1
|
Tan JK, Awuah WA, Ahluwalia A, Sanker V, Ben-Jaafar A, Tenkorang PO, Aderinto N, Mehta A, Darko K, Shah MH, Roy S, Abdul-Rahman T, Atallah O. Genes to therapy: a comprehensive literature review of whole-exome sequencing in neurology and neurosurgery. Eur J Med Res 2024; 29:538. [PMID: 39523358 PMCID: PMC11552425 DOI: 10.1186/s40001-024-02063-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/12/2024] [Indexed: 11/16/2024] Open
Abstract
Whole-exome sequencing (WES), a ground-breaking technology, has emerged as a linchpin in neurology and neurosurgery, offering a comprehensive elucidation of the genetic landscape of various neurological disorders. This transformative methodology concentrates on the exonic portions of DNA, which constitute approximately 1% of the human genome, thus facilitating an expedited and efficient sequencing process. WES has been instrumental in advancing our understanding of neurodegenerative diseases, neuro-oncology, cerebrovascular disorders, and epilepsy by revealing rare variants and novel mutations and providing intricate insights into their genetic complexities. This has been achieved while maintaining a substantial diagnostic yield, thereby offering novel perspectives on the pathophysiology and personalized management of these conditions. The utilization of WES boasts several advantages over alternative genetic sequencing methodologies, including cost-effectiveness, reduced incidental findings, simplified analysis and interpretation process, and reduced computational demands. However, despite its benefits, there are challenges, such as the interpretation of variants of unknown significance, cost considerations, and limited accessibility in resource-constrained settings. Additionally, ethical, legal, and social concerns are raised, particularly in the context of incidental findings and patient consent. As we look to the future, the integration of WES with other omics-based approaches could help revolutionize the field of personalized medicine through its implications in predictive models and the development of targeted therapeutic strategies, marking a significant stride toward more effective and clinically oriented solutions.
Collapse
Affiliation(s)
- Joecelyn Kirani Tan
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.
| | | | | | - Vivek Sanker
- Department of Neurosurgery, Trivandrum Medical College, Thiruvananthapuram, India
| | - Adam Ben-Jaafar
- University College Dublin, School of Medicine, Belfield, Dublin 4, Ireland
| | | | - Nicholas Aderinto
- Internal Medicine Department, LAUTECH Teaching Hospital, Ogbomoso, Nigeria
| | - Aashna Mehta
- University of Debrecen-Faculty of Medicine, Debrecen, Hungary
| | - Kwadwo Darko
- Department of Neurosurgery, Korle Bu Teaching Hospital, Accra, Ghana
| | | | - Sakshi Roy
- School of Medicine, Queen's University Belfast, Belfast, UK
| | | | - Oday Atallah
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| |
Collapse
|
2
|
Reis MC, Mandler L, Kang J, Oliver D, Halaszovich C, Nolte D. A novel KCND3 variant in the N-terminus impairs the ionic current of Kv4.3 and is associated with SCA19/22. J Cell Mol Med 2024; 28:e70039. [PMID: 39180521 PMCID: PMC11344468 DOI: 10.1111/jcmm.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/05/2024] [Indexed: 08/26/2024] Open
Abstract
Spinocerebellar ataxias (SCAs) are a genetically heterogeneous group of autosomal dominant movement disorders. Among the SCAs associated with impaired ion channel function, SCA19/22 is caused by pathogenic variants in KCND3, which encodes the voltage-gated potassium channel Kv4.3. SCA19/22 is clinically characterized by ataxia, dysarthria and oculomotor dysfunction in combination with other signs and symptoms, including mild cognitive impairment, peripheral neuropathy and pyramidal signs. The known KCND3 pathogenic variants are localized either in the transmembrane segments, the connecting loops, or the C-terminal region of Kv4.3. We have identified a novel pathogenic variant, c.455A>G (p.D152G), localized in the N-terminus of Kv4.3. It is located in the immediate neighbourhood of the T1 domain, which is responsible for multimerization with the β-subunit KChIP2b and thus for the formation of functional heterooctamers. Electrophysiological studies showed that p.D152G does not affect channel gating, but reduces the ionic current in Kv4.3, even though the variant is not located in the transmembrane domains. Impaired channel trafficking to the plasma membrane may contribute to this effect. In a patient with a clinical picture corresponding to SCA19/22, p.D152G is the first pathogenic variant in the N-terminus of Kv4.3 to be described to date with an effect on ion channel activity.
Collapse
Affiliation(s)
| | - Laura Mandler
- Institute of Human GeneticsJustus‐Liebig‐University GiessenGiessenGermany
- Present address:
Department of NeurologyJustus‐Liebig‐University GiessenGiessenGermany
| | - Jun‐Suk Kang
- Department of NeurologyGoethe‐University FrankfurtFrankfurtGermany
- Present address:
NeuropraxisFrankfurtGermany
| | - Dominik Oliver
- Institute of PhysiologyPhilipps‐University MarburgMarburgGermany
| | | | - Dagmar Nolte
- Institute of Human GeneticsJustus‐Liebig‐University GiessenGiessenGermany
| |
Collapse
|
3
|
Contaldi E, Gallo S, Corrado L, D'Alfonso S, Magistrelli L. Parkinsonism in SCA19/22: Dopamine Transporter Imaging in an Italian Family Harboring a Novel Mutation. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1226-1230. [PMID: 37857779 DOI: 10.1007/s12311-023-01619-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 10/21/2023]
Abstract
Spinocerebellar ataxia (SCA)19/22 is a channelopathy caused by mutations in the KCND3 gene encoding for the voltage-gated potassium channel Kv4.3. In the present work, we report an Italian family harboring a novel KCND3 missense mutation characterized by ataxia and mild parkinsonism. Patients underwent dopamine transporter single-photon emission computed tomography to assess dopaminergic degeneration. Normal findings were observed, and treatment with levodopa did not yield any benefit, thus suggesting the involvement of other mechanisms to explain parkinsonian symptoms in SCA19/22. Our cases expand the genetic and imaging spectrum of this rare disease and emphasize a cautious approach in managing parkinsonism in these patients.
Collapse
Affiliation(s)
- Elena Contaldi
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Corso Mazzini 18, 28100, Novara, Italy.
- PhD Program in Medical Sciences and Biotechnology, University of Piemonte Orientale, 28100, Novara, Italy.
| | - Silvia Gallo
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Corso Mazzini 18, 28100, Novara, Italy
| | - Lucia Corrado
- Department of Health Sciences, Centre of Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale, 28100, Novara, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, Centre of Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale, 28100, Novara, Italy
| | - Luca Magistrelli
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Corso Mazzini 18, 28100, Novara, Italy
| |
Collapse
|
4
|
Gazulla J, Berciano J. Potential Benefit of Channel Activators in Loss-of-Function Primary Potassium Channelopathies Causing Heredoataxia. CEREBELLUM (LONDON, ENGLAND) 2024; 23:833-837. [PMID: 37460907 DOI: 10.1007/s12311-023-01584-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/29/2023] [Indexed: 03/20/2024]
Abstract
Potassium channels (KCN) are transmembrane complexes that regulate the resting membrane potential and the duration of action potentials in cells. The opening of KCN brings about an efflux of K+ ions that induces cell repolarization after depolarization, returns the transmembrane potential to its resting state, and enables for continuous spiking ability. The aim of this work was to assess the role of KCN dysfunction in the pathogenesis of hereditary ataxias and the mechanisms of action of KCN opening agents (KCO). In consequence, a review of the ad hoc medical literature was performed. Among hereditary KCN diseases causing ataxia, mutated Kv3.3, Kv4.3, and Kv1.1 channels provoke spinocerebellar ataxia (SCA) type 13, SCA19/22, and episodic ataxia type 1 (EA1), respectively. The K+ efflux was found to be reduced in experimental models of these diseases, resulting in abnormally prolonged depolarization and incomplete repolarization, thereby interfering with repetitive discharges in the cells. Hence, substances able to promote normal spiking activity in the cerebellum could provide symptomatic benefit. Although drugs used in clinical practice do not activate Kv3.3 or Kv4.3 directly, available KCO probably could ameliorate ataxic symptoms in SCA13 and SCA19/22, as verified with acetazolamide in EA1, and retigabine in a mouse model of hypokalemic periodic paralysis. To summarize, ataxia could possibly be improved by non-specific KCO in SCA13 and SCA19/22. The identification of new specific KCO agents will undoubtedly constitute a promising therapeutic strategy for these diseases.
Collapse
Affiliation(s)
- José Gazulla
- Department of Neurology, Hospital Universitario Miguel Servet, Isabel la Católica, 1-3, 50009, Saragossa, Spain.
| | - José Berciano
- Department of Neurology, Hospital Universitario Marqués de Valdecilla (IDIVAL), University of Cantabria, CIBERNED, Avenida de Valdecilla S/N, 39008, Santander, Spain
| |
Collapse
|
5
|
Orfali R, Alwatban AZ, Orfali RS, Lau L, Chea N, Alotaibi AM, Nam YW, Zhang M. Oxidative stress and ion channels in neurodegenerative diseases. Front Physiol 2024; 15:1320086. [PMID: 38348223 PMCID: PMC10859863 DOI: 10.3389/fphys.2024.1320086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/12/2024] [Indexed: 02/15/2024] Open
Abstract
Numerous neurodegenerative diseases result from altered ion channel function and mutations. The intracellular redox status can significantly alter the gating characteristics of ion channels. Abundant neurodegenerative diseases associated with oxidative stress have been documented, including Parkinson's, Alzheimer's, spinocerebellar ataxia, amyotrophic lateral sclerosis, and Huntington's disease. Reactive oxygen and nitrogen species compounds trigger posttranslational alterations that target specific sites within the subunits responsible for channel assembly. These alterations include the adjustment of cysteine residues through redox reactions induced by reactive oxygen species (ROS), nitration, and S-nitrosylation assisted by nitric oxide of tyrosine residues through peroxynitrite. Several ion channels have been directly investigated for their functional responses to oxidizing agents and oxidative stress. This review primarily explores the relationship and potential links between oxidative stress and ion channels in neurodegenerative conditions, such as cerebellar ataxias and Parkinson's disease. The potential correlation between oxidative stress and ion channels could hold promise for developing innovative therapies for common neurodegenerative diseases.
Collapse
Affiliation(s)
- Razan Orfali
- Neuroscience Research Department, Research Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Adnan Z. Alwatban
- Neuroscience Research Department, Research Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | - Liz Lau
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| | - Noble Chea
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| | - Abdullah M. Alotaibi
- Neuroscience Research Department, Research Centre, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, United States
| |
Collapse
|
6
|
Khan R, Chaturvedi P, Sahu P, Ludhiadch A, Singh P, Singh G, Munshi A. Role of Potassium Ion Channels in Epilepsy: Focus on Current Therapeutic Strategies. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:67-87. [PMID: 36578258 DOI: 10.2174/1871527322666221227112621] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Epilepsy is one of the prevalent neurological disorders characterized by disrupted synchronization between inhibitory and excitatory neurons. Disturbed membrane potential due to abnormal regulation of neurotransmitters and ion transport across the neural cell membrane significantly contributes to the pathophysiology of epilepsy. Potassium ion channels (KCN) regulate the resting membrane potential and are involved in neuronal excitability. Genetic alterations in the potassium ion channels (KCN) have been reported to result in the enhancement of the release of neurotransmitters, the excitability of neurons, and abnormal rapid firing rate, which lead to epileptic phenotypes, making these ion channels a potential therapeutic target for epilepsy. The aim of this study is to explore the variations reported in different classes of potassium ion channels (KCN) in epilepsy patients, their functional evaluation, and therapeutic strategies to treat epilepsy targeting KCN. METHODOLOGY A review of all the relevant literature was carried out to compile this article. RESULTS A large number of variations have been reported in different genes encoding various classes of KCN. These genetic alterations in KCN have been shown to be responsible for disrupted firing properties of neurons. Antiepileptic drugs (AEDs) are the main therapeutic strategy to treat epilepsy. Some patients do not respond favorably to the AEDs treatment, resulting in pharmacoresistant epilepsy. CONCLUSION Further to address the challenges faced in treating epilepsy, recent approaches like optogenetics, chemogenetics, and genome editing, such as clustered regularly interspaced short palindromic repeats (CRISPR), are emerging as target-specific therapeutic strategies.
