1
|
Wang M, Li H, Zhou Q, Zhao Q, Wang M, Geng Y, Kang H. Case report: Misdiagnosed orolingual dyskinesia as a consequence of seizures in a chorea-acanthocytosis patient with a novel VPS13A variation from a family with consanguineous marriage. Front Neurol 2024; 15:1352467. [PMID: 38933328 PMCID: PMC11199768 DOI: 10.3389/fneur.2024.1352467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Chorea-acanthocytosis (ChAc) is a rare autosomal recessive inherited syndrome with heterogeneous symptoms, which makes it a challenge for early diagnosis. The mutation of VPS13A is considered intimately related to the pathogenesis of ChAc. To date, diverse mutation patterns of VPS13A, consisting of missense, nonsense, and frameshift mutations, have been reported. In this study, we first report a clinical case that was misdiagnosed as epilepsy due to recurrent seizures accompanied by tongue bite for 9 months, which was not rectified until seizures were controlled and involuntary orolingual movements with awareness became prominent and were confirmed to be orolingual dyskinesia. The patient was eventually diagnosed as ChAc based on whole-exome sequencing revealing novel homozygous c.2061dup (frameshift mutation) and c.6796A > T dual mutations in VPS13A. The patient from a family with consanguineous marriage manifested epileptic seizures at onset, including both generalized tonic-clonic seizures and absence but normal long-term electroencephalography, and gradually developed orofacial dyskinesia, including involuntary tongue protrusion, tongue biting and ulcers, involuntary open jaws, occasionally frequent eye blinks, and head swings. The first test of the peripheral blood smear was negative, and repeated checks confirmed an elevated percentage of acanthocytes by 15-21.3%. Structural brain MRI indicated a mildly swollen left hippocampus and parahippocampal gyrus and a progressively decreased volume of the bilateral hippocampus 1 year later, along with atrophy of the head of the caudate nucleus but no progression in 1 year. We deeply analyzed the reasons for long-term misdiagnosis in an effort to achieve a more comprehensive understanding of ChAc, thus facilitating early diagnosis and treatment in future clinical practice.
Collapse
Affiliation(s)
- Mengying Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huimin Li
- Department of Neurology, The Central Hospital of Wuhan, Wuhan, Hubei, China
| | - Qing Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Zhao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Man Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yumei Geng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huicong Kang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Neurology, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
2
|
Neiman AM. Pharmacological interventions for lipid transport disorders. Front Neurosci 2023; 17:1321250. [PMID: 38156273 PMCID: PMC10752963 DOI: 10.3389/fnins.2023.1321250] [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: 10/13/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
The recent discovery that defects in inter-organelle lipid transport are at the heart of several neurological and neurodegenerative disorders raises the challenge of identifying therapeutic strategies to correct lipid transport defects. This perspective highlights two potential strategies suggested by the study of lipid transport in budding yeast. In the first approach, small molecules are proposed that enhance the lipid transfer activity of VPS13 proteins and thereby compensate for reduced transport. In the second approach, molecules that act as inter-organelle tethers could be used to create artificial contact sites and bypass the loss of endogenous contacts.
Collapse
Affiliation(s)
- Aaron M. Neiman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
3
|
Ditzel RM, Walker RH, Nirenberg MJ, Tetlow AM, Farrell K, Lind-Watson KJ, Thorn EL, Dangoor DK, Gordon R, De Sanctis C, Barton B, Karp BI, Kirby A, Lett DJ, Mente K, Simon DK, Velayos-Baeza A, Miltenberger-Miltenyi G, Humphrey J, Crary JF. An Autopsy Series of Seven Cases of VPS13A Disease (Chorea-Acanthocytosis). Mov Disord 2023; 38:2163-2172. [PMID: 37670483 PMCID: PMC10841393 DOI: 10.1002/mds.29589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/29/2023] [Accepted: 08/04/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Vacuolar protein sorting 13 homolog A (VPS13A) disease, historically known as chorea-acanthocytosis, is a rare neurodegenerative disorder caused by biallelic mutations in VPS13A, usually resulting in reduced or absent levels of its protein product, VPS13A. VPS13A localizes to contact sites between subcellular organelles, consistent with its recently identified role in lipid transfer between membranes. Mutations are associated with neuronal loss in the striatum, most prominently in the caudate nucleus, and associated marked astrogliosis. There are no other known disease-specific cellular changes (eg, protein aggregation), but autopsy reports to date have been limited, often lacking genetic or biochemical diagnostic confirmation. OBJECTIVE The goal of this study was to characterize