encoding within the initial synapses in the auditory brainstem requires reliable and precise synaptic transmission in response to rapid and large fluctuations in action potential (AP) firing rates. The magnitude and location of Ca2+ entry through voltage-gated Ca2+ channels (CaV) in the presynaptic terminal are key determinants in triggering AP-mediated release. In the mammalian central nervous system (CNS), the CaV2.1 subtype is the critical subtype for CNS function, since it is the most efficient CaV2 subtype in triggering AP-mediated synaptic vesicle (SV) release. Auditory brainstem synapses utilize CaV2.1 to sustain fast and repetitive SV release to encode sound information. Therefore, understanding the presynaptic mechanisms that control CaV2.1 localization, organization and biophysical properties are integral to understanding auditory processing. Here, we review our current knowledge about the control of presynaptic CaV2 abundance and organization in the auditory brainstem and impact on the regulation of auditory processing.
Collapse
Affiliation(s)
- Samuel M Young
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Otolaryngology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | | |
Collapse
|
38
|
Gandini MA, Souza IA, Ferron L, Innes AM, Zamponi GW. The de novo CACNA1A pathogenic variant Y1384C associated with hemiplegic migraine, early onset cerebellar atrophy and developmental delay leads to a loss of Cav2.1 channel function. Mol Brain 2021; 14:27. [PMID: 33557884 PMCID: PMC7871581 DOI: 10.1186/s13041-021-00745-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
CACNA1A pathogenic variants have been linked to several neurological disorders including familial hemiplegic migraine and cerebellar conditions. More recently, de novo variants have been associated with severe early onset developmental encephalopathies. CACNA1A is highly expressed in the central nervous system and encodes the pore-forming CaVα1 subunit of P/Q-type (Cav2.1) calcium channels. We have previously identified a patient with a de novo missense mutation in CACNA1A (p.Y1384C), characterized by hemiplegic migraine, cerebellar atrophy and developmental delay. The mutation is located at the transmembrane S5 segment of the third domain. Functional analysis in two predominant splice variants of the neuronal Cav2.1 channel showed a significant loss of function in current density and changes in gating properties. Moreover, Y1384 variants exhibit differential splice variant-specific effects on recovery from inactivation. Finally, structural analysis revealed structural damage caused by the tyrosine substitution and changes in electrostatic potentials.
Collapse
Affiliation(s)
- Maria A Gandini
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ivana A Souza
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - 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
| | - A Micheil Innes
- Department of Medical Genetics and Alberta Children's Hospital Research Institute, 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
|
39
|
A CACNA1A variant associated with trigeminal neuralgia alters the gating of Cav2.1 channels. Mol Brain 2021; 14:4. [PMID: 33413531 PMCID: PMC7789175 DOI: 10.1186/s13041-020-00725-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/28/2020] [Indexed: 11/25/2022] Open
Abstract
A novel missense mutation in the CACNA1A gene that encodes the pore forming α1 subunit of the CaV2.1 voltage-gated calcium channel was identified in a patient with trigeminal neuralgia. This mutation leads to a substitution of proline 2455 by histidine (P2455H) in the distal C-terminus region of the channel. Due to the well characterized role of this channel in neurotransmitter release, our aim was to characterize the biophysical properties of the P2455H variant in heterologously expressed CaV2.1 channels. Whole-cell patch clamp recordings of wild type and mutant CaV2.1 channels expressed in tsA-201 cells reveal that the mutation mediates a depolarizing shift in the voltage-dependence of activation and inactivation. Moreover, the P2455H mutant strongly reduced calcium-dependent inactivation of the channel that is consistent with an overall gain of function. Hence, the P2455H CaV2.1 missense mutation alters the gating properties of the channel, suggesting that associated changes in CaV2.1-dependent synaptic communication in the trigeminal system may contribute to the development of trigeminal neuralgia.