Collapse
Affiliation(s)
- Rahul Khan
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda 151401, India
| | - Pragya Chaturvedi
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda 151401, India
| | - Prachi Sahu
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda 151401, India
| | - Abhilash Ludhiadch
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda 151401, India
| | - Paramdeep Singh
- Department of Radiology, All India Institute of Medical Sciences, Bathinda, Punjab, 151001 India
| | - Gagandeep Singh
- Department of Neurology, Dayanand Medical College and Hospital, Ludhiana, Punjab, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda 151401, India
| |
Collapse
|
7
|
Karatug Kacar A. Exploring dual effects of dinutuximab beta on cell death and proliferation of insulinoma. Chem Biol Drug Des 2024; 103:e14368. [PMID: 37802653 DOI: 10.1111/cbdd.14368] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/13/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023]
Abstract
Insulinoma INS-1 cells are pancreatic beta cell tumors. Dinutuximab beta (DB) is a monoclonal antibody used in the treatment of neuroblastoma. The aim of this study is to investigate the effects of DB on pancreatic beta cell tumors at the molecular level. DB (Qarziba®) was available from EUSA Pharma. Streptozotocin (STZ) was used induce to cell cytotoxicity. DB was applied to the cells before or after the STZ application. KCND3, KCNN4, KCNK1, and PTHrP gene expression levels were analyzed by q-RT-PCR, and protein levels were analyzed by Western blotting. Analysis of glucose-stimulated insulin secretion was performed. Ca+2 and CA19-9 levels were determined by the ELISA kit. PERK, CHOP, HSP90, p-c-Jun, p-Atf2, and p-Elk1 protein levels were analyzed by simple WES. Decreased KCND3, KCNK1, and PTHrP protein levels and increased KCND3, KCNN4, KCNK1, and PTHrP gene expression levels were observed with DB applied after STZ application. Cell dysfunction was detected with DB applied before and after STZ application. Ca19-9 and Ca+2 levels were increased with DB applied after STZ application. PERK, CHOP, and p-Elk1 levels decreased, while HSP90 levels increased with DB applied after STZ application. CHOP, p-Akt-2, and p-c-Jun levels increased in the DB group. As a result, INS-1 cells go to cell death via the ERK signaling pathway without ER stress and release insulin with the decrease of K+ channels and an increase in Ca+2 levels with DB applied after STZ application. Moreover, the cells proliferate via JNK signaling with DB application. DB holds promise for the treatment of insulinoma. The study should be supported by in vivo studies.
Collapse
Affiliation(s)
- Ayse Karatug Kacar
- Faculty of Science, Department of Biology, Istanbul University, Istanbul, Turkey
| |
Collapse
|
8
|
Alam KA, Svalastoga P, Martinez A, Glennon JC, Haavik J. Potassium channels in behavioral brain disorders. Molecular mechanisms and therapeutic potential: A narrative review. Neurosci Biobehav Rev 2023; 152:105301. [PMID: 37414376 DOI: 10.1016/j.neubiorev.2023.105301] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
Potassium channels (K+-channels) selectively control the passive flow of potassium ions across biological membranes and thereby also regulate membrane excitability. Genetic variants affecting many of the human K+-channels are well known causes of Mendelian disorders within cardiology, neurology, and endocrinology. K+-channels are also primary targets of many natural toxins from poisonous organisms and drugs used within cardiology and metabolism. As genetic tools are improving and larger clinical samples are being investigated, the spectrum of clinical phenotypes implicated in K+-channels dysfunction is rapidly expanding, notably within immunology, neurosciences, and metabolism. K+-channels that previously were considered to be expressed in only a few organs and to have discrete physiological functions, have recently been found in multiple tissues and with new, unexpected functions. The pleiotropic functions and patterns of expression of K+-channels may provide additional therapeutic opportunities, along with new emerging challenges from off-target effects. Here we review the functions and therapeutic potential of K+-channels, with an emphasis on the nervous system, roles in neuropsychiatric disorders and their involvement in other organ systems and diseases.
Collapse
Affiliation(s)
| | - Pernille Svalastoga
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway; Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
| | | | - Jeffrey Colm Glennon
- Conway Institute for Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland.
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Norway; Division of Psychiatry, Haukeland University Hospital, Norway.
| |
Collapse
|
9
|
Ågren R, Geerdink N, Brunner HG, Paucar M, Kamsteeg EJ, Sahlholm K. An E280K Missense Variant in KCND3/Kv4.3-Case Report and Functional Characterization. Int J Mol Sci 2023; 24:10924. [PMID: 37446101 DOI: 10.3390/ijms241310924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
A five-year-old girl presented with headache attacks, clumsiness, and a history of transient gait disturbances. She and her father, mother, twin sister, and brother underwent neurological evaluation, neuroimaging, and exome sequencing covering 357 genes associated with movement disorders. Sequencing revealed the new variant KCND3 c.838G>A, p.E280K in the father and sisters, but not in the mother and brother. KCND3 encodes voltage-gated potassium channel D3 (Kv4.3) and mutations have been associated with spinocerebellar ataxia type 19/22 (SCA19/22) and cardiac arrhythmias. SCA19/22 is characterized by ataxia, Parkinsonism, peripheral neuropathy, and sometimes, intellectual disability. Neuroimaging, EEG, and ECG were unremarkable. Mild developmental delay with impaired fluid reasoning was observed in both sisters, but not in the brother. None of the family members demonstrated ataxia or parkinsonism. In Xenopus oocyte electrophysiology experiments, E280K was associated with a rightward shift in the Kv4.3 voltage-activation relationship of 11 mV for WT/E280K and +17 mV for E280K/E280K relative to WT/WT. Steady-state inactivation was similarly right-shifted. Maximal peak current amplitudes were similar for WT/WT, WT/E280K, and E280K/E280K. Our data indicate that Kv4.3 E280K affects channel activation and inactivation and is associated with developmental delay. However, E280K appears to be relatively benign considering it does not result in overt ataxia.
Collapse
Affiliation(s)
- Richard Ågren
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Niels Geerdink
- Department of Pediatrics, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| | - Han G Brunner
- Department of Human Genetics, Donders Centre for Brain, Cognition and Behavior, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Department of Clinical Genetics, MUMC Maastricht, GROW School for Oncology and Developmental Biology, MHENS School for Mental Health and Neuroscience, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Martin Paucar
- Department of Neurology, Karolinska University Hospital, 141 86 Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud UMC, 6525 GA Nijmegen, The Netherlands
| | - Kristoffer Sahlholm
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Integrative Medical Biology, Wallenberg Centre for Molecular Medicine, Umeå University, 901 87 Umeå, Sweden
| |
Collapse
|
10
|
Huang H, Shakkottai VG. Targeting Ion Channels and Purkinje Neuron Intrinsic Membrane Excitability as a Therapeutic Strategy for Cerebellar Ataxia. Life (Basel) 2023; 13:1350. [PMID: 37374132 DOI: 10.3390/life13061350] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
In degenerative neurological disorders such as Parkinson's disease, a convergence of widely varying insults results in a loss of dopaminergic neurons and, thus, the motor symptoms of the disease. Dopamine replacement therapy with agents such as levodopa is a mainstay of therapy. Cerebellar ataxias, a heterogeneous group of currently untreatable conditions, have not been identified to have a shared physiology that is a target of therapy. In this review, we propose that perturbations in cerebellar Purkinje neuron intrinsic membrane excitability, a result of ion channel dysregulation, is a common pathophysiologic mechanism that drives motor impairment and vulnerability to degeneration in cerebellar ataxias of widely differing genetic etiologies. We further propose that treatments aimed at restoring Purkinje neuron intrinsic membrane excitability have the potential to be a shared therapy in cerebellar ataxia akin to levodopa for Parkinson's disease.
Collapse
Affiliation(s)
- Haoran Huang
- Medical Scientist Training Program, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Vikram G Shakkottai
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
11
|
Spagnoli C, Fusco C, Pisani F. Pediatric-Onset Epilepsy and Developmental Epileptic Encephalopathies Followed by Early-Onset Parkinsonism. Int J Mol Sci 2023; 24:ijms24043796. [PMID: 36835207 PMCID: PMC9965035 DOI: 10.3390/ijms24043796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Genetic early-onset Parkinsonism is unique due to frequent co-occurrence of hyperkinetic movement disorder(s) (MD), or additional neurological of systemic findings, including epilepsy in up to 10-15% of cases. Based on both the classification of Parkinsonism in children proposed by Leuzzi and coworkers and the 2017 ILAE epilepsies classification, we performed a literature review in PubMed. A few discrete presentations can be identified: Parkinsonism as a late manifestation of complex neurodevelopmental disorders, characterized by developmental and epileptic encephalopathies (DE-EE), with multiple, refractory seizure types and severely abnormal EEG characteristics, with or without preceding hyperkinetic MD; Parkinsonism in the context of syndromic conditions with unspecific reduced seizure threshold in infancy and childhood; neurodegenerative conditions with brain iron accumulation, in which childhood DE-EE is followed by neurodegeneration; and finally, monogenic juvenile Parkinsonism, in which a subset of patients with intellectual disability or developmental delay (ID/DD) develop hypokinetic MD between 10 and 30 years of age, following unspecific, usually well-controlled, childhood epilepsy. This emerging group of genetic conditions leading to epilepsy or DE-EE in childhood followed by juvenile Parkinsonism highlights the need for careful long-term follow-up, especially in the context of ID/DD, in order to readily identify individuals at increased risk of later Parkinsonism.
Collapse
Affiliation(s)
- Carlotta Spagnoli
- Child Neurology and Psychiatry Unit, Department of Pediatrics, Presidio Ospedaliero Santa Maria Nuova, AUSL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy
- Correspondence: ; Tel.: +39-0522-296033
| | - Carlo Fusco
- Child Neurology and Psychiatry Unit, Department of Pediatrics, Presidio Ospedaliero Santa Maria Nuova, AUSL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy
| | - Francesco Pisani
- Human Neurosciences Department, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
12
|
Elmalik SA. Electrophysiology of ataxia-telangiectasia-like disorder 1. Sudan J Paediatr 2023; 23:153-162. [PMID: 38380400 PMCID: PMC10876264 DOI: 10.24911/sjp.106-1703054783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 12/20/2023] [Indexed: 02/22/2024]
Abstract
Ataxia-telangiectasia-like disorder-1 (ATLD1, OMIM # 604391) is a very rare clinical condition, characterized by slowly progressive ataxia with onset in childhood, associated with oculomotor apraxia and dysarthria. Laboratory findings reveal increased susceptibility to radiation, with a defect in DNA repair. Patients with ATLD1 show no telangiectasia, have no immunodeficiency, and also have preserved cognition. Reflexes might be initially brisk and later becomes reduced associated with axonal sensorimotor neuropathy. Brain magnetic resonance imaging (MRI) detects cerebellar atrophy. The condition is caused by mutations in the meiotic recombination 11 (MRE11A) gene. The present study reports on the neurophysiologic finding in eight Saudi patients, belonging to three Saudi families, who have genetically confirmed ATLD1. All investigated patients had cerebellar atrophy on brain MRI (5/5). Electrophysiologic studies showed normal motor conduction velocity (MCV) of the median (8/8) and tibial (2/2) nerves, while 5/6 (83%) had normal peroneal nerve MCV. The distal motor latency (DML) for median, tibial, and peroneal nerves was within the normal range in all examined patients. The amplitude of compound muscle action potential (CMAP) of median and tibial nerves was also normal, while that of the peroneal nerve was normal in 3/6 (50%). Two of seven (29%) patients had reduced amplitude of median nerve sensory nerve action potential (SNAP) while 3/8 (38%) had a reduction in the amplitude of sural nerve SNAP. These findings favour an axonal type of neuropathy predominately affecting the sensory fibres (axonal sensorimotor neuropathy). The present study constitutes the largest cohort of ATLD1 patients worldwide who had electrophysiologic tests.
Collapse
Affiliation(s)
- Salah A Elmalik
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
13
|
Younger DS. Neurogenetic motor disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:183-250. [PMID: 37562870 DOI: 10.1016/b978-0-323-98818-6.00003-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Advances in the field of neurogenetics have practical applications in rapid diagnosis on blood and body fluids to extract DNA, obviating the need for invasive investigations. The ability to obtain a presymptomatic diagnosis through genetic screening and biomarkers can be a guide to life-saving disease-modifying therapy or enzyme replacement therapy to compensate for the deficient disease-causing enzyme. The benefits of a comprehensive neurogenetic evaluation extend to family members in whom identification of the causal gene defect ensures carrier detection and at-risk counseling for future generations. This chapter explores the many facets of the neurogenetic evaluation in adult and pediatric motor disorders as a primer for later chapters in this volume and a roadmap for the future applications of genetics in neurology.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
14
|
Baviera-Muñoz R, Carretero-Vilarroig L, Vázquez-Costa JF, Morata-Martínez C, Campins-Romeu M, Muelas N, Sastre-Bataller I, Martínez-Torres I, Pérez-García J, Sivera R, Sevilla T, Vilchez JJ, Jaijo T, Espinós C, Millán JM, Bataller L, Aller E. Diagnostic Efficacy of Genetic Studies in a Series of Hereditary Cerebellar Ataxias in Eastern Spain. NEUROLOGY GENETICS 2022; 8:e200038. [DOI: 10.1212/nxg.0000000000200038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022]
Abstract
Background and ObjectivesTo determine the diagnostic efficacy of clinical exome-targeted sequencing (CES) and spinocerebellar ataxia 36 (SCA36) screening in a real-life cohort of patients with cerebellar ataxia (CA) from Eastern Spain.MethodsA total of 130 unrelated patients with CA, negative for common trinucleotide repeat expansions (SCA1, SCA2, SCA3, SCA6, SCA7, SCA8, SCA12, SCA17, dentatorubral pallidoluysian atrophy [DRPLA], and Friedreich ataxia), were studied with CES. Bioinformatic and genotype-phenotype analyses were performed to assess the pathogenicity of the variants encountered. Copy number variants were analyzed when appropriate. In undiagnosed dominant and sporadic cases, repeat primed PCR was used to screen for the presence of a repeat expansion in theNOP56gene.ResultsCES identified pathogenic or likely pathogenic variants in 50 families (39%), including 23 novel variants. Overall, there was a high genetic heterogeneity, and the most frequent genetic diagnosis wasSPG7(n = 15), followed bySETX(n = 6),CACNA1A(n = 5),POLR3A(n = 4), andSYNE1(n = 3). In addition, 17 families displayed likely pathogenic/pathogenic variants in 14 different genes:KCND3(n = 2),KIF1C(n = 2),CYP27A1A(n = 2),AFG3L2(n = 1),ANO10(n = 1),CAPN1(n = 1),CWF19L1(n = 1),ITPR1(n = 1),KCNA1(n = 1),OPA1(n = 1),PNPLA6(n = 1),SPG11(n = 1),SPTBN2(n = 1), andTPP1(n = 1). Twenty-two novel variants were characterized. SCA36 was diagnosed in 11 families, all with autosomal dominant (AD) presentation. SCA36 screening increased the total diagnostic rate to 47% (n = 61/130). Ultimately, undiagnosed patients showed delayed age at onset (p< 0.05) and were more frequently sporadic.DiscussionOur study provides insight into the genetic landscape of CA in Eastern Spain. Although CES was an effective approach to capture genetic heterogeneity, most patients remained undiagnosed. SCA36 was found to be a relatively frequent form and, therefore, should be tested prior to CES in familial AD presentations in particular geographical regions.