neuropathological findings in the brains of seven patients with VPS13A disease (chorea-acanthocytosis). METHODS In this study, we collected brain tissues and clinical data from seven cases of VPS13A for neuropathological analysis. The clinical diagnosis was confirmed by the presence of VPS13A mutations and/or immunoblot showing the loss or reduction of VPS13A protein. Tissues underwent routine, special, and immunohistochemical staining focused on neurodegeneration. Electron microscopy was performed in one case. RESULTS Gross examination showed severe striatal atrophy. Microscopically, there was neuronal loss and astrogliosis in affected regions. Luxol fast blue staining showed variable lipid accumulation with diverse morphology, which was further characterized by electron microscopy. In some cases, rare degenerating p62- and ubiquitin-positive cells were present in affected regions. Calcifications were present in four cases, being extensive in one. CONCLUSIONS We present the largest autopsy series of biochemically and genetically confirmed VPS13A disease and identify novel histopathological findings implicating abnormal lipid accumulation. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ricky M. Ditzel
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ruth H. Walker
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - Melissa J. Nirenberg
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - Amber M. Tetlow
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kurt Farrell
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kourtni J. Lind-Watson
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Emma L. Thorn
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Diana K. Dangoor
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ronald Gordon
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claudia De Sanctis
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Brandon Barton
- Rush University Medical Center, Chicago, Illinois, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Barbara I. Karp
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Alana Kirby
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Debra J. Lett
- Newcastle Brain Tissue Resource, Newcastle University, Newcastle, UK
| | - Karin Mente
- Departments of Neurology and Pathology, Case Western Reserve University, Cleveland, OH, USA
- Louis Stokes Cleveland VA Medical Center, Cleveland OH, USA
| | - David K. Simon
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Antonio Velayos-Baeza
- Department of Physiology, Anatomy, and Genetics, University of Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Gabriel Miltenberger-Miltenyi
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- Reference Center on Lysosomal Storage Diseases, Hospital Senhora da Oliveira, Guimarães, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Jack Humphrey
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F. Crary
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
4
|
Krajnik A, Nimmer E, Brazzo JA, Biber JC, Drewes R, Tumenbayar BI, Sullivan A, Pham K, Krug A, Heo Y, Kolega J, Heo SJ, Lee K, Weil BR, Kim DH, Gupte SA, Bae Y. Survivin regulates intracellular stiffness and extracellular matrix production in vascular smooth muscle cells. APL Bioeng 2023; 7:046104. [PMID: 37868708 PMCID: PMC10590228 DOI: 10.1063/5.0157549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
Vascular dysfunction is a common cause of cardiovascular diseases characterized by the narrowing and stiffening of arteries, such as atherosclerosis, restenosis, and hypertension. Arterial narrowing results from the aberrant proliferation of vascular smooth muscle cells (VSMCs) and their increased synthesis and deposition of extracellular matrix (ECM) proteins. These, in turn, are modulated by arterial stiffness, but the mechanism for this is not fully understood. We found that survivin is an important regulator of stiffness-mediated ECM synthesis and intracellular stiffness in VSMCs. Whole-transcriptome analysis and cell culture experiments showed that survivin expression is upregulated in injured femoral arteries in mice and in human VSMCs cultured on stiff fibronectin-coated hydrogels. Suppressed expression of survivin in human VSMCs significantly decreased the stiffness-mediated expression of ECM components related to arterial stiffening, such as collagen-I, fibronectin, and lysyl oxidase. By contrast, expression of these ECM proteins was rescued by ectopic expression of survivin in human VSMCs cultured on soft hydrogels. Interestingly, atomic force microscopy analysis showed that suppressed or ectopic expression of survivin decreases or increases intracellular stiffness, respectively. Furthermore, we observed that inhibiting Rac and Rho reduces survivin expression, elucidating a mechanical pathway connecting intracellular tension, mediated by Rac and Rho, to survivin induction. Finally, we found that survivin inhibition decreases FAK phosphorylation, indicating that survivin-dependent intracellular tension feeds back to maintain signaling through FAK. These findings suggest a novel mechanism by which survivin potentially modulates arterial stiffness.