Collapse
|
40
|
Yeow SQZ, Loh KWZ, Soong TW. Calcium Channel Splice Variants and Their Effects in Brain and Cardiovascular Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:67-86. [DOI: 10.1007/978-981-16-4254-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
41
|
Abstract
Potassium channels are present in every living cell and essential to setting up a stable, non-zero transmembrane electrostatic potential which manifests the off-equilibrium livelihood of the cell. They are involved in other cellular activities and regulation, such as the controlled release of hormones, the activation of T-cells for immune response, the firing of action potential in muscle cells and neurons, etc. Pharmacological reagents targeting potassium channels are important for treating various human diseases linked to dysfunction of the channels. High-resolution structures of these channels are very useful tools for delineating the detailed chemical basis underlying channel functions and for structure-based design and optimization of their pharmacological and pharmaceutical agents. Structural studies of potassium channels have revolutionized biophysical understandings of key concepts in the field - ion selectivity, conduction, channel gating, and modulation, making them multi-modality targets of pharmacological regulation. In this chapter, I will select a few high-resolution structures to illustrate key structural insights, proposed allostery behind channel functions, disagreements still open to debate, and channel-lipid interactions and co-evolution. The known structural consensus allows the inference of conserved molecular mechanisms shared among subfamilies of K+ channels and makes it possible to develop channel-specific pharmaceutical agents.
Collapse
Affiliation(s)
- Qiu-Xing Jiang
- Laboratory of Molecular Physiology and Biophysics and the Cryo-EM Center, Hauptmann-Woodward Medical Research Institute, Buffalo, NY, USA.
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, USA.
- Departments of Materials Design and Invention and Physiology and Biophysics, University of Buffalo (SUNY), Buffalo, NY, USA.
| |
Collapse
|
42
|
Koźmiński W, Pera J. Involvement of the Peripheral Nervous System in Episodic Ataxias. Biomedicines 2020; 8:biomedicines8110448. [PMID: 33105744 PMCID: PMC7690566 DOI: 10.3390/biomedicines8110448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/04/2022] Open
Abstract
Episodic ataxias comprise a group of inherited disorders, which have a common hallmark—transient attacks of ataxia. The genetic background is heterogeneous and the causative genes are not always identified. Furthermore, the clinical presentation, including intraictal and interictal symptoms, as well as the retention and progression of neurological deficits, is heterogeneous. Spells of ataxia can be accompanied by other symptoms—mostly from the central nervous system. However, in some of episodic ataxias involvement of peripheral nervous system is a part of typical clinical picture. This review intends to provide an insight into involvement of peripheral nervous system in episodic ataxias.
Collapse
Affiliation(s)
- Wojciech Koźmiński
- Department of Neurology, University Hospital, ul. Jakubowskiego 2, 30-688 Krakow, Poland;
| | - Joanna Pera
- Department of Neurology, Jagiellonian University Medical College, ul. Botaniczna 3, 31-503 Krakow, Poland
- Correspondence:
| |
Collapse
|
43
|
Maddox JW, Randall KL, Yadav RP, Williams B, Hagen J, Derr PJ, Kerov V, Della Santina L, Baker SA, Artemyev N, Hoon M, Lee A. A dual role for Ca v1.4 Ca 2+ channels in the molecular and structural organization of the rod photoreceptor synapse. eLife 2020; 9:e62184. [PMID: 32940604 PMCID: PMC7561352 DOI: 10.7554/elife.62184] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Synapses are fundamental information processing units that rely on voltage-gated Ca2+ (Cav) channels to trigger Ca2+-dependent neurotransmitter release. Cav channels also play Ca2+-independent roles in other biological contexts, but whether they do so in axon terminals is unknown. Here, we addressed this unknown with respect to the requirement for Cav1.4 L-type channels for the formation of rod photoreceptor synapses in the retina. Using a mouse strain expressing a non-conducting mutant form of Cav1.4, we report that the Cav1.4 protein, but not its Ca2+ conductance, is required for the molecular assembly of rod synapses; however, Cav1.4 Ca2+ signals are needed for the appropriate recruitment of postsynaptic partners. Our results support a model in which presynaptic Cav channels serve both as organizers of synaptic building blocks and as sources of Ca2+ ions in building the first synapse of the visual pathway and perhaps more broadly in the nervous system.