Collapse
|
15
|
Palombo F, La Morgia C, Fiorini C, Caporali L, Valentino ML, Donadio V, Liguori R, Carelli V. A Second Case With the V374A KCND3 Pathogenic Variant in an Italian Patient With Early-Onset Spinocerebellar Ataxia. Neurol Genet 2022; 8:e200004. [PMID: 35949253 PMCID: PMC9359624 DOI: 10.1212/nxg.0000000000200004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/13/2022] [Indexed: 11/18/2022]
Abstract
Background and Objectives To date, approximately 20 heterozygous mainly loss-of-function variants in KCND3 have been associated with spinocerebellar ataxia (SCA) type 19 and 22, a clinically heterogeneous group of neurodegenerative disorders. We aimed at reporting the second patients with the V374A KCND3 mutation from an independent family, confirming its pathogenic role. Methods We describe the clinical history of a patient with SCA and conducted genetic investigations including mitochondrial DNA analysis and exome sequencing. Results This male patient was reported to have unstable gait with tremors at the lower limbs and dysarthric speech since childhood. A neurologic examination also showed dysarthria, nystagmus, action tremor, dysmetria, and weak deep tendon reflexes. He had marked cerebellar atrophy at brain MRI, more evident at vermis. Molecular analysis, including exome sequencing and an in silico panel analysis of genes associated with SCA, revealed the c.1121T>C [p.V374A] mutation in KCND3. Discussion This report consolidates the pathogenicity of the V374A KCND3 mutation and suggests that the ataxic paroxysmal exacerbations are not a key phenotypic feature of this mutation.
Collapse
|
16
|
Agostini F, Agostinis R, Medina DL, Bisaglia M, Greggio E, Plotegher N. The Regulation of MiTF/TFE Transcription Factors Across Model Organisms: from Brain Physiology to Implication for Neurodegeneration. Mol Neurobiol 2022; 59:5000-5023. [PMID: 35665902 PMCID: PMC9363479 DOI: 10.1007/s12035-022-02895-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/21/2022] [Indexed: 12/30/2022]
Abstract
The microphthalmia/transcription factor E (MiTF/TFE) transcription factors are responsible for the regulation of various key processes for the maintenance of brain function, including autophagy-lysosomal pathway, lipid catabolism, and mitochondrial homeostasis. Among them, autophagy is one of the most relevant pathways in this frame; it is evolutionary conserved and crucial for cellular homeostasis. The dysregulation of MiTF/TFE proteins was shown to be involved in the development and progression of neurodegenerative diseases. Thus, the characterization of their function is key in the understanding of the etiology of these diseases, with the potential to develop novel therapeutics targeted to MiTF/TFE proteins and to the autophagic process. The fact that these proteins are evolutionary conserved suggests that their function and dysfunction can be investigated in model organisms with a simpler nervous system than the mammalian one. Building not only on studies in mammalian models but also in complementary model organisms, in this review we discuss (1) the mechanistic regulation of MiTF/TFE transcription factors; (2) their roles in different regions of the central nervous system, in different cell types, and their involvement in the development of neurodegenerative diseases, including lysosomal storage disorders; (3) the overlap and the compensation that occur among the different members of the family; (4) the importance of the evolutionary conservation of these protein and the process they regulate, which allows their study in different model organisms; and (5) their possible role as therapeutic targets in neurodegeneration.
Collapse
Affiliation(s)
| | - Rossella Agostinis
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Scuola Superiore Meridionale SSM, Federico II University, Naples, Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Department of Medical and Translational, Science, II University, Naples, Federico, Italy
| | - Marco Bisaglia
- Department of Biology, University of Padova, Padua, Italy
| | - Elisa Greggio
- Department of Biology, University of Padova, Padua, Italy
| | | |
Collapse
|
17
|
Li M, Liu F, Hao X, Fan Y, Li J, Hu Z, Shi J, Fan L, Zhang S, Ma D, Guo M, Xu Y, Shi C. Rare KCND3 Loss-of-Function Mutation Associated With the SCA19/22. Front Mol Neurosci 2022; 15:919199. [PMID: 35813061 PMCID: PMC9261871 DOI: 10.3389/fnmol.2022.919199] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/19/2022] [Indexed: 12/15/2022] Open
Abstract
Spinocerebellar ataxia 19/22 (SCA19/22) is a rare neurodegenerative disorder caused by mutations of the KCND3 gene, which encodes the Kv4. 3 protein. Currently, only 22 KCND3 single-nucleotide mutation sites of SCA19/22 have been reported worldwide, and detailed pathogenesis remains unclear. In this study, Sanger sequencing was used to screen 115 probands of cerebellar ataxia families in 67 patients with sporadic cerebellar ataxia and 200 healthy people to identify KCND3 mutations. Mutant gene products showed pathogenicity damage, and the polarity was changed. Next, we established induced pluripotent stem cells (iPSCs) derived from SCA19/22 patients. Using a transcriptome sequencing technique, we found that protein processing in the endoplasmic reticulum was significantly enriched in SCA19/22-iPS-derived neurons and was closely related to endoplasmic reticulum stress (ERS) and apoptosis. In addition, Western blotting of the SCA19/22-iPS-derived neurons showed a reduction in Kv4.3; but, activation of transcription factor 4 (ATF4) and C/EBP homologous protein was increased. Therefore, the c.1130 C>T (p.T377M) mutation of the KCND3 gene may mediate misfold and aggregation of Kv4.3, which activates the ERS and further induces neuron apoptosis involved in SCA19/22.
Collapse
Affiliation(s)
- Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Fen Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Yu Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Jiadi Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Zhengwei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Jingjing Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Department of Cell Biology and Medical Genetics, Basic Medical College of Zhengzhou University, Zhengzhou, China
| | - Liyuan Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Dongrui Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Mengnan Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Department of Cell Biology and Medical Genetics, Basic Medical College of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Hereditary Neurodegenerative Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Key Laboratory of Cerebrovascular Diseases Prevention and Treatment, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Hereditary Neurodegenerative Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Key Laboratory of Cerebrovascular Diseases Prevention and Treatment, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- *Correspondence: Changhe Shi
| |
Collapse
|
18
|
Bushart DD, Shakkottai VG. Vulnerability of Human Cerebellar Neurons to Degeneration in Ataxia-Causing Channelopathies. Front Syst Neurosci 2022; 16:908569. [PMID: 35757096 PMCID: PMC9219590 DOI: 10.3389/fnsys.2022.908569] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023] Open
Abstract
Mutations in ion channel genes underlie a number of human neurological diseases. Historically, human mutations in ion channel genes, the so-called channelopathies, have been identified to cause episodic disorders. In the last decade, however, mutations in ion channel genes have been demonstrated to result in progressive neurodegenerative and neurodevelopmental disorders in humans, particularly with ion channels that are enriched in the cerebellum. This was unexpected given prior rodent ion channel knock-out models that almost never display neurodegeneration. Human ataxia-causing channelopathies that result in even haploinsufficiency can result in cerebellar atrophy and cerebellar Purkinje neuron loss. Rodent neurons with ion channel loss-of-function appear to, therefore, be significantly more resistant to neurodegeneration compared to human neurons. Fundamental differences in susceptibility of human and rodent cerebellar neurons in ataxia-causing channelopathies must therefore be present. In this review, we explore the properties of human neurons that may contribute to their vulnerability to cerebellar degeneration secondary to ion channel loss-of-function mutations. We present a model taking into account the known allometric scaling of neuronal ion channel density in humans and other mammals that may explain the preferential vulnerability of human cerebellar neurons to degeneration in ataxia-causing channelopathies. We also speculate on the vulnerability of cerebellar neurons to degeneration in mouse models of spinocerebellar ataxia (SCA) where ion channel transcript dysregulation has recently been implicated in disease pathogenesis.
Collapse
Affiliation(s)
- David D. Bushart
- Ohio State University College of Medicine, Columbus, OH, United States
| | - Vikram G. Shakkottai
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States,*Correspondence: Vikram G. Shakkottai,
| |
Collapse
|
19
|
Cendelin J, Cvetanovic M, Gandelman M, Hirai H, Orr HT, Pulst SM, Strupp M, Tichanek F, Tuma J, Manto M. Consensus Paper: Strengths and Weaknesses of Animal Models of Spinocerebellar Ataxias and Their Clinical Implications. CEREBELLUM (LONDON, ENGLAND) 2022; 21:452-481. [PMID: 34378174 PMCID: PMC9098367 DOI: 10.1007/s12311-021-01311-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/21/2021] [Indexed: 01/02/2023]
Abstract
Spinocerebellar ataxias (SCAs) represent a large group of hereditary degenerative diseases of the nervous system, in particular the cerebellum, and other systems that manifest with a variety of progressive motor, cognitive, and behavioral deficits with the leading symptom of cerebellar ataxia. SCAs often lead to severe impairments of the patient's functioning, quality of life, and life expectancy. For SCAs, there are no proven effective pharmacotherapies that improve the symptoms or substantially delay disease progress, i.e., disease-modifying therapies. To study SCA pathogenesis and potential therapies, animal models have been widely used and are an essential part of pre-clinical research. They mainly include mice, but also other vertebrates and invertebrates. Each animal model has its strengths and weaknesses arising from model animal species, type of genetic manipulation, and similarity to human diseases. The types of murine and non-murine models of SCAs, their contribution to the investigation of SCA pathogenesis, pathological phenotype, and therapeutic approaches including their advantages and disadvantages are reviewed in this paper. There is a consensus among the panel of experts that (1) animal models represent valuable tools to improve our understanding of SCAs and discover and assess novel therapies for this group of neurological disorders characterized by diverse mechanisms and differential degenerative progressions, (2) thorough phenotypic assessment of individual animal models is required for studies addressing therapeutic approaches, (3) comparative studies are needed to bring pre-clinical research closer to clinical trials, and (4) mouse models complement cellular and invertebrate models which remain limited in terms of clinical translation for complex neurological disorders such as SCAs.
Collapse
Affiliation(s)
- Jan Cendelin
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 75, 323 00, Plzen, Czech Republic.
- Laboratory of Neurodegenerative Disorders, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 75, 323 00, Plzen, Czech Republic.
| | - Marija Cvetanovic
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Mandi Gandelman
- Department of Neurology, University of Utah, 175 North Medical Drive East, Salt Lake City, UT, 84132, USA
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, 3-39-22, Gunma, 371-8511, Japan
- Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Gunma, 371-8511, Japan
| | - Harry T Orr
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, 175 North Medical Drive East, Salt Lake City, UT, 84132, USA
| | - Michael Strupp
- Department of Neurology and German Center for Vertigo and Balance Disorders, Hospital of the Ludwig-Maximilians University, Munich, Campus Grosshadern, Marchioninistr. 15, 81377, Munich, Germany
| | - Filip Tichanek
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 75, 323 00, Plzen, Czech Republic
- Laboratory of Neurodegenerative Disorders, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 75, 323 00, Plzen, Czech Republic
| | - Jan Tuma
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 75, 323 00, Plzen, Czech Republic
- The Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, MC 7843, San Antonio, TX, 78229, USA
| | - Mario Manto
- Unité des Ataxies Cérébelleuses, Service de Neurologie, CHU-Charleroi, Charleroi, Belgium
- Service des Neurosciences, Université de Mons, UMons, Mons, Belgium
| |
Collapse
|
20
|
Martinez-Rojas VA, Juarez-Hernandez LJ, Musio C. Ion channels and neuronal excitability in polyglutamine neurodegenerative diseases. Biomol Concepts 2022; 13:183-199. [DOI: 10.1515/bmc-2022-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Polyglutamine (polyQ) diseases are a family composed of nine neurodegenerative inherited disorders (NDDs) caused by pathological expansions of cytosine-adenine-guanine (CAG) trinucleotide repeats which encode a polyQ tract in the corresponding proteins. CAG polyQ repeat expansions produce neurodegeneration via multiple downstream mechanisms; among those the neuronal activity underlying the ion channels is affected directly by specific channelopathies or indirectly by secondary dysregulation. In both cases, the altered excitability underlies to gain- or loss-of-function pathological effects. Here we summarize the repertoire of ion channels in polyQ NDDs emphasizing the biophysical features of neuronal excitability and their pathogenic role. The aim of this review is to point out the value of a deeper understanding of those functional mechanisms and processes as crucial elements for the designing and targeting of novel therapeutic avenues.