Collapse
Affiliation(s)
- Amanda Krajnik
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Erik Nimmer
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | - Joseph A. Brazzo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - John C. Biber
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Rhonda Drewes
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Bat-Ider Tumenbayar
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Andra Sullivan
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | - Khanh Pham
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Alanna Krug
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | | | - John Kolega
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Su-Jin Heo
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | - Brian R. Weil
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Sachin A. Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA
| | - Yongho Bae
- Author to whom correspondence should be addressed:
| |
Collapse
|
5
|
Alkahtani S, Alkahtane AA, Stournaras C, Alarifi S. Chorein sensitive microtubule organization in tumor cells. PeerJ 2023; 11:e16074. [PMID: 37744224 PMCID: PMC10517657 DOI: 10.7717/peerj.16074] [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: 05/02/2023] [Accepted: 08/20/2023] [Indexed: 09/26/2023] Open
Abstract
Background The purpose of this study is to analyzed the involvement of chorein in microtubules organization of three types of malignant; rhabdomyosarcoma tumor cells (ZF), rhabdomyosarcoma cells (RH30), and rhabdomyosarcoma cells (RD). ZF are expressing high chorein levels. Previous studies revealed that chorein protein silencing in ZF tumor cells persuaded apoptotic response followed by cell death. In addition, in numerous malignant and non-malignant cells this protein regulates actin cytoskeleton structure and cellular signaling. However, the function of chorein protein in microtubular organization is yet to be established. Methods In a current research study, we analyzed the involvement of chorein in microtubules organization by using three types of malignant rhabdomyosarcoma cells. We have applied confocal laser-scanning microscopy to analyze microtubules structure and RT-PCR to examine cytoskeletal gene transcription. Results We report here that in rhabdomyosarcoma cells (RH30), chorein silencing induced disarrangement of microtubular network. This was documented by laser scanning microscopy and further quantified by FACS analysis. Interestingly and in agreement with previous reports, tubulin gene transcription in RH cells was unchanged upon silencing of chorein protein. Equally, confocal analysis showed minor disordered microtubules organization with evidently weakened staining in rhabdomyosarcoma cells (RD and ZF) after silencing of chorein protein. Conclusion These results disclose that chorein silencing induces considerable structural disorganization of tubulin network in RH30 human rhabdomyosarcoma tumor cells. Additional studies are now needed to establish the role of chorein in regulating cytoskeleton architecture in tumor cells.
Collapse
Affiliation(s)
- Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah A. Alkahtane
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Christos Stournaras
- Department of Biochemistry, University of Crete Medical School, Heraklion, Greece
| | - Saud Alarifi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
6
|
Miltenberger-Miltenyi G, Jones A, Tetlow AM, Conceição VA, Crary JF, Ditzel RM, Farrell K, Nandakumar R, Barton B, Karp BI, Kirby A, Lett DJ, Mente K, Morgello S, Simon DK, Walker RH. Sphingolipid and Phospholipid Levels Are Altered in Human Brain in Chorea-Acanthocytosis. Mov Disord 2023; 38:1535-1541. [PMID: 37307400 DOI: 10.1002/mds.29445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/24/2023] [Accepted: 04/28/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Chorea-acanthocytosis (ChAc) is associated with mutations of VPS13A, which encodes for chorein, a protein implicated in lipid transport at intracellular membrane contact sites. OBJECTIVES The goal of this study was to establish the lipidomic profile of patients with ChAc. METHODS We analyzed 593 lipid species in the caudate nucleus (CN), putamen, and dorsolateral prefrontal cortex (DLPFC) from postmortem tissues of four patients with ChAc and six patients without ChAc. RESULTS We found increased levels of bis(monoacylglycerol)phosphate, sulfatide, lysophosphatidylserine, and phosphatidylcholine ether in the CN and putamen, but not in the DLPFC, of patients with ChAc. Phosphatidylserine and monoacylglycerol were increased in the CN and N-acyl phosphatidylserine in the putamen. N-acyl serine was decreased in the CN and DLPFC, whereas lysophosphatidylinositol was decreased in the DLPFC. CONCLUSIONS We present the first evidence of altered sphingolipid and phospholipid levels in the brains of patients with ChAc. Our observations are congruent with recent findings in cellular and animal models, and implicate defects of lipid processing in VPS13A disease pathophysiology. © 2023 International Parkinson and Movement Disorder Society. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Gabriel Miltenberger-Miltenyi
- Laboratório de Genética, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- Reference Center on Lysosomal Storage Diseases, Hospital Senhora da Oliveira, Guimarães, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Attila Jones
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Amber M Tetlow
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Department of Neuroscience and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Vasco A Conceição