Collapse
Affiliation(s)
- J Wesley Maddox
- Department of Molecular Physiology and Biophysics, University of IowaIowa CityUnited States
- Iowa Neuroscience Institute, University of IowaIowa CityUnited States
- Pappajohn Biomedical Institute, University of IowaIowa CityUnited States
| | - Kate L Randall
- Department of Molecular Physiology and Biophysics, University of IowaIowa CityUnited States
- Iowa Neuroscience Institute, University of IowaIowa CityUnited States
- Pappajohn Biomedical Institute, University of IowaIowa CityUnited States
| | - Ravi P Yadav
- Department of Molecular Physiology and Biophysics, University of IowaIowa CityUnited States
| | - Brittany Williams
- Department of Molecular Physiology and Biophysics, University of IowaIowa CityUnited States
- Iowa Neuroscience Institute, University of IowaIowa CityUnited States
- Pappajohn Biomedical Institute, University of IowaIowa CityUnited States
| | - Jussara Hagen
- Department of Molecular Physiology and Biophysics, University of IowaIowa CityUnited States
- Iowa Neuroscience Institute, University of IowaIowa CityUnited States
- Pappajohn Biomedical Institute, University of IowaIowa CityUnited States
| | - Paul J Derr
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Vasily Kerov
- Department of Molecular Physiology and Biophysics, University of IowaIowa CityUnited States
- Iowa Neuroscience Institute, University of IowaIowa CityUnited States
| | - Luca Della Santina
- Department of Ophthalmology, University of California, San FranciscoSan FranciscoUnited States
| | - Sheila A Baker
- Iowa Neuroscience Institute, University of IowaIowa CityUnited States
- Department of Biochemistry, University of IowaIowa CityUnited States
- Department of OphthalmologyIowa CityUnited States
| | - Nikolai Artemyev
- Department of Molecular Physiology and Biophysics, University of IowaIowa CityUnited States
- Iowa Neuroscience Institute, University of IowaIowa CityUnited States
- Department of OphthalmologyIowa CityUnited States
| | - Mrinalini Hoon
- Department of Neuroscience, University of Wisconsin-MadisonMadisonUnited States
- Department of Ophthalmology and Visual Science, University of Wisconsin-MadisonMadisonUnited States
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of IowaIowa CityUnited States
- Iowa Neuroscience Institute, University of IowaIowa CityUnited States
- Pappajohn Biomedical Institute, University of IowaIowa CityUnited States
| |
Collapse
|
44
|
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
|
45
|
Giunti P, Mantuano E, Frontali M. Episodic Ataxias: Faux or Real? Int J Mol Sci 2020; 21:ijms21186472. [PMID: 32899446 PMCID: PMC7555854 DOI: 10.3390/ijms21186472] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 11/22/2022] Open
Abstract
The term Episodic Ataxias (EA) was originally used for a few autosomal dominant diseases, characterized by attacks of cerebellar dysfunction of variable duration and frequency, often accompanied by other ictal and interictal signs. The original group subsequently grew to include other very rare EAs, frequently reported in single families, for some of which no responsible gene was found. The clinical spectrum of these diseases has been enormously amplified over time. In addition, episodes of ataxia have been described as phenotypic variants in the context of several different disorders. The whole group is somewhat confused, since a strong evidence linking the mutation to a given phenotype has not always been established. In this review we will collect and examine all instances of ataxia episodes reported so far, emphasizing those for which the pathophysiology and the clinical spectrum is best defined.
Collapse
Affiliation(s)
- Paola Giunti
- Laboratory of Neurogenetics, Department of Molecular Neuroscience, UCL Institute of Neurology, London WC2N 5DU, UK
- Correspondence: (P.G.); (M.F.)
| | - Elide Mantuano
- Laboratory of Neurogenetics, Institute of Translational Pharmacology, National Research Council of Italy, 00133 Rome, Italy;
| | - Marina Frontali
- Laboratory of Neurogenetics, Institute of Translational Pharmacology, National Research Council of Italy, 00133 Rome, Italy;
- Correspondence: (P.G.); (M.F.)