Collapse
Affiliation(s)
- Vladimir A. Martinez-Rojas
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) , Via Sommarive 18 , 38123 Trento , Italy
| | - Leon J. Juarez-Hernandez
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) , Via Sommarive 18 , 38123 Trento , Italy
| | - Carlo Musio
- Institute of Biophysics (IBF), Trento Unit, National Research Council (CNR) , Via Sommarive 18 , 38123 Trento , Italy
| |
Collapse
|
21
|
Ha H, Kim M, Chung B, Lee CH, Oh SH, Kang H, Kwon OY. A Novel KCND3 Variant in a Korean Family With SCA19. J Clin Neurol 2022; 18:90-92. [PMID: 35021282 PMCID: PMC8762501 DOI: 10.3988/jcn.2022.18.1.90] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 12/05/2022] Open
Affiliation(s)
- Hongmin Ha
- Department of Neurology, Gyeongsang National University Hospital, Jinju, Korea
| | - Minkyeong Kim
- Department of Neurology, Gyeongsang National University Hospital, Jinju, Korea.
| | - Bora Chung
- Department of Neurology, Gyeongsang National University Hospital, Jinju, Korea
| | - Chan Hyun Lee
- Department of Neurology, Gyeongsang National University Hospital, Jinju, Korea
| | - Seung Hwan Oh
- Department of Laboratory Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Heeyoung Kang
- Department of Neurology, Gyeongsang National University Hospital, Jinju, Korea.,Department of Neurology, Gyeongsang National University College of Medicine, Jinju, Korea.,Institute of Health Science, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Oh-Young Kwon
- Department of Neurology, Gyeongsang National University Hospital, Jinju, Korea.,Department of Neurology, Gyeongsang National University College of Medicine, Jinju, Korea.,Institute of Health Science, Gyeongsang National University College of Medicine, Jinju, Korea.
| |
Collapse
|
22
|
Corral-Juan M, Casquero P, Giraldo-Restrepo N, Laurie S, Martinez-Piñeiro A, Mateo-Montero RC, Ispierto L, Vilas D, Tolosa E, Volpini V, Alvarez-Ramo R, Sánchez I, Matilla-Dueñas A. OUP accepted manuscript. Brain Commun 2022; 4:fcac030. [PMID: 35310830 PMCID: PMC8928420 DOI: 10.1093/braincomms/fcac030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/20/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Spinocerebellar ataxias consist of a highly heterogeneous group of inherited movement disorders clinically characterized by progressive cerebellar ataxia variably associated with additional distinctive clinical signs. The genetic heterogeneity is evidenced by the myriad of associated genes and underlying genetic defects identified. In this study, we describe a new spinocerebellar ataxia subtype in nine members of a Spanish five-generation family from Menorca with affected individuals variably presenting with ataxia, nystagmus, dysarthria, polyneuropathy, pyramidal signs, cerebellar atrophy and distinctive cerebral demyelination. Affected individuals presented with horizontal and vertical gaze-evoked nystagmus and hyperreflexia as initial clinical signs, and a variable age of onset ranging from 12 to 60 years. Neurophysiological studies showed moderate axonal sensory polyneuropathy with altered sympathetic skin response predominantly in the lower limbs. We identified the c.1877C > T (p.Ser626Leu) pathogenic variant within the SAMD9L gene as the disease causative genetic defect with a significant log-odds score (Zmax = 3.43; θ = 0.00; P < 3.53 × 10−5). We demonstrate the mitochondrial location of human SAMD9L protein, and its decreased levels in patients’ fibroblasts in addition to mitochondrial perturbations. Furthermore, mutant SAMD9L in zebrafish impaired mobility and vestibular/sensory functions. This study describes a novel spinocerebellar ataxia subtype caused by SAMD9L mutation, SCA49, which triggers mitochondrial alterations pointing to a role of SAMD9L in neurological motor and sensory functions.
Collapse
Affiliation(s)
- Marc Corral-Juan
- Functional and Translational Neurogenetics Unit, Department of Neuroscience, Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona-Can Ruti Campus, Badalona, Barcelona, Spain
| | - Pilar Casquero
- Neurology and Neurophysiology Section, Hospital Mateu Orfila, Mahón, Menorca, Spain
| | | | - Steve Laurie
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Alicia Martinez-Piñeiro
- Neuromuscular and Functional Studies Unit, Neurology Service, University Hospital Germans Trias i Pujol (HUGTiP), Universitat Autònoma de Barcelona-Can Ruti Campus, Badalona, Barcelona, Spain
| | | | - Lourdes Ispierto
- Neurodegenerative Diseases Unit, Neurology Service, Department of Neuroscience, University Hospital Germans Trias i Pujol (HUGTiP), Universitat Autònoma de Barcelona-Can Ruti Campus, Badalona, Barcelona, Spain
| | - Dolores Vilas
- Neurodegenerative Diseases Unit, Neurology Service, Department of Neuroscience, University Hospital Germans Trias i Pujol (HUGTiP), Universitat Autònoma de Barcelona-Can Ruti Campus, Badalona, Barcelona, Spain
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Spain
| | - Eduardo Tolosa
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Spain
| | | | - Ramiro Alvarez-Ramo
- Neurodegenerative Diseases Unit, Neurology Service, Department of Neuroscience, University Hospital Germans Trias i Pujol (HUGTiP), Universitat Autònoma de Barcelona-Can Ruti Campus, Badalona, Barcelona, Spain
| | - Ivelisse Sánchez
- Functional and Translational Neurogenetics Unit, Department of Neuroscience, Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona-Can Ruti Campus, Badalona, Barcelona, Spain
| | - Antoni Matilla-Dueñas
- Functional and Translational Neurogenetics Unit, Department of Neuroscience, Research Institute Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona-Can Ruti Campus, Badalona, Barcelona, Spain
- Correspondence to: Dr Antoni Matilla-Dueñas Head of the Neurogenetics Unit Health Sciences Research Institute Germans Trias i Pujol (IGTP) Ctra. de Can Ruti, Camí de les Escoles s/n 08916 Badalona, Barcelona, Spain E-mail:
| |
Collapse
|
23
|
Abstract
The term SCA refers to a phenotypically and genetically heterogeneous group of autosomal dominant spinocerebellar ataxias. Phenotypically they present as gait ataxia frequently in combination with dysarthria and oculomotor problems. Additional signs and symptoms are common and can include various pyramidal and extrapyramidal signs and intellectual impairment. Genetic causes of SCAs are either repeat expansions within disease genes or common mutations (point mutations, deletions, insertions etc.). Frequently the two types of mutations cause indistinguishable phenotypes (locus heterogeneity). This article focuses on SCAs caused by common mutations. It describes phenotype and genotype of the presently 27 types known and discusses the molecular pathogenesis in those 21 types where the disease gene has been identified. Apart from the dominant types, the article also summarizes findings in a variant caused by mutations in a mitochondrial gene. Possible common disease mechanisms are considered based on findings in the various SCAs described.
Collapse
Affiliation(s)
- Ulrich Müller
- Institute of Human Genetics, JLU-Gießen, Schlangenzahl 14, 35392, Giessen, Germany.
| |
Collapse
|
24
|
NGS in Hereditary Ataxia: When Rare Becomes Frequent. Int J Mol Sci 2021; 22:ijms22168490. [PMID: 34445196 PMCID: PMC8395181 DOI: 10.3390/ijms22168490] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022] Open
Abstract
The term hereditary ataxia (HA) refers to a heterogeneous group of neurological disorders with multiple genetic etiologies and a wide spectrum of ataxia-dominated phenotypes. Massive gene analysis in next-generation sequencing has entered the HA scenario, broadening our genetic and clinical knowledge of these conditions. In this study, we employed a targeted resequencing panel (TRP) in a large and highly heterogeneous cohort of 377 patients with a clinical diagnosis of HA, but no molecular diagnosis on routine genetic tests. We obtained a positive result (genetic diagnosis) in 33.2% of the patients, a rate significantly higher than those reported in similar studies employing TRP (average 19.4%), and in line with those performed using exome sequencing (ES, average 34.6%). Moreover, 15.6% of the patients had an uncertain molecular diagnosis. STUB1, PRKCG, and SPG7 were the most common causative genes. A comparison with published literature data showed that our panel would have identified 97% of the positive cases reported in previous TRP-based studies and 92% of those diagnosed by ES. Proper use of multigene panels, when combined with detailed phenotypic data, seems to be even more efficient than ES in clinical practice.
Collapse
|
25
|
Rare Gain-of-Function KCND3 Variant Associated with Cerebellar Ataxia, Parkinsonism, Cognitive Dysfunction, and Brain Iron Accumulation. Int J Mol Sci 2021; 22:ijms22158247. [PMID: 34361012 PMCID: PMC8347726 DOI: 10.3390/ijms22158247] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/08/2023] Open
Abstract
Loss-of-function mutations in the KV4.3 channel-encoding KCND3 gene are linked to neurodegenerative cerebellar ataxia. Patients suffering from neurodegeneration associated with iron deposition may also present with cerebellar ataxia. The mechanism underlying brain iron accumulation remains unclear. Here, we aim to ascertain the potential pathogenic role of KCND3 variant in iron accumulation-related cerebellar ataxia. We presented a patient with slowly progressive cerebellar ataxia, parkinsonism, cognitive impairment, and iron accumulation in the basal ganglia and the cerebellum. Whole exome sequencing analyses identified in the patient a heterozygous KCND3 c.1256G>A (p.R419H) variant predicted to be disease-causing by multiple bioinformatic analyses. In vitro biochemical and immunofluorescence examinations revealed that, compared to the human KV4.3 wild-type channel, the p.R419H variant exhibited normal protein abundance and subcellular localization pattern. Electrophysiological investigation, however, demonstrated that the KV4.3 p.R419H variant was associated with a dominant increase in potassium current amplitudes, as well as notable changes in voltage-dependent gating properties leading to enhanced potassium window current. These observations indicate that, in direct contrast with the loss-of-function KCND3 mutations previously reported in cerebellar ataxia patients, we identified a rare gain-of-function KCND3 variant that may expand the clinical and molecular spectra of neurodegenerative cerebellar disorders associated with brain iron accumulation.
Collapse
|
26
|
Novel KCND3 Variant Underlying Nonprogressive Congenital Ataxia or SCA19/22 Disrupt K V4.3 Protein Expression and K+ Currents with Variable Effects on Channel Properties. Int J Mol Sci 2021; 22:ijms22094986. [PMID: 34067185 PMCID: PMC8125845 DOI: 10.3390/ijms22094986] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023] Open
Abstract
KCND3 encodes the voltage-gated potassium channel KV4.3 that is highly expressed in the cerebellum, where it regulates dendritic excitability and calcium influx. Loss-of-function KV4.3 mutations have been associated with dominant spinocerebellar ataxia (SCA19/22). By targeted NGS sequencing, we identified two novel KCND3 missense variants of the KV4.3 channel: p.S347W identified in a patient with adult-onset pure cerebellar syndrome and p.W359G detected in a child with congenital nonprogressive ataxia. Neuroimaging showed mild cerebellar atrophy in both patients. We performed a two-electrode voltage-clamp recording of KV4.3 currents in Xenopus oocytes: both the p.G345V (previously reported in a SCA19/22 family) and p.S347W mutants exhibited reduced peak currents by 50%, while no K+ current was detectable for the p.W359G mutant. We assessed the effect of the mutations on channel gating by measuring steady-state voltage-dependent activation and inactivation properties: no significant alterations were detected in p.G345V and p.S347W disease-associated variants, compared to controls. KV4.3 expression studies in HEK293T cells showed 53% (p.G345V), 45% (p.S347W) and 75% (p.W359G) reductions in mutant protein levels compared with the wildtype. The present study broadens the spectrum of the known phenotypes and identifies additional variants for KCND3-related disorders, outlining the importance of SCA gene screening in early-onset and congenital ataxia.
Collapse
|
27
|
Nakajima T, Tamura S, Kurabayashi M, Kaneko Y. Towards Mutation-Specific Precision Medicine in Atypical Clinical Phenotypes of Inherited Arrhythmia Syndromes. Int J Mol Sci 2021; 22:ijms22083930. [PMID: 33920294 PMCID: PMC8069124 DOI: 10.3390/ijms22083930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/08/2021] [Indexed: 12/19/2022] Open
Abstract
Most causal genes for inherited arrhythmia syndromes (IASs) encode cardiac ion channel-related proteins. Genotype-phenotype studies and functional analyses of mutant genes, using heterologous expression systems and animal models, have revealed the pathophysiology of IASs and enabled, in part, the establishment of causal gene-specific precision medicine. Additionally, the utilization of induced pluripotent stem cell (iPSC) technology have provided further insights into the pathophysiology of IASs and novel promising therapeutic strategies, especially in long QT syndrome. It is now known that there are atypical clinical phenotypes of IASs associated with specific mutations that have unique electrophysiological properties, which raises a possibility of mutation-specific precision medicine. In particular, patients with Brugada syndrome harboring an SCN5A R1632C mutation exhibit exercise-induced cardiac events, which may be caused by a marked activity-dependent loss of R1632C-Nav1.5 availability due to a marked delay of recovery from inactivation. This suggests that the use of isoproterenol should be avoided. Conversely, the efficacy of β-blocker needs to be examined. Patients harboring a KCND3 V392I mutation exhibit both cardiac (early repolarization syndrome and paroxysmal atrial fibrillation) and cerebral (epilepsy) phenotypes, which may be associated with a unique mixed electrophysiological property of V392I-Kv4.3. Since the epileptic phenotype appears to manifest prior to cardiac events in this mutation carrier, identifying KCND3 mutations in patients with epilepsy and providing optimal therapy will help prevent sudden unexpected death in epilepsy. Further studies using the iPSC technology may provide novel insights into the pathophysiology of atypical clinical phenotypes of IASs and the development of mutation-specific precision medicine.