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - John F Crary
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Department of Neuroscience and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Ricky Michael Ditzel
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Department of Neuroscience and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Kurt Farrell
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Department of Neuroscience and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Renu Nandakumar
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York City, New York, USA
| | - Brandon Barton
- Rush University Medical Center, Chicago, Illinois, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Barbara I Karp
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Alana Kirby
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Debra J Lett
- Newcastle Brain Tissue Resource, Newcastle University, Newcastle, United Kingdom
| | - Karin Mente
- Departments of Neurology and Pathology, Case Western Reserve University, Cleveland, Ohio, USA
- Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, USA
| | - Susan Morgello
- Department of Pathology, Molecular, and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - David K Simon
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ruth H Walker
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, USA
| |
Collapse
|
7
|
Chaudhari S, Ware AP, Jasti DB, Gorthi SP, Acharya LP, Bhat M, Mallya S, Satyamoorthy K. Exome sequencing of choreoacanthocytosis reveals novel mutations in VPS13A and co-mutation in modifier gene(s). Mol Genet Genomics 2023; 298:965-976. [PMID: 37209156 DOI: 10.1007/s00438-023-02032-2] [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: 09/01/2022] [Accepted: 05/04/2023] [Indexed: 05/22/2023]
Abstract
Choreoacanthocytosis, one of the forms of neuroacanthocytosis, is caused by mutations in vacuolar protein sorting-associated protein A (VPS13A), and is often misdiagnosed with other form of neuroacanthocytosis with discrete genetic defects. The phenotypic variations among the patients with VPS13A mutations significantly obfuscates the understanding of the disease and treatment strategies. In this study, two unrelated cases were identified, exhibiting the core phenotype of neuroacanthocytosis but with considerable clinical heterogeneity. Case 1 presented with an additional Parkinsonism phenotype, whereas seizures were evident in case 2. To decipher the genetic basis, whole exome sequencing followed by validation with Sanger sequencing was performed. A known homozygous pathogenic nonsense mutation (c.799C > T; p.R267X) in exon 11 of the VPS13A gene was identified in case 1 that resulted in a truncated protein. A novel missense mutation (c.9263T > G; p.M3088R) in exon 69 of VPS13A identified in case 2 was predicted as pathogenic. In silico analysis of the p.M3088R mutation at the C-terminus of VPS13A suggests a loss of interaction with TOMM40 and may disrupt mitochondrial localization. We also observed an increase in mitochondrial DNA copy numbers in case 2. Mutation analysis revealed benign heterozygous variants in interacting partners of VPS13A such as VAPA in case 1. Our study confirmed the cases as ChAc and identified the novel homozygous variant of VPS13A (c.9263T > G; p.M3088R) within the mutation spectrum of VPS13A-associated ChAc. Furthermore, mutations in VPS13A and co-mutations in its potential interacting partner(s) might contribute to the diverse clinical manifestations of ChAc, which requires further study.
Collapse
Affiliation(s)
- Sima Chaudhari
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Akshay Pramod Ware
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Dushyanth Babu Jasti
- Department of Neurology, Kasturba Medical College, Manipal, Karnataka, 576104, India
| | - Sankar Prasad Gorthi
- Department of Neurology, Kasturba Medical College, Manipal, Karnataka, 576104, India
- Department of Neurology, Bharati Hospital and Research Center, Bharati Vidyapeeth (Deemed to be University) Medical College and Hospital, Dhankawadi, Pune, Maharashtra, 411043, India
| | - Lavanya Prakash Acharya
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Manoj Bhat
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sandeep Mallya
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
8
|
Guillén-Samander A, De Camilli P. Endoplasmic Reticulum Membrane Contact Sites, Lipid Transport, and Neurodegeneration. Cold Spring Harb Perspect Biol 2023; 15:a041257. [PMID: 36123033 PMCID: PMC10071438 DOI: 10.1101/cshperspect.a041257] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The Endoplasmic Reticulum (ER) is an endomembrane system that plays a multiplicity of roles in cell physiology and populates even the most distal cell compartments, including dendritic tips and axon terminals of neurons. Some of its functions are achieved by a cross talk with other intracellular membranous organelles and with the plasma membrane at membrane contacts sites (MCSs). As the ER synthesizes most membrane lipids, lipid exchanges mediated by lipid transfer proteins at MCSs are a particularly important aspect of this cross talk, which synergizes with the cross talk mediated by vesicular transport. Several mutations of genes that encode proteins localized at ER MCSs result in familial neurodegenerative diseases, emphasizing the importance of the normal lipid traffic within cells for a healthy brain. Here, we provide an overview of such diseases, with a specific focus on proteins that directly or indirectly impact lipid transport.