| |
Collapse
|
46
|
Ortner NJ, Kaserer T, Copeland JN, Striessnig J. De novo CACNA1D Ca 2+ channelopathies: clinical phenotypes and molecular mechanism. Pflugers Arch 2020; 472:755-773. [PMID: 32583268 PMCID: PMC7351864 DOI: 10.1007/s00424-020-02418-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/04/2020] [Accepted: 06/10/2020] [Indexed: 12/15/2022]
Abstract
The identification of rare disease-causing variants in humans by large-scale next-generation sequencing (NGS) studies has also provided us with new insights into the pathophysiological role of de novo missense variants in the CACNA1D gene that encodes the pore-forming α1-subunit of voltage-gated Cav1.3 L-type Ca2+ channels. These CACNA1D variants have been identified somatically in aldosterone-producing adenomas as well as germline in patients with neurodevelopmental and in some cases endocrine symptoms. In vitro studies in heterologous expression systems have revealed typical gating changes that indicate enhanced Ca2+ influx through Cav1.3 channels as the underlying disease-causing mechanism. Here we summarize the clinical findings of 12 well-characterized individuals with a total of 9 high-risk pathogenic CACNA1D variants. Moreover, we propose how information from somatic mutations in aldosterone-producing adenomas could be used to predict the potential pathogenicity of novel germline variants. Since these pathogenic de novo variants can cause a channel-gain-of function, we also discuss the use of L-type Ca2+ channel blockers as a potential therapeutic option.
Collapse
Affiliation(s)
- Nadine J Ortner
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| | - Teresa Kaserer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - J Nathan Copeland
- Duke Center for Autism and Brain Development, Duke Child and Family Mental Health and Developmental Neuroscience, Durham, USA
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
47
|
Cardoso FC. Multi-targeting sodium and calcium channels using venom peptides for the treatment of complex ion channels-related diseases. Biochem Pharmacol 2020; 181:114107. [PMID: 32579958 DOI: 10.1016/j.bcp.2020.114107] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/13/2020] [Accepted: 06/17/2020] [Indexed: 02/08/2023]
Abstract
Venom peptides are amongst the most exquisite group of bioactive molecules able to alter the normal physiology of organisms. These bioactive peptides penetrate tissues and blood vessels to encounter a number of receptors and ion channels to which they bind with high affinity and execute modulatory activities. Arachnid is the most diverse class of venomous animals often rich in peptides modulating voltage-gated sodium (NaV), calcium (CaV), and potassium (KV) channels. Spider venoms, in particular, contain potent and selective peptides targeting these channels, with a few displaying interesting multi-target properties for NaV and CaV channels underlying disease mechanisms such as in neuropathic pain, motor neuron disease and cancer. The elucidation of the pharmacology and structure-function properties of these venom peptides are invaluable for the development of effective drugs targeting NaV and CaV channels. This perspective discusses spider venom peptides displaying multi-target properties to modulate NaV and CaV channels in regard to their pharmacological features, structure-function relationships and potential to become the next generation of effective drugs to treat neurological disorders and other multi-ion channels related diseases.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd., St Lucia, QLD AU 4072, Australia
| |
Collapse
|
48
|
Rare CACNA1A mutations leading to congenital ataxia. Pflugers Arch 2020; 472:791-809. [DOI: 10.1007/s00424-020-02396-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/03/2023]
|
49
|
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] [MESH Headings] [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
|
50
|
Dolphin AC, Lee A. Presynaptic calcium channels: specialized control of synaptic neurotransmitter release. Nat Rev Neurosci 2020; 21:213-229. [PMID: 32161339 PMCID: PMC7873717 DOI: 10.1038/s41583-020-0278-2] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 11/09/2022]
Abstract
Chemical synapses are heterogeneous junctions formed between neurons that are specialized for the conversion of electrical impulses into the exocytotic release of neurotransmitters. Voltage-gated Ca2+ channels play a pivotal role in this process as they are the major conduits for the Ca2+ ions that trigger the fusion of neurotransmitter-containing vesicles with the presynaptic membrane. Alterations in the intrinsic function of these channels and their positioning within the active zone can profoundly alter the timing and strength of synaptic output. Advances in optical and electron microscopic imaging, structural biology and molecular techniques have facilitated recent breakthroughs in our understanding of the properties of voltage-gated Ca2+ channels that support their presynaptic functions. Here we examine the nature of these channels, how they are trafficked to and anchored within presynaptic boutons, and the mechanisms that allow them to function optimally in shaping the flow of information through neural circuits.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|