Collapse
|
28
|
Bushart DD, Zalon AJ, Zhang H, Morrison LM, Guan Y, Paulson HL, Shakkottai VG, McLoughlin HS. Antisense Oligonucleotide Therapy Targeted Against ATXN3 Improves Potassium Channel-Mediated Purkinje Neuron Dysfunction in Spinocerebellar Ataxia Type 3. CEREBELLUM (LONDON, ENGLAND) 2021; 20:41-53. [PMID: 32789747 PMCID: PMC7930886 DOI: 10.1007/s12311-020-01179-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Spinocerebellar ataxia type 3 (SCA3) is the second-most common CAG repeat disease, caused by a glutamine-encoding expansion in the ATXN3 protein. SCA3 is characterized by spinocerebellar degeneration leading to progressive motor incoordination and early death. Previous studies suggest that potassium channel dysfunction underlies early abnormalities in cerebellar cortical Purkinje neuron firing in SCA3. However, cerebellar cortical degeneration is often modest both in the human disease and mouse models of SCA3, raising uncertainty about the role of cerebellar dysfunction in SCA3. Here, we address this question by investigating Purkinje neuron excitability in SCA3. In early-stage SCA3 mice, we confirm a previously identified increase in excitability of cerebellar Purkinje neurons and associate this excitability with reduced transcripts of two voltage-gated potassium (KV) channels, Kcna6 and Kcnc3, as well as motor impairment. Intracerebroventricular delivery of antisense oligonucleotides (ASO) to reduce mutant ATXN3 restores normal excitability to SCA3 Purkinje neurons and rescues transcript levels of Kcna6 and Kcnc3. Interestingly, while an even broader range of KV channel transcripts shows reduced levels in late-stage SCA3 mice, cerebellar Purkinje neuron physiology was not further altered despite continued worsening of motor impairment. These results suggest the progressive motor phenotype observed in SCA3 may not reflect ongoing changes in the cerebellar cortex but instead dysfunction of other neuronal structures within and beyond the cerebellum. Nevertheless, the early rescue of both KV channel expression and neuronal excitability by ASO treatment suggests that cerebellar cortical dysfunction contributes meaningfully to motor dysfunction in SCA3.
Collapse
Affiliation(s)
- David D. Bushart
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Annie J. Zalon
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Hongjiu Zhang
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109,Microsoft, Inc. Bellevue, WA 98004
| | - Logan M. Morrison
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109
| | - Yuanfang Guan
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | - Henry L. Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| | - Vikram G. Shakkottai
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109,Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109,Address correspondence to: Vikram G. Shakkottai, 4009 BSRB, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, ; Hayley S. McLoughlin, 4017 BSRB, 109 Zina Pitcher Pl., Ann Arbor, MI 48109,
| | - Hayley S. McLoughlin
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109,Address correspondence to: Vikram G. Shakkottai, 4009 BSRB, 109 Zina Pitcher Pl., Ann Arbor, MI 48109, ; Hayley S. McLoughlin, 4017 BSRB, 109 Zina Pitcher Pl., Ann Arbor, MI 48109,
| |
Collapse
|
29
|
Paucar M, Ågren R, Li T, Lissmats S, Bergendal Å, Weinberg J, Nilsson D, Savichetva I, Sahlholm K, Nilsson J, Svenningsson P. V374A KCND3 Pathogenic Variant Associated With Paroxysmal Ataxia Exacerbations. NEUROLOGY-GENETICS 2021; 7:e546. [PMID: 33575485 PMCID: PMC7862093 DOI: 10.1212/nxg.0000000000000546] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/27/2020] [Indexed: 12/27/2022]
Abstract
Objective Ataxia channelopathies share common features such as slow motor progression and variable degrees of cognitive dysfunction. Mutations in potassium voltage-gated channel subfamily D member 3 (KCND3), encoding the K+ channel, Kv4.3, are associated with spinocerebellar ataxia (SCA) 19, allelic with SCA22. Mutations in potassium voltage-gated channel subfamily C member 3 (KCNC3), encoding another K+ channel, Kv3.3, cause SCA13. First, a comprehensive phenotype assessment was carried out in a family with autosomal dominant ataxia harboring 2 genetic variants in KCNC3 and KCND3. To evaluate the physiological impact of these variants on channel currents, in vitro studies were performed. Methods Clinical and psychometric evaluations, neuroimaging, and genotyping of a family (mother and son) affected by ataxia were carried out. Heterozygous and homozygous Kv3.3 A671V and Kv4.3 V374A variants were evaluated in Xenopus laevis oocytes using 2-electrode voltage-clamp. The influence of Kv4 conductance on neuronal activity was investigated computationally using a Purkinje neuron model. Results The main clinical findings were consistent with adult-onset ataxia with cognitive dysfunction and acetazolamide-responsive paroxysmal motor exacerbations in the index case. Despite cognitive deficits, fluorodeoxyglucose (FDG)-PET displayed hypometabolism mainly in the severely atrophic cerebellum. Genetic analyses revealed the new variant c.1121T>C (V374A) in KCND3 and c.2012T>C (A671V) in KCNC3. In vitro electrophysiology experiments on Xenopus oocytes demonstrated that the V374A mutant was nonfunctional when expressed on its own. Upon equal co-expression of wild-type (WT) and V374A channel subunits, Kv4.3 currents were significantly reduced in a dominant negative manner, without alterations of the gating properties of the channel. By contrast, Kv3.3 A671V, when expressed alone, exhibited moderately reduced currents compared with WT, with no effects on channel activation or inactivation. Immunohistochemistry demonstrated adequate cell membrane translocation of the Kv4.3 V374A variant, thus suggesting an impairment of channel function, rather than of expression. Computational modeling predicted an increased Purkinje neuron firing frequency upon reduced Kv4.3 conductance. Conclusions Our findings suggest that Kv4.3 V374A is likely pathogenic and associated with paroxysmal ataxia exacerbations, a new trait for SCA19/22. The present FDG PET findings contrast with a previous study demonstrating widespread brain hypometabolism in SCA19/22.
Collapse
Affiliation(s)
- Martin Paucar
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| | - Richard Ågren
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| | - Tianyi Li
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| | - Simon Lissmats
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| | - Åsa Bergendal
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| | - Jan Weinberg
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| | - Daniel Nilsson
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| | - Irina Savichetva
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| | - Kristoffer Sahlholm
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| | - Johanna Nilsson
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience (M.P., R.Å., T.L., Å.B., J.N., P.S.), Department of Molecular Medicine and Surgery (D.N.), Center for Molecular Medicine (D.N.), and Science for Life Laboratory (D.N.), Karolinska Institutet (S.L., I.S.), Stockholm; Department of Neurology (M.P., J.W., P.S.), Department of Clinical Genetics (D.N.), Department of Nuclear Medicine (I.S.), and Department of Neurophysiology (J.N.), Karolinska University Hospital (R.Å.), Stockholm; Department of Integrative Medical Biology (K.S.), Umeå University; and Department of Medical Sciences (J.N.), Örebro University, Sweden
| |
Collapse
|
30
|
Li Z, Feng J, Yuan Z. Key Modules and Hub Genes Identified by Coexpression Network Analysis for Revealing Novel Biomarkers for Spina Bifida. Front Genet 2020; 11:583316. [PMID: 33343629 PMCID: PMC7738565 DOI: 10.3389/fgene.2020.583316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/09/2020] [Indexed: 11/13/2022] Open
Abstract
Spina bifida is a common neural tube defect (NTD) accounting for 5–10% of perinatal mortalities. As a polygenic disease, spina bifida is caused by a combination of genetic and environmental factors, for which the precise molecular pathogenesis is still not systemically understood. In the present study, we aimed to identify the related gene module that might play a vital role in the occurrence and development of spina bifida by using weighted gene co-expression network analysis (WGCNA). Transcription profiling according to an array of human amniocytes from patients with spina bifida and healthy controls was downloaded from the Gene Expression Omnibus database. First, outliers were identified and removed by principal component analysis (PCA) and sample clustering. Then, genes in the top 25% of variance in the GSE4182 dataset were then determined in order to explore candidate genes in potential hub modules using WGCNA. After data preprocessing, 5407 genes were obtained for further WGCNA. Highly correlated genes were divided into nineteen modules. Combined with a co-expression network and significant differentially expressed genes, 967 candidate genes were identified that may be involved in the pathological processes of spina bifida. Combined with our previous microRNA (miRNA) microarray results, we constructed an miRNA–mRNA network including four miRNAs and 39 mRNA among which three key genes were, respectively, linked to two miRNA-associated gene networks. Following the verification of qRT-PCR and KCND3 was upregulated in the spina bifida. KCND3 and its related miR-765 and miR-142-3p are worthy of further study. These findings may be conducive for early detection and intervention in spina bifida, as well as be of great significance to pregnant women and clinical staff.
Collapse
Affiliation(s)
- Zijian Li
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China.,Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
31
|
Xu Y, Wu Z, Liu L, Liu J, Wang Y. Rat Model of Cockayne Syndrome Neurological Disease. Cell Rep 2020; 29:800-809.e5. [PMID: 31644904 DOI: 10.1016/j.celrep.2019.09.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 08/26/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022] Open
Abstract
Cockayne syndrome (CS) is a rare genetic neurodevelopmental disorder, characterized by a deficiency in transcription-coupled subpathway of nucleotide excision DNA repair (TC-NER). Mutation of the Cockayne syndrome B (CSB) gene affects basal transcription, which is considered a major cause of CS neurologic dysfunction. Here, we generate a rat model by mimicking a nonsense mutation in the CSB gene. In contrast to that of the Csb-/- mouse models, the brains of the CSB-deficient rats are more profoundly affected. The cerebellar cortex shows significant atrophy and dysmyelination. Aberrant foliation of the cerebellum and deformed hippocampus are visible. The white matter displays high glial fibrillary acidic protein (GFAP) staining indicative of reactive astrogliosis. RNA sequencing (RNA-seq) analysis reveals that CSB deficiency affects the expression of hundreds of genes, many of which are neuronal genes, suggesting that transcription dysregulation could contribute to the neurologic disease seen in the CSB rat models.
Collapse
Affiliation(s)
- Yingying Xu
- Key Laboratory of Neurological Function and Health, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhenzhen Wu
- Key Laboratory of Neurological Function and Health, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Lingyun Liu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiena Liu
- Key Laboratory of Neurological Function and Health, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Yuming Wang
- Key Laboratory of Neurological Function and Health, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
32
|
Nakajima T, Kawabata-Iwakawa R, Kaneko Y, Hamano SI, Sano R, Tamura S, Hasegawa H, Kobari T, Kominato Y, Nishiyama M, Kurabayashi M. Novel Cardiocerebral Channelopathy Associated with a KCND3 V392I Mutation. Int Heart J 2020; 61:1049-1055. [PMID: 32921676 DOI: 10.1536/ihj.20-203] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
While a KCND3 V392I mutation uniquely displays a mixed electrophysiological phenotype of Kv4.3, only limited clinical information on the mutation carriers is available. We report two teenage siblings exhibiting both cardiac (early repolarization syndrome and paroxysmal atrial fibrillation) and cerebral phenotypes (epilepsy and intellectual disability), in whom we identified the KCND3 V392I mutation. We propose a link between the KCND3 mutation with a mixed electrophysiological phenotype and cardiocerebral phenotypes, which may be defined as a novel cardiocerebral channelopathy.
Collapse
Affiliation(s)
- Tadashi Nakajima
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
| | - Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research
| | - Yoshiaki Kaneko
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
| | | | - Rie Sano
- Department of Legal Medicine, Gunma University Graduate School of Medicine
| | - Shuntaro Tamura
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
| | - Hiroshi Hasegawa
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
| | - Takashi Kobari
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
| | - Yoshihiko Kominato
- Department of Legal Medicine, Gunma University Graduate School of Medicine
| | - Masahiko Nishiyama
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research.,Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine
| | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine
| |
Collapse
|
33
|
KCND3-Related Neurological Disorders: From Old to Emerging Clinical Phenotypes. Int J Mol Sci 2020; 21:ijms21165802. [PMID: 32823520 PMCID: PMC7461103 DOI: 10.3390/ijms21165802] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/06/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
KCND3 encodes the voltage-gated potassium ion channel subfamily D member 3, a six trans-membrane protein (Kv4.3), involved in the transient outward K+ current. KCND3 defect causes both cardiological and neurological syndromes. From a neurological perspective, Kv4.3 defect has been associated to SCA type 19/22, a complex neurological disorder encompassing a wide spectrum of clinical features beside ataxia. To better define the phenotypic spectrum and course of KCND3-related neurological disorder, we review the clinical presentation and evolution in 68 reported cases. We delineated two main clinical phenotypes according to the age of onset. Neurodevelopmental disorder with epilepsy and/or movement disorders with ataxia later in the disease course characterized the early onset forms, while a prominent ataxic syndrome with possible cognitive decline, movement disorders, and peripheral neuropathy were observed in the late onset forms. Furthermore, we described a 37-year-old patient with a de novo KCND3 variant [c.901T>C (p.Ser301Pro)], previously reported in dbSNP as rs79821338, and a clinical phenotype paradigmatic of the early onset forms with neurodevelopmental disorder, epilepsy, parkinsonism-dystonia, and ataxia in adulthood, further expanding the clinical spectrum of this condition.