Collapse
Affiliation(s)
- Andrés Guillén-Samander
- Departments of Neuroscience and of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06520, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Pietro De Camilli
- Departments of Neuroscience and of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06520, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| |
Collapse
|
9
|
Dall'Armellina F, Stagi M, Swan LE. In silico modeling human VPS13 proteins associated with donor and target membranes suggests lipid transfer mechanisms. Proteins 2023; 91:439-455. [PMID: 36404287 PMCID: PMC10953354 DOI: 10.1002/prot.26446] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/14/2022] [Accepted: 11/03/2022] [Indexed: 11/22/2022]
Abstract
The VPS13 protein family constitutes a novel class of bridge-like lipid transferases. Autosomal recessive inheritance of mutations in VPS13 genes is associated with the development of neurodegenerative diseases in humans. Bioinformatic approaches previously recognized the domain architecture of these proteins. In this study, we model the first ever full-length structures of the four human homologs VPS13A, VPS13B, VPS13C, and VPS13D in association with model membranes, to investigate their lipid transfer ability and potential structural association with membrane leaflets. We analyze the evolutionary conservation and physicochemical properties of these proteins, focusing on conserved C-terminal amphipathic helices that disturb organelle surfaces and that, adjoined, resemble a traditional Venetian gondola. The gondola domains share significant structural homology with lipid droplet surface-binding proteins. We introduce in silico protein-membrane models displaying the mode of association of VPS13A, VPS13B, VPS13C, and VPS13D to donor and target membranes, and present potential models of action for protein-mediated lipid transfer.
Collapse
Affiliation(s)
- Filippo Dall'Armellina
- Department of Biochemistry and Systems BiologyInstitute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUK
| | - Massimiliano Stagi
- Department of Biochemistry and Systems BiologyInstitute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUK
| | - Laura E. Swan
- Department of Biochemistry and Systems BiologyInstitute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUK
| |
Collapse
|
10
|
Peikert K, Danek A. VPS13 Forum Proceedings: XK, XK-Related and VPS13 Proteins in Membrane Lipid Dynamics. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2023; 6:25152564231156994. [PMID: 37366410 PMCID: PMC10243564 DOI: 10.1177/25152564231156994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 01/23/2023] [Indexed: 06/28/2023]
Abstract
In 2020, the pandemic interrupted the series of biannual International Neuroacanthocytosis Meetings that brought together clinicians, scientists, and patient groups to share research into a small group of devastating genetic diseases that combine both acanthocytosis (deformed red blood cells) and neurodegeneration with movement disorders. This Meeting Report describes talks at the 5th VPS13 Forum in January 2022, one of a series of online meetings held to fill the gap. The meeting addressed the basic biology of two key proteins implicated in chorea-acanthocytosis (mutations in VPS13A) and McLeod syndrome (mutations in XK). In a remarkable confluence of ideas, the speakers described different aspects of a single functional unit that comprises of VPS13A and XK proteins working together. Conditions caused by VPS13 (A-D) gene family mutations and related genes, such as XK, previously footnote knowledge, seem to turn central for a novel disease paradigm: bulk lipid transfer disorders.
Collapse
Affiliation(s)
- Kevin Peikert
- Translational Neurodegeneration Section
“Albrecht-Kossel”, Department of Neurology, University Medical Center Rostock, University of
Rostock, Rostock, Germany
- Center for Transdisciplinary Neurosciences
Rostock (CTNR), University Medical Center Rostock, Rostock, Germany
- United Neuroscience Campus Lund-Rostock
(UNC), Rostock site, Rostock, Germany
| | - Adrian Danek
- Department of Neurology, University Hospital,
LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases
(Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE), Research Site Munich, Munich,
Germany
| |
Collapse
|
11
|
Park JS, Hu Y, Hollingsworth NM, Miltenberger-Miltenyi G, Neiman AM. Interaction between VPS13A and the XK scramblase is important for VPS13A function in humans. J Cell Sci 2022; 135:jcs260227. [PMID: 35950506 PMCID: PMC9482346 DOI: 10.1242/jcs.260227] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022] Open
Abstract
VPS13 family proteins form conduits between the membranes of different organelles through which lipids are transferred. In humans, there are four VPS13 paralogs, and mutations in the genes encoding each of them are associated with different inherited disorders. VPS13 proteins contain multiple conserved domains. The Vps13 adaptor-binding (VAB) domain binds to adaptor proteins that recruit VPS13 to specific membrane contact sites. This work demonstrates the importance of a different domain in VPS13A function. The pleckstrin homology (PH) domain at the C-terminal region of VPS13A is required to form a complex with the XK scramblase and for the co-localization of VPS13A with XK within the cell. Alphafold modeling was used to predict an interaction surface between VPS13A and XK. Mutations in this region disrupt both complex formation and co-localization of the two proteins. Mutant VPS13A alleles found in patients with VPS13A disease truncate the PH domain. The phenotypic similarities between VPS13A disease and McLeod syndrome caused by mutations in VPS13A and XK, respectively, argue that loss of the VPS13A-XK complex is the basis of both diseases.