Collapse
|
34
|
Inter-Regulation of K v4.3 and Voltage-Gated Sodium Channels Underlies Predisposition to Cardiac and Neuronal Channelopathies. Int J Mol Sci 2020; 21:ijms21145057. [PMID: 32709127 PMCID: PMC7404392 DOI: 10.3390/ijms21145057] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/04/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Genetic variants in voltage-gated sodium channels (Nav) encoded by SCNXA genes, responsible for INa, and Kv4.3 channels encoded by KCND3, responsible for the transient outward current (Ito), contribute to the manifestation of both Brugada syndrome (BrS) and spinocerebellar ataxia (SCA19/22). We examined the hypothesis that Kv4.3 and Nav variants regulate each other’s function, thus modulating INa/Ito balance in cardiomyocytes and INa/I(A) balance in neurons. Methods: Bicistronic and other constructs were used to express WT or variant Nav1.5 and Kv4.3 channels in HEK293 cells. INa and Ito were recorded. Results: SCN5A variants associated with BrS reduced INa, but increased Ito. Moreover, BrS and SCA19/22 KCND3 variants associated with a gain of function of Ito, significantly reduced INa, whereas the SCA19/22 KCND3 variants associated with a loss of function (LOF) of Ito significantly increased INa. Auxiliary subunits Navβ1, MiRP3 and KChIP2 also modulated INa/Ito balance. Co-immunoprecipitation and Duolink studies suggested that the two channels interact within the intracellular compartments and biotinylation showed that LOF SCN5A variants can increase Kv4.3 cell-surface expression. Conclusion: Nav and Kv4.3 channels modulate each other’s function via trafficking and gating mechanisms, which have important implications for improved understanding of these allelic cardiac and neuronal syndromes.
Collapse
|
35
|
Robinson KJ, Watchon M, Laird AS. Aberrant Cerebellar Circuitry in the Spinocerebellar Ataxias. Front Neurosci 2020; 14:707. [PMID: 32765211 PMCID: PMC7378801 DOI: 10.3389/fnins.2020.00707] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022] Open
Abstract
The spinocerebellar ataxias (SCAs) are a heterogeneous group of neurodegenerative diseases that share convergent disease features. A common symptom of these diseases is development of ataxia, involving impaired balance and motor coordination, usually stemming from cerebellar dysfunction and neurodegeneration. For most spinocerebellar ataxias, pathology can be attributed to an underlying gene mutation and the impaired function of the encoded protein through loss or gain-of-function effects. Strikingly, despite vast heterogeneity in the structure and function of disease-causing genes across the SCAs and the cellular processes affected, the downstream effects have considerable overlap, including alterations in cerebellar circuitry. Interestingly, aberrant function and degeneration of Purkinje cells, the major output neuronal population present within the cerebellum, precedes abnormalities in other neuronal populations within many SCAs, suggesting that Purkinje cells have increased vulnerability to cellular perturbations. Factors that are known to contribute to perturbed Purkinje cell function in spinocerebellar ataxias include altered gene expression resulting in altered expression or functionality of proteins and channels that modulate membrane potential, downstream impairments in intracellular calcium homeostasis and changes in glutamatergic input received from synapsing climbing or parallel fibers. This review will explore this enhanced vulnerability and the aberrant cerebellar circuitry linked with it in many forms of SCA. It is critical to understand why Purkinje cells are vulnerable to such insults and what overlapping pathogenic mechanisms are occurring across multiple SCAs, despite different underlying genetic mutations. Enhanced understanding of disease mechanisms will facilitate the development of treatments to prevent or slow progression of the underlying neurodegenerative processes, cerebellar atrophy and ataxic symptoms.
Collapse
Affiliation(s)
| | | | - Angela S. Laird
- Centre for Motor Neuron Disease Research, Department of Biomedical Science, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
36
|
Binda F, Pernaci C, Saxena S. Cerebellar Development and Circuit Maturation: A Common Framework for Spinocerebellar Ataxias. Front Neurosci 2020; 14:293. [PMID: 32300292 PMCID: PMC7145357 DOI: 10.3389/fnins.2020.00293] [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: 12/22/2019] [Accepted: 03/13/2020] [Indexed: 01/24/2023] Open
Abstract
Spinocerebellar ataxias (SCAs) affect the cerebellum and its afferent and efferent systems that degenerate during disease progression. In the cerebellum, Purkinje cells (PCs) are the most vulnerable and their prominent loss in the late phase of the pathology is the main characteristic of these neurodegenerative diseases. Despite the constant advancement in the discovery of affected molecules and cellular pathways, a comprehensive description of the events leading to the development of motor impairment and degeneration is still lacking. However, in the last years the possible causal role for altered cerebellar development and neuronal circuit wiring in SCAs has been emerging. Not only wiring and synaptic transmission deficits are a common trait of SCAs, but also preventing the expression of the mutant protein during cerebellar development seems to exert a protective role. By discussing this tight relationship between cerebellar development and SCAs, in this review, we aim to highlight the importance of cerebellar circuitry for the investigation of SCAs.
Collapse
Affiliation(s)
- Francesca Binda
- Department of Neurology, Center for Experimental Neurology, University Hospital of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Carla Pernaci
- Department of Neurology, Center for Experimental Neurology, University Hospital of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Smita Saxena
- Department of Neurology, Center for Experimental Neurology, University Hospital of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
37
|
Tada Y, Kume K, Matsuda Y, Kurashige T, Kanaya Y, Ohsawa R, Morino H, Tabu H, Kaneko S, Suenaga T, Kakizuka A, Kawakami H. Genetic screening for potassium channel mutations in Japanese autosomal dominant spinocerebellar ataxia. J Hum Genet 2020; 65:363-369. [PMID: 31907387 DOI: 10.1038/s10038-019-0717-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 12/04/2019] [Accepted: 12/19/2019] [Indexed: 01/17/2023]
Abstract
Spinocerebellar ataxia (SCA) is a genetically heterogeneous disease characterized by cerebellar ataxia. Many causative genes have been identified to date, the most common etiology being the abnormal expansion of repeat sequences, and the mutation of ion channel genes also play an important role in the development of SCA. Some of them encode calcium and potassium channels. However, due to limited reports about potassium genes in SCA, we screened 192 Japanese individuals with dominantly inherited SCA who had no abnormal repeat expansions of causative genes for potassium channel mutations (KCNC3 for SCA13 and KCND3 for SCA19/SCA22) by target sequencing. As a result, two variants were identified from two patients: c.1973G>A, p.R658Q and c.1018G>A, p.V340M for KCNC3, and no pathogenic variant was identified for KCND3. The newly identified p.V340M exists in the extracellular domain, and p.R658Q exists in the intracellular domain on the C-terminal side, although most of the reported KCNC3 mutations are present at the transmembrane site. Adult-onset and slowly progressive cerebellar ataxia are the main clinical features of SCA13 and SCA19 caused by potassium channel mutations, which was similar in our cases. SCA13 caused by KCNC3 mutations may present with deep sensory loss and cognitive impairment in addition to cerebellar ataxia. In this study, mild deep sensory loss was observed in one case. SCA caused by potassium channel gene mutations is extremely rare, and more cases should be accumulated in the future to elucidate its pathogenesis due to channel dysfunction.
Collapse
Affiliation(s)
- Yui Tada
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.,Laboratory of Functional Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kodai Kume
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yukiko Matsuda
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Takashi Kurashige
- Department of Neurology, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Kure, Japan
| | - Yuhei Kanaya
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Ryosuke Ohsawa
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Hiroyuki Morino
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Hayato Tabu
- Department of Neurology, Kitano Hospital, Osaka, Japan
| | - Satoshi Kaneko
- Department of Neurology, Kansai Medical University, Osaka, Japan
| | | | - Akira Kakizuka
- Laboratory of Functional Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hideshi Kawakami
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
38
|
Jenkins CA, Kalmar L, Matiasek K, Mari L, Kyöstilä K, Lohi H, Schofield EC, Mellersh CS, De Risio L, Ricketts SL. Characterisation of canine KCNIP4: A novel gene for cerebellar ataxia identified by whole-genome sequencing two affected Norwegian Buhund dogs. PLoS Genet 2020; 16:e1008527. [PMID: 31999692 PMCID: PMC7012447 DOI: 10.1371/journal.pgen.1008527] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/11/2020] [Accepted: 11/15/2019] [Indexed: 12/14/2022] Open
Abstract
A form of hereditary cerebellar ataxia has recently been described in the Norwegian Buhund dog breed. This study aimed to identify the genetic cause of the disease. Whole-genome sequencing of two Norwegian Buhund siblings diagnosed with progressive cerebellar ataxia was carried out, and sequences compared with 405 whole genome sequences of dogs of other breeds to filter benign common variants. Nine variants predicted to be deleterious segregated among the genomes in concordance with an autosomal recessive mode of inheritance, only one of which segregated within the breed when genotyped in additional Norwegian Buhunds. In total this variant was assessed in 802 whole genome sequences, and genotyped in an additional 505 unaffected dogs (including 146 Buhunds), and only four affected Norwegian Buhunds were homozygous for the variant. The variant identified, a T to C single nucleotide polymorphism (SNP) (NC_006585.3:g.88890674T>C), is predicted to cause a tryptophan to arginine substitution in a highly conserved region of the potassium voltage-gated channel interacting protein KCNIP4. This gene has not been implicated previously in hereditary ataxia in any species. Evaluation of KCNIP4 protein expression through western blot and immunohistochemical analysis using cerebellum tissue of affected and control dogs demonstrated that the mutation causes a dramatic reduction of KCNIP4 protein expression. The expression of alternative KCNIP4 transcripts within the canine cerebellum, and regional differences in KCNIP4 protein expression, were characterised through RT-PCR and immunohistochemistry respectively. The voltage-gated potassium channel protein KCND3 has previously been implicated in spinocerebellar ataxia, and our findings suggest that the Kv4 channel complex KCNIP accessory subunits also have an essential role in voltage-gated potassium channel function in the cerebellum and should be investigated as potential candidate genes for cerebellar ataxia in future studies in other species.
Collapse
Affiliation(s)
| | - Lajos Kalmar
- Department of Veterinary Medicine, University of Cambridge, Cambridge, Cambridgeshire, United Kingdom
| | - Kaspar Matiasek
- Section of Clinical & Comparative Neuropathology, Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität Munich, München, Germany
| | - Lorenzo Mari
- Neurology/Neurosurgery Service, Centre for Small Animal Studies, Animal Health Trust, Newmarket, Suffolk, United Kingdom
| | - Kaisa Kyöstilä
- Department of Veterinary Biosciences, and Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Hannes Lohi
- Department of Veterinary Biosciences, and Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Ellen C. Schofield
- Kennel Club Genetics Centre, Animal Health Trust, Newmarket, Suffolk, United Kingdom
| | - Cathryn S. Mellersh
- Kennel Club Genetics Centre, Animal Health Trust, Newmarket, Suffolk, United Kingdom
| | - Luisa De Risio
- Neurology/Neurosurgery Service, Centre for Small Animal Studies, Animal Health Trust, Newmarket, Suffolk, United Kingdom
| | - Sally L. Ricketts
- Kennel Club Genetics Centre, Animal Health Trust, Newmarket, Suffolk, United Kingdom
| |
Collapse
|
39
|
Catte A, Ferbel L, Bhattacharjee N, Jan Akhunzada M, D'Agostino T, Brancato G. In silico investigation of the interaction between the voltage-gated potassium channel Kv4.3 and its auxiliary protein KChIP1. Phys Chem Chem Phys 2019; 21:25290-25301. [PMID: 31701097 DOI: 10.1039/c9cp04082j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The voltage-gated potassium channel Kv4.3 plays a vital role in shaping the timing, frequency, and backpropagation of electrical signals in the brain and heart by generating fast transient currents at subthreshold membrane potentials in repetitive firing neurons. To achieve its physiological function, Kv4.3 is assisted by auxiliary β-subunits that become integral parts of the native A-type potassium channels, among which there are the Kv channel-interacting proteins (KChIPs). KChIPs are a family of cytosolic proteins that, when coexpressed with Kv4, lead to higher current density, modulation of channel inactivation and faster recovery from inactivation, while the loss of KChIP function may lead to severe pathological states. Recently, the structural basis of the KChIP1-Kv4.3 interaction was reported by using two similar X-ray crystallographic structures, which supported a crucial role for KChIP1 in enhancing the stability of the Kv4.3 tetrameric assembly, thus helping the trafficking of the channel to the plasma membrane. Here, we investigate through fully atomistic simulations the structure and stability of the human Kv4.3 tetramerization (T1) domain in complex with KChIP1 upon specific mutations located in the first and second interfaces of the complex, as compared to the wild-type (WT). Our results nicely complement the available structural and biophysical information collected so far on these complex variants. In particular, the degree of structural deviations and energetic instability, from small to substantial, observed in these variants with respect to the WT model seems to parallel well the level of channel dysfunction known from electrophysiology data. Our simulations provide an octameric structure of the WT KChIP1-Kv4.3 assembly very similar to the known crystal structures, and, at the same time, highlight the importance of a previously overlooked site of interaction between KChIP1 and the Kv4.3 T1 domain.