Collapse
Affiliation(s)
- Jae-Sook Park
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Yiying Hu
- Fish Core Unit, German Center for Neurodegenerative Diseases München (DZNE), 81377 Munich, Germany
- Munich Medical Research School (MMRS), 80336 Munich, Germany
| | - Nancy M. Hollingsworth
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | | | - Aaron M. Neiman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| |
Collapse
|
12
|
A partnership between the lipid scramblase XK and the lipid transfer protein VPS13A at the plasma membrane. Proc Natl Acad Sci U S A 2022; 119:e2205425119. [PMID: 35994651 PMCID: PMC9436381 DOI: 10.1073/pnas.2205425119] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Chorea-acanthocytosis (ChAc) and McLeod syndrome are diseases with shared clinical manifestations caused by mutations in VPS13A and XK, respectively. Key features of these conditions are the degeneration of caudate neurons and the presence of abnormally shaped erythrocytes. XK belongs to a family of plasma membrane (PM) lipid scramblases whose action results in exposure of PtdSer at the cell surface. VPS13A is an endoplasmic reticulum (ER)-anchored lipid transfer protein with a putative role in the transport of lipids at contacts of the ER with other membranes. Recently VPS13A and XK were reported to interact by still unknown mechanisms. So far, however, there is no evidence for a colocalization of the two proteins at contacts of the ER with the PM, where XK resides, as VPS13A was shown to be localized at contacts between the ER and either mitochondria or lipid droplets. Here we show that VPS13A can also localize at ER-PM contacts via the binding of its PH domain to a cytosolic loop of XK, that such interaction is regulated by an intramolecular interaction within XK, and that both VPS13A and XK are highly expressed in the caudate neurons. Binding of the PH domain of VPS13A to XK is competitive with its binding to intracellular membranes that mediate other tethering functions of VPS13A. Our findings support a model according to which VPS13A-dependent lipid transfer between the ER and the PM is coupled to lipid scrambling within the PM. They raise the possibility that defective cell surface exposure of PtdSer may be responsible for neurodegeneration.
Collapse
|
13
|
Ryoden Y, Nagata S. The XK plasma membrane scramblase and the VPS13A cytosolic lipid transporter for ATP-induced cell death. Bioessays 2022; 44:e2200106. [PMID: 35996795 DOI: 10.1002/bies.202200106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/25/2022] [Accepted: 08/08/2022] [Indexed: 11/12/2022]
Abstract
Extracellular ATP released from necrotic cells in inflamed tissues activates the P2X7 receptor, stimulates the exposure of phosphatidylserine, and causes cell lysis. Recent findings indicated that XK, a paralogue of XKR8 lipid scramblase, forms a complex with VPS13A at the plasma membrane of T cells. Upon engagement by ATP, an unidentified signal(s) from the P2X7 receptor activates the XK-VPS13A complex to scramble phospholipids, followed by necrotic cell death. P2X7 is expressed highly in CD25+ CD4+ T cells but weakly in CD8+ T cells, suggesting a role of this system in the activation of the immune system to prevent infection. On the other hand, a loss-of-function mutation in XK or VPS13A causes neuroacanthocytosis, indicating the crucial involvement of XK-VPS13A-mediated phospholipid scrambling at plasma membranes in the maintenance of homeostasis in the nervous and red blood cell systems.
Collapse
Affiliation(s)
- Yuta Ryoden
- Laboratory of Biochemistry and Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shigekazu Nagata
- Laboratory of Biochemistry and Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
14
|
Kaminska J, Soczewka P, Rzepnikowska W, Zoladek T. Yeast as a Model to Find New Drugs and Drug Targets for VPS13-Dependent Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23095106. [PMID: 35563497 PMCID: PMC9104724 DOI: 10.3390/ijms23095106] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 12/10/2022] Open
Abstract
Mutations in human VPS13A-D genes result in rare neurological diseases, including chorea-acanthocytosis. The pathogenesis of these diseases is poorly understood, and no effective treatment is available. As VPS13 genes are evolutionarily conserved, the effects of the pathogenic mutations could be studied in model organisms, including yeast, where one VPS13 gene is present. In this review, we summarize advancements obtained using yeast. In recent studies, vps13Δ and vps13-I2749 yeast mutants, which are models of chorea-acanthocytosis, were used to screen for multicopy and chemical suppressors. Two of the suppressors, a fragment of the MYO3 and RCN2 genes, act by downregulating calcineurin activity. In addition, vps13Δ suppression was achieved by using calcineurin inhibitors. The other group of multicopy suppressors were genes: FET4, encoding iron transporter, and CTR1, CTR3 and CCC2, encoding copper transporters. Mechanisms of their suppression rely on causing an increase in the intracellular iron content. Moreover, among the identified chemical suppressors were copper ionophores, which require a functional iron uptake system for activity, and flavonoids, which bind iron. These findings point at areas for further investigation in a higher eukaryotic model of VPS13-related diseases and to new therapeutic targets: calcium signalling and copper and iron homeostasis. Furthermore, the identified drugs are interesting candidates for drug repurposing for these diseases.