Collapse
Affiliation(s)
- Andrea Catte
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126 Pisa, Italy. and Istituto Nazionale di Fisica Nucleare, Largo Pontecorvo 3, I-56100 Pisa, Italy
| | - Letizia Ferbel
- Università di Pisa, Dipartimento di Ingegneria Civile ed Industriale, Largo Lucio Lazzarino 2, I-56124 Pisa, Italy
| | - Nicholus Bhattacharjee
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126 Pisa, Italy. and Istituto Nazionale di Fisica Nucleare, Largo Pontecorvo 3, I-56100 Pisa, Italy
| | - Muhammad Jan Akhunzada
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126 Pisa, Italy. and Istituto Nazionale di Fisica Nucleare, Largo Pontecorvo 3, I-56100 Pisa, Italy
| | - Tommaso D'Agostino
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126 Pisa, Italy. and Istituto Nazionale di Fisica Nucleare, Largo Pontecorvo 3, I-56100 Pisa, Italy
| | - Giuseppe Brancato
- Scuola Normale Superiore, Piazza dei Cavalieri 7, I-56126 Pisa, Italy. and Istituto Nazionale di Fisica Nucleare, Largo Pontecorvo 3, I-56100 Pisa, Italy
| |
Collapse
|
40
|
Jędrychowska J, Korzh V. Kv2.1 voltage-gated potassium channels in developmental perspective. Dev Dyn 2019; 248:1180-1194. [PMID: 31512327 DOI: 10.1002/dvdy.114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/01/2019] [Accepted: 09/03/2019] [Indexed: 11/11/2022] Open
Abstract
Kv2.1 voltage-gated potassium channels consist of two types of α-subunits: (a) electrically-active Kcnb1 α-subunits and (b) silent or modulatory α-subunits plus β-subunits that, similar to silent α-subunits, also regulate electrically-active subunits. Voltage-gated potassium channels were traditionally viewed, mainly by electrophysiologists, as regulators of the electrical activity of the plasma membrane in excitable cells, a role that is performed by transmembrane protein domains of α-subunits that form the electric pore. Genetic studies revealed a role for this region of α-subunits of voltage-gated potassium channels in human neurodevelopmental disorders, such as epileptic encephalopathy. The N- and C-terminal domains of α-subunits interact to form the cytoplasmic subunit of heterotetrameric potassium channels that regulate electric pores. Subsequent animal studies revealed the developmental functions of Kcnb1-containing voltage-gated potassium channels and illustrated their role during brain development and reproduction. These functions of potassium channels are discussed in this review in the context of regulatory interactions between electrically-active and regulatory subunits.
Collapse
Affiliation(s)
- Justyna Jędrychowska
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, Poland
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| |
Collapse
|
41
|
Hsiao CT, Fu SJ, Liu YT, Lu YH, Zhong CY, Tang CY, Soong BW, Jeng CJ. Novel SCA19/22-associated KCND3 mutations disrupt human K V 4.3 protein biosynthesis and channel gating. Hum Mutat 2019; 40:2088-2107. [PMID: 31293010 DOI: 10.1002/humu.23865] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/05/2019] [Accepted: 07/07/2019] [Indexed: 11/07/2022]
Abstract
Mutations in the human voltage-gated K+ channel subunit KV 4.3-encoding KCND3 gene have been associated with the autosomal dominant neurodegenerative disorder spinocerebellar ataxia types 19 and 22 (SCA19/22). The precise pathophysiology underlying the dominant inheritance pattern of SCA19/22 remains elusive. Using cerebellar ataxia-specific targeted next-generation sequencing technology, we identified two novel KCND3 mutations, c.950 G>A (p.C317Y) and c.1123 C>T (p.P375S) from a cohort with inherited cerebellar ataxias in Taiwan. The patients manifested notable phenotypic heterogeneity that includes cognitive impairment. We employed in vitro heterologous expression systems to inspect the biophysical and biochemical properties of human KV 4.3 harboring the two novel mutations, as well as two previously reported but uncharacterized disease-related mutations, c.1013 T>A (p.V338E) and c.1130 C>T (p.T377M). Electrophysiological analyses revealed that all of these SCA19/22-associated KV 4.3 mutant channels manifested loss-of-function phenotypes. Protein chemistry and immunofluorescence analyses further demonstrated that these mutants displayed enhanced protein degradation and defective membrane trafficking. By coexpressing KV 4.3 wild-type with the disease-related mutants, we provided direct evidence showing that the mutants instigated anomalous protein biosynthesis and channel gating of KV 4.3. We propose that the dominant inheritance pattern of SCA19/22 may be explained by the dominant-negative effects of the mutants on protein biosynthesis and voltage-dependent gating of KV 4.3 wild-type channel.
Collapse
Affiliation(s)
- Cheng-Tsung Hsiao
- Department of Internal Medicine, Taipei Veterans General Hospital Taoyuan Branch, Taoyuan, Taiwan
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Neurology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ssu-Ju Fu
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yo-Tsen Liu
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Neurology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Hsiang Lu
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ciao-Yu Zhong
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Yung Tang
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bing-Wen Soong
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Chung-Jiuan Jeng
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
42
|
Szpisjak L, Zadori D, Klivenyi P, Vecsei L. Clinical Characteristics and Possible Drug Targets in Autosomal Dominant Spinocerebellar Ataxias. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:279-293. [DOI: 10.2174/1871527318666190311155846] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/10/2018] [Accepted: 01/31/2019] [Indexed: 12/28/2022]
Abstract
Background & Objective:
The autosomal dominant spinocerebellar ataxias (SCAs) belong
to a large and expanding group of neurodegenerative disorders. SCAs comprise more than 40 subtypes
characterized by progressive ataxia as a common feature. The most prevalent diseases among SCAs
are caused by CAG repeat expansions in the coding-region of the causative gene resulting in polyglutamine
(polyQ) tract formation in the encoded protein. Unfortunately, there is no approved therapy to
treat cerebellar motor dysfunction in SCA patients. In recent years, several studies have been conducted
to recognize the clinical and pathophysiological aspects of the polyQ SCAs more accurately.
This scientific progress has provided new opportunities to develop promising gene therapies, including
RNA interference and antisense oligonucleotides.
Conclusion:
The aim of the current work is to give a brief summary of the clinical features of SCAs
and to review the cardinal points of pathomechanisms of the most common polyQ SCAs. In addition,
we review the last few year’s promising gene suppression therapies of the most frequent polyQ SCAs
in animal models, on the basis of which human trials may be initiated in the near future.
Collapse
Affiliation(s)
- Laszlo Szpisjak
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - Denes Zadori
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - Peter Klivenyi
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - Laszlo Vecsei
- Department of Neurology, University of Szeged, Szeged, Hungary
| |
Collapse
|
43
|
Novel De Novo KCND3 Mutation in a Japanese Patient with Intellectual Disability, Cerebellar Ataxia, Myoclonus, and Dystonia. THE CEREBELLUM 2019; 17:237-242. [PMID: 28895081 DOI: 10.1007/s12311-017-0883-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Spinocerebellar ataxia 19/22 (SCA19/22) is a rare type of autosomal dominant SCA that was previously described in 11 families. We report the case of a 30-year-old Japanese man presenting with intellectual disability, early onset cerebellar ataxia, myoclonus, and dystonia without a family history. MRI showed cerebellar atrophy, and electroencephalograms showed paroxysmal sharp waves during hyperventilation and photic stimulation. Trio whole-exome sequencing analysis of DNA samples from the patient and his parents revealed a de novo novel missense mutation (c.1150G>A, p.G384S) in KCND3, the causative gene of SCA19/22, substituting for evolutionally conserved glycine. The mutation was predicted to be functionally deleterious by bioinformatic analysis. Although pure cerebellar ataxia is the most common clinical feature in SCA19/22 families, extracerebellar symptoms including intellectual disability and myoclonus are reported in a limited number of families, suggesting a genotype-phenotype correlation for particular mutations. Although autosomal recessive diseases are more common in patients with early onset sporadic cerebellar ataxia, the present study emphasizes that such a possibility of de novo mutation should be considered.
Collapse
|
44
|
Abstract
Spinocerebellar ataxia type 19 (SCA19), allelic with spinocerebellar ataxia type 22 (SCA22), is a rare syndrome caused by mutations in the KCND3 gene which encodes the potassium channel Kv4.3. Only 18 SCA19/22 families and sporadic cases of different ethnic backgrounds have been previously reported. As in other SCAs, the SCA19/22 phenotype is variable and usually consists of adult-onset slowly progressive ataxia and cognitive impairment; myoclonus and seizures; mild Parkinsonism occurs in some cases. Here we describe a Swedish SCA19/22 family spanning five generations and harboring the T377M mutation in KCND3. For the first time for this disease, 18F-fluorodeoxyglucose PET was assessed revealing widespread brain hypometabolism. In addition, we identified white matter abnormalities and found unusual features for SCA19/22 including early age of onset and fast rate of progression in the late course of disease in the oldest patient of this family.
Collapse
|
45
|
Noh W, Pak S, Choi G, Yang S, Yang S. Transient Potassium Channels: Therapeutic Targets for Brain Disorders. Front Cell Neurosci 2019; 13:265. [PMID: 31263403 PMCID: PMC6585177 DOI: 10.3389/fncel.2019.00265] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 05/28/2019] [Indexed: 01/04/2023] Open
Abstract
Transient potassium current channels (IA channels), which are expressed in most brain areas, have a central role in modulating feedforward and feedback inhibition along the dendroaxonic axis. Loss of the modulatory channels is tightly associated with a number of brain diseases such as Alzheimer’s disease, epilepsy, fragile X syndrome (FXS), Parkinson’s disease, chronic pain, tinnitus, and ataxia. However, the functional significance of IA channels in these diseases has so far been underestimated. In this review, we discuss the distribution and function of IA channels. Particularly, we posit that downregulation of IA channels results in neuronal (mostly dendritic) hyperexcitability accompanied by the imbalanced excitation and inhibition ratio in the brain’s networks, eventually causing the brain diseases. Finally, we propose a potential therapeutic target: the enhanced action of IA channels to counteract Ca2+-permeable channels including NMDA receptors could be harnessed to restore dendritic excitability, leading to a balanced neuronal state.
Collapse
Affiliation(s)
- Wonjun Noh
- Department of Nano-Bioengineering, Incheon National University, Incheon, South Korea
| | - Sojeong Pak
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Geunho Choi
- Department of Computer Science and Engineering, Incheon National University, Incheon, South Korea
| | - Sungchil Yang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Sunggu Yang
- Department of Nano-Bioengineering, Incheon National University, Incheon, South Korea
| |
Collapse
|
46
|
van Setten J, Verweij N, Mbarek H, Niemeijer MN, Trompet S, Arking DE, Brody JA, Gandin I, Grarup N, Hall LM, Hemerich D, Lyytikäinen LP, Mei H, Müller-Nurasyid M, Prins BP, Robino A, Smith AV, Warren HR, Asselbergs FW, Boomsma DI, Caulfield MJ, Eijgelsheim M, Ford I, Hansen T, Harris TB, Heckbert SR, Hottenga JJ, Iorio A, Kors JA, Linneberg A, MacFarlane PW, Meitinger T, Nelson CP, Raitakari OT, Silva Aldana CT, Sinagra G, Sinner M, Soliman EZ, Stoll M, Uitterlinden A, van Duijn CM, Waldenberger M, Alonso A, Gasparini P, Gudnason V, Jamshidi Y, Kääb S, Kanters JK, Lehtimäki T, Munroe PB, Peters A, Samani NJ, Sotoodehnia N, Ulivi S, Wilson JG, de Geus EJC, Jukema JW, Stricker B, van der Harst P, de Bakker PIW, Isaacs A. Genome-wide association meta-analysis of 30,000 samples identifies seven novel loci for quantitative ECG traits. Eur J Hum Genet 2019; 27:952-962. [PMID: 30679814 PMCID: PMC6777533 DOI: 10.1038/s41431-018-0295-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 10/05/2018] [Accepted: 10/16/2018] [Indexed: 01/09/2023] Open
Abstract
Genome-wide association studies (GWAS) of quantitative electrocardiographic (ECG) traits in large consortia have identified more than 130 loci associated with QT interval, QRS duration, PR interval, and heart rate (RR interval). In the current study, we meta-analyzed genome-wide association results from 30,000 mostly Dutch samples on four ECG traits: PR interval, QRS duration, QT interval, and RR interval. SNP genotype data was imputed using the Genome of the Netherlands reference panel encompassing 19 million SNPs, including millions of rare SNPs (minor allele frequency < 5%). In addition to many known loci, we identified seven novel locus-trait associations: KCND3, NR3C1, and PLN for PR interval, KCNE1, SGIP1, and NFKB1 for QT interval, and ATP2A2 for QRS duration, of which six were successfully replicated. At these seven loci, we performed conditional analyses and annotated significant SNPs (in exons and regulatory regions), demonstrating involvement of cardiac-related pathways and regulation of nearby genes.
Collapse
Affiliation(s)
- Jessica van Setten
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands.
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hamdi Mbarek
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leanne M Hall
- Department of Cardiovascular Sciences, University of Leicester, Leicester, England
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester, UK
| | - Daiane Hemerich
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- CAPES Foundation, Ministry of Education of Brazil, Brasília, DF, 70040-020, Brazil
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Life Sciences, University of Tampere, 33520, Tampere, Finland
| | - Hao Mei
- Center of Biostatistics and Bioinformatics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Germany
| | - Bram P Prins
- Human Genetics Research Centre, ICCS, St George's University of London, London, UK
| | - Antonietta Robino
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykavik, Iceland
| | - Helen R Warren
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Research Centre, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Folkert W Asselbergs
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Durrer Center for Cardiovascular Research, Netherlands Heart Institute, Utrecht, The Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, and Farr Institute of Health Informatics Research and Institute of Health Informatics, University College London, London, UK
| | - Dorret I Boomsma
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mark J Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Research Centre, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Mark Eijgelsheim
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tamara B Harris
- Laboratory of Epidemiology, Demography and Biometry, National Institute on Aging, Bethesda, MD, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Annamaria Iorio
- Cardiovascular Department, "Ospedali Riuniti and University of Trieste", Trieste, Italy
| | - Jan A Kors
- Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital-The Capital Region, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Thomas Meitinger
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, England
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester, UK
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, and Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, 20520, Finland
| | - Claudia T Silva Aldana
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Doctoral Program in Biomedical Sciences, Universidad del Rosario, Bogotá, Colombia
- Institute of translational Medicine-IMT-Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Rosario, Colombia
| | - Gianfranco Sinagra
- Cardiovascular Department, "Ospedali Riuniti and University of Trieste", Trieste, Italy
| | - Moritz Sinner
- Department of Medicine I, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Elsayed Z Soliman
- Epidemiological Cardiology Research Center (EPICARE), Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Monika Stoll
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
| | - Andre Uitterlinden
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cornelia M van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Melanie Waldenberger
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Research unit of Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Paolo Gasparini
- DSM, University of Trieste, Trieste, Italy
- IRCCS-Burlo Garofolo Children Hospital, Via dell'Istria 65, Trieste, Italy
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykavik, Iceland
| | - Yalda Jamshidi
- Human Genetics Research Centre, ICCS, St George's University of London, London, UK
| | - Stefan Kääb
- Department of Medicine I, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Jørgen K Kanters
- Laboratory of Experimental Cardiology, University of Copenhagen, Copenhagen, Denmark
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Life Sciences, University of Tampere, 33520, Tampere, Finland
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Research Centre, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Annette Peters
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, England
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Groby Road, Leicester, UK
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, WA, USA
| | - Sheila Ulivi
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Eco J C de Geus
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bruno Stricker
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, The Netherlands
| | - Paul I W de Bakker
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Aaron Isaacs
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands.
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands.
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
47
|
Johnston KJA, Adams MJ, Nicholl BI, Ward J, Strawbridge RJ, Ferguson A, McIntosh AM, Bailey MES, Smith DJ. Genome-wide association study of multisite chronic pain in UK Biobank. PLoS Genet 2019; 15:e1008164. [PMID: 31194737 PMCID: PMC6592570 DOI: 10.1371/journal.pgen.1008164] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/25/2019] [Accepted: 04/27/2019] [Indexed: 12/20/2022] Open
Abstract
Chronic pain is highly prevalent worldwide and represents a significant socioeconomic and public health burden. Several aspects of chronic pain, for example back pain and a severity-related phenotype 'chronic pain grade', have been shown previously to be complex heritable traits with a polygenic component. Additional pain-related phenotypes capturing aspects of an individual's overall sensitivity to experiencing and reporting chronic pain have also been suggested as a focus for investigation. We made use of a measure of the number of sites of chronic pain in individuals within the UK general population. This measure, termed Multisite Chronic Pain (MCP), is a complex trait and its genetic architecture has not previously been investigated. To address this, we carried out a large-scale genome-wide association study (GWAS) of MCP in ~380,000 UK Biobank participants. Our findings were consistent with MCP having a significant polygenic component, with a Single Nucleotide Polymorphism (SNP) heritability of 10.2%. In total 76 independent lead SNPs at 39 risk loci were associated with MCP. Additional gene-level association analyses identified neurogenesis, synaptic plasticity, nervous system development, cell-cycle progression and apoptosis genes as enriched for genetic association with MCP. Genetic correlations were observed between MCP and a range of psychiatric, autoimmune and anthropometric traits, including major depressive disorder (MDD), asthma and Body Mass Index (BMI). Furthermore, in Mendelian randomisation (MR) analyses a causal effect of MCP on MDD was observed. Additionally, a polygenic risk score (PRS) for MCP was found to significantly predict chronic widespread pain (pain all over the body), indicating the existence of genetic variants contributing to both of these pain phenotypes. Overall, our findings support the proposition that chronic pain involves a strong nervous system component with implications for our understanding of the physiology of chronic pain. These discoveries may also inform the future development of novel treatment approaches.
Collapse
Affiliation(s)
- Keira J. A. Johnston
- Institute of Health and Wellbeing, University of Glasgow, Scotland, United Kingdom
- Deanery of Molecular, Genetic and Population Health Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Scotland, United Kingdom
- School of Life Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Scotland, United Kingdom
| | - Mark J. Adams
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Scotland, United Kingdom
| | - Barbara I. Nicholl
- Institute of Health and Wellbeing, University of Glasgow, Scotland, United Kingdom
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Scotland, United Kingdom
| | - Rona J. Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Scotland, United Kingdom
- Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Amy Ferguson
- Institute of Health and Wellbeing, University of Glasgow, Scotland, United Kingdom
| | - Andrew M. McIntosh
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Scotland, United Kingdom
| | - Mark E. S. Bailey
- School of Life Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Scotland, United Kingdom
| | - Daniel J. Smith
- Institute of Health and Wellbeing, University of Glasgow, Scotland, United Kingdom
| |
Collapse
|
48
|
Kang C, Liang C, Ahmad KE, Gu Y, Siow SF, Colebatch JG, Whyte S, Ng K, Cremer PD, Corbett AJ, Davis RL, Roscioli T, Cowley MJ, Park JS, Sue CM, Kumar KR. High Degree of Genetic Heterogeneity for Hereditary Cerebellar Ataxias in Australia. THE CEREBELLUM 2019; 18:137-146. [PMID: 30078120 DOI: 10.1007/s12311-018-0969-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genetic testing strategies such as next-generation sequencing (NGS) panels and whole genome sequencing (WGS) can be applied to the hereditary cerebellar ataxias (HCAs), but their exact role in the diagnostic pathway is unclear. We aim to determine the yield from genetic testing strategies and the genetic and phenotypic spectrum of HCA in Australia by analysing real-world data. We performed a retrospective review on 87 HCA cases referred to the Neurogenetics Clinic at the Royal North Shore Hospital, Sydney, Australia. Probands underwent triplet repeat expansion testing; those that tested negative had NGS-targeted panels and WGS testing when available. In our sample, 58.6% were male (51/87), with an average age at onset of 37.1 years. Individuals with sequencing variants had a prolonged duration of illness compared to those with a triplet repeat expansion. The detection rate in probands for routine repeat expansion panels was 13.8% (11/80). NGS-targeted panels yielded a further 11 individuals (11/32, 34.4%), with WGS yielding 1 more diagnosis (1/3, 33.3%). NGS panels and WGS improved the overall diagnostic rate to 28.8% (23/80) in 14 known HCA loci. The genetic findings included novel variants in ANO10, CACNA1A, PRKCG and SPG7. Our findings highlight the genetic heterogeneity of HCAs and support the use of NGS approaches for individuals who were negative on repeat expansion testing. In comparison to repeat disorders, individuals with sequencing variants may have a prolonged duration of illness, consistent with slower progression of disease.
Collapse
Affiliation(s)
- Ce Kang
- Faculty of Medicine and Health, Kolling Institute of Medical Research, University of Sydney Northern Clinical School, St Leonards, Australia
| | - Christina Liang
- Department of Neurogenetics, Kolling Institute, University of Sydney and Northern Sydney Local Health District, St Leonards, Australia.,Department of Neurology, Royal North Shore Hospital, St Leonards, Australia
| | - Kate E Ahmad
- Department of Neurogenetics, Kolling Institute, University of Sydney and Northern Sydney Local Health District, St Leonards, Australia.,Department of Neurology, Royal North Shore Hospital, St Leonards, Australia
| | - Yufan Gu
- Department of Neurogenetics, Kolling Institute, University of Sydney and Northern Sydney Local Health District, St Leonards, Australia.,Department of Neurology, Royal North Shore Hospital, St Leonards, Australia
| | - Sue-Faye Siow
- Department of Neurogenetics, Kolling Institute, University of Sydney and Northern Sydney Local Health District, St Leonards, Australia.,Department of Neurology, Royal North Shore Hospital, St Leonards, Australia
| | - James G Colebatch
- Prince of Wales Clinical School and Neuroscience Research Australia, University of New South Wales, Randwick, Australia.,Institute of Neurological Sciences, Prince of Wales Hospital, Randwick, Australia
| | - Scott Whyte
- Department of Neurology, Gosford Hospital, Gosford, Australia
| | - Karl Ng
- Department of Neurology, Royal North Shore Hospital, St Leonards, Australia
| | - Philip D Cremer
- Department of Neurology, Royal North Shore Hospital, St Leonards, Australia
| | - Alastair J Corbett
- Department of Neurology, Concord Repatriation General Hospital, Concord, Australia
| | - Ryan L Davis
- Faculty of Medicine and Health, Kolling Institute of Medical Research, University of Sydney Northern Clinical School, St Leonards, Australia.,Department of Neurogenetics, Kolling Institute, University of Sydney and Northern Sydney Local Health District, St Leonards, Australia
| | - Tony Roscioli
- Prince of Wales Clinical School and Neuroscience Research Australia, University of New South Wales, Randwick, Australia.,Department of Clinical Genetics, Sydney Children's Hospital, Randwick, Australia
| | - Mark J Cowley
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, Australia.,St Vincent's Clinical School, University of New South Wales, Darlinghurst, Australia
| | - Jin-Sung Park
- Department of Neurogenetics, Kolling Institute, University of Sydney and Northern Sydney Local Health District, St Leonards, Australia.,Department of Experimental Animal Research, Seoul National University Hospital, Biomedical Research Institute, Seoul, Republic of Korea
| | - Carolyn M Sue
- Department of Neurogenetics, Kolling Institute, University of Sydney and Northern Sydney Local Health District, St Leonards, Australia.,Department of Neurology, Royal North Shore Hospital, St Leonards, Australia
| | - Kishore R Kumar
- Department of Neurogenetics, Kolling Institute, University of Sydney and Northern Sydney Local Health District, St Leonards, Australia. .,Department of Neurology, Royal North Shore Hospital, St Leonards, Australia. .,Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, Australia.
| |
Collapse
|
49
|
Abstract
The spinocerebellar ataxias (SCAs) are a genetically heterogeneous group of autosomal dominantly inherited progressive disorders, the clinical hallmark of which is loss of balance and coordination accompanied by slurred speech; onset is most often in adult life. Genetically, SCAs are grouped as repeat expansion SCAs, such as SCA3/Machado-Joseph disease (MJD), and rare SCAs that are caused by non-repeat mutations, such as SCA5. Most SCA mutations cause prominent damage to cerebellar Purkinje neurons with consecutive cerebellar atrophy, although Purkinje neurons are only mildly affected in some SCAs. Furthermore, other parts of the nervous system, such as the spinal cord, basal ganglia and pontine nuclei in the brainstem, can be involved. As there is currently no treatment to slow or halt SCAs (many SCAs lead to premature death), the clinical care of patients with SCA focuses on managing the symptoms through physiotherapy, occupational therapy and speech therapy. Intense research has greatly expanded our understanding of the pathobiology of many SCAs, revealing that they occur via interrelated mechanisms (including proteotoxicity, RNA toxicity and ion channel dysfunction), and has led to the identification of new targets for treatment development. However, the development of effective therapies is hampered by the heterogeneity of the SCAs; specific therapeutic approaches may be required for each disease.
Collapse
|
50
|
Wang J, Wen Y, Zhang Q, Yu S, Chen Y, Wu X, Zhang Y, Bao X. Gene mutational analysis in a cohort of Chinese children with unexplained epilepsy: Identification of a new KCND3 phenotype and novel genes causing Dravet syndrome. Seizure 2019; 66:26-30. [PMID: 30776697 DOI: 10.1016/j.seizure.2019.01.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 01/24/2019] [Accepted: 01/26/2019] [Indexed: 01/10/2023] Open
Abstract
PURPOSE This study aimed to investigate the genetic etiology of epilepsy in a cohort of Chinese children. METHODS Targeted next-generation sequencing (NGS) was performed for 120 patients with unexplained epilepsy, including 71 patients with early-onset epileptic encephalopathies, and 16 patients with Dravet syndrome (including three patients with a Dravet-like phenotype) but without SCN1A pathogenic variants. RESULTS Pathogenic variants of 14 genes were discovered in 22 patients (18%). A de novo KCND3 pathogenic variant (c.1174G > A, p.Val392Ile) was identified in a boy with refractory epilepsy, psychomotor regression, attention deficit, and visual decline. Pathogenic variants in other coding genes were excluded via whole exome sequencing. This KCND3 variant was previously confirmed to be pathogenic by Giudicessi, et al. However, the clinical profile was different: sudden death at 20 years old without any medical history of neurological disorders, nor with any diseases typically caused by KCND3 pathogenic variants such as Brugada syndrome, spinocerebellar ataxia type 19/22 or ataxia accompanied by epilepsy. This indicates that we have identified a new KCND3 phenotype. In addition, we also uncovered a GRIN1 pathogenic variant and a novel HCN1 pathogenic variant in the Dravet cohort. CONCLUSION Our study highlights the significant utility of NGS panels in the genetic diagnosis of pediatric epilepsy. Our findings indicate that KCND3 pathogenic variants may be responsible for a wider phenotypic spectrum than previously thought, by including childhood epileptic encephalopathy. Furthermore, this study provides evidence that GRIN1 and HCN1 are candidate genes for Dravet and Dravet-like phenotypes.
Collapse
Affiliation(s)
- Jiaping Wang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Yongxin Wen
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Qingping Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Shujie Yu
- Department of Neurology, Harbin Children's Hospital, Harbin 150010, Heilongjiang Province, China
| | - Yan Chen
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Xiru Wu
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - YueHua Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Xinhua Bao
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China.
| |
Collapse
|