Collapse
Affiliation(s)
- Joanna Kaminska
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, 02-106 Warsaw, Poland; (J.K.); (P.S.)
| | - Piotr Soczewka
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, 02-106 Warsaw, Poland; (J.K.); (P.S.)
| | - Weronika Rzepnikowska
- Neuromuscular Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Teresa Zoladek
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, 02-106 Warsaw, Poland; (J.K.); (P.S.)
- Correspondence:
| |
Collapse
|
15
|
Reichel F, Kräter M, Peikert K, Glaß H, Rosendahl P, Herbig M, Rivera Prieto A, Kihm A, Bosman G, Kaestner L, Hermann A, Guck J. Changes in Blood Cell Deformability in Chorea-Acanthocytosis and Effects of Treatment With Dasatinib or Lithium. Front Physiol 2022; 13:852946. [PMID: 35444561 PMCID: PMC9013823 DOI: 10.3389/fphys.2022.852946] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/08/2022] [Indexed: 12/29/2022] Open
Abstract
Misshaped red blood cells (RBCs), characterized by thorn-like protrusions known as acanthocytes, are a key diagnostic feature in Chorea-Acanthocytosis (ChAc), a rare neurodegenerative disorder. The altered RBC morphology likely influences their biomechanical properties which are crucial for the cells to pass the microvasculature. Here, we investigated blood cell deformability of five ChAc patients compared to healthy controls during up to 1-year individual off-label treatment with the tyrosine kinase inhibitor dasatinib or several weeks with lithium. Measurements with two microfluidic techniques allowed us to assess RBC deformability under different shear stresses. Furthermore, we characterized leukocyte stiffness at high shear stresses. The results showed that blood cell deformability–including both RBCs and leukocytes - in general was altered in ChAc patients compared to healthy donors. Therefore, this study shows for the first time an impairment of leukocyte properties in ChAc. During treatment with dasatinib or lithium, we observed alterations in RBC deformability and a stiffness increase for leukocytes. The hematological phenotype of ChAc patients hinted at a reorganization of the cytoskeleton in blood cells which partly explains the altered mechanical properties observed here. These findings highlight the need for a systematic assessment of the contribution of impaired blood cell mechanics to the clinical manifestation of ChAc.
Collapse
Affiliation(s)
- Felix Reichel
- Max-Planck-Institut für die Physik des Lichts and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Martin Kräter
- Max-Planck-Institut für die Physik des Lichts and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Kevin Peikert
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Hannes Glaß
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Philipp Rosendahl
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Maik Herbig
- Max-Planck-Institut für die Physik des Lichts and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Alejandro Rivera Prieto
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Alexander Kihm
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
| | - Giel Bosman
- Department of Biochemistry, Radboud UMC, Nijmegen, Netherlands
| | - Lars Kaestner
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
- Theoretical Medicine and Biosciences, Saarland University, Homburg, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Dresden, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Rostock/Greifswald, Rostock, Germany
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Jochen Guck
- Max-Planck-Institut für die Physik des Lichts and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
- *Correspondence: Jochen Guck,
| |
Collapse
|
16
|
Huang S, Zhang J, Tao M, Lv Y, Xu L, Liang Z. Two case reports of chorea-acanthocytosis and review of literature. Eur J Med Res 2022; 27:22. [PMID: 35130982 PMCID: PMC8822714 DOI: 10.1186/s40001-022-00646-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 01/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chorea-acanthocytosis (ChAc), as the most common subtype of neuroacanthocytosis syndrome, is characterized by the presence of acanthocytes and neurological symptoms. It is thought to be caused by the VPS13A (vacuolar protein sorting-associated protein 13A) mutations. This article reports two confirmed cases of ChAc and summarizes some suggestive features, which provide direction for the diagnosis and treatment of acanthocytosis in the future. CASE PRESENTATION Here, we present two cases of ChAc diagnosed based on typical clinical symptoms, neuroimaging features, genetic findings of VPS13A, and response to the symptomatic treatment. CONCLUSIONS Chorea-acanthocytosis is a rare neurodegenerative disease with various early clinical manifestations. The final diagnosis of the ChAc can be established by either genetic analysis or protein expression by Western blotting. Supportive treatments and nursing are helpful to improve the quality of the patient's life. Nevertheless, it is imperative to investigate the impact of neuroimaging and neuropathological diagnosis in a larger group of ChAc in future studies.
Collapse
Affiliation(s)
- Shuangfeng Huang
- Second Clinical Medical College, Binzhou Medical University, Yantai, Shandong, China.,Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Junliang Zhang
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Manli Tao
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yaodong Lv
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Luyao Xu
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Zhigang Liang
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China.
| |
Collapse
|
17
|
|
18
|
Adaptative Up-Regulation of PRX2 and PRX5 Expression Characterizes Brain from a Mouse Model of Chorea-Acanthocytosis. Antioxidants (Basel) 2021; 11:antiox11010076. [PMID: 35052580 PMCID: PMC8772732 DOI: 10.3390/antiox11010076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 02/04/2023] Open
Abstract
The peroxiredoxins (PRXs) constitute a ubiquitous antioxidant. Growing evidence in neurodegenerative disorders such as Parkinson’s disease (PD) or Alzheimer’s disease (AD) has highlighted a crucial role for PRXs against neuro-oxidation. Chorea-acanthocytosis/Vps13A disease (ChAc) is a devastating, life-shortening disorder characterized by acanthocytosis, neurodegeneration and abnormal proteostasis. We recently developed a Vps13a−/− ChAc-mouse model, showing acanthocytosis, neurodegeneration and neuroinflammation which could be restored by LYN inactivation. Here, we show in our Vps13a−/− mice protein oxidation, NRF2 activation and upregulation of downstream cytoprotective systems NQO1, SRXN1 and TRXR in basal ganglia. This was associated with upregulation of PRX2/5 expression compared to wild-type mice. PRX2 expression was age-dependent in both mouse strains, whereas only Vps13a−/− PRX5 expression was increased independent of age. LYN deficiency or nilotinib-mediated LYN inhibition improved autophagy in Vps13a−/− mice. In Vps13a−/−; Lyn−/− basal ganglia, absence of LYN resulted in reduced NRF2 activation and down-regulated expression of PRX2/5, SRXN1 and TRXR. Nilotinib treatment of Vps13a−/− mice reduced basal ganglia oxidation, and plasma PRX5 levels, suggesting plasma PRX5 as a possible ChAc biomarker. Our data support initiation of therapeutic Lyn inhibition as promptly as possible after ChAc diagnosis to minimize development of irreversible neuronal damage during otherwise inevitable ChAc progression.
Collapse
|
19
|
Abbas MM, Thenral SG, Ramprasad VL, Walker RH, Kukkle PL. Chorea-acanthocytosis: 3 New Families with Novel Genetic and Metabolic Findings. Ann Indian Acad Neurol 2021; 24:452-456. [PMID: 34447025 PMCID: PMC8370142 DOI: 10.4103/aian.aian_215_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/27/2020] [Accepted: 09/23/2020] [Indexed: 11/05/2022] Open
Affiliation(s)
| | - S G Thenral
- Medgenome Labs Pvt Ltd, Bangalore, Karnataka, India
| | | | - Ruth H Walker
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, New York, USA.,Department of Neurology, Mount Sinai School of Medicine, New York, USA
| | - Prashanth Lingappa Kukkle
- Parkinson and Ageing Research Foundation, Bangalore, Karnataka, India.,Center for Parkinson's Disease and Movement Disorders Clinic, Vikram Hospitals, Bangalore, Karnataka, India.,Parkinson's Disease and Movement Disorders Clinic, Bangalore, Karnataka, India
| |
Collapse
|
20
|
Mitchell SD, Albin RL, Dauer WT, Goudreau JL, Sidiropoulos C. Heterozygous VPS13A and PARK2 Mutations in a Patient with Parkinsonism and Seizures. Case Rep Neurol 2021; 13:341-346. [PMID: 34248567 PMCID: PMC8255736 DOI: 10.1159/000515805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/05/2021] [Indexed: 01/09/2023] Open
Abstract
Neuroacanthocytosis (NA) is a diverse group of disorders in which nervous system abnormalities co-occur with irregularly shaped red blood cells called acanthocytes. Chorea-acanthocytosis is the most common of these syndromes and is an autosomal recessive disease caused by mutations in the vacuolar protein sorting 13A (VPS13A) gene. We report a case of early onset parkinsonism and seizures in a 43-year-old male with a family history of NA. Neurologic examinations showed cognitive impairment and marked parkinsonian signs. MRI showed bilateral basal ganglia gliosis. He was found to have a novel heterozygous mutation in the VPS13A gene, in addition a heterozygous mutation in the PARK2 gene. His clinical picture was atypical for typical chorea-acanthocytosis (ChAc). The compound heterozygous mutations of VPS13A and PARK2 provide the most plausible explanation for this patient's clinical symptoms. This case adds to the phenotypic diversity of ChAc. More research is needed to fully understand the roles of epistatic interactions on phenotypic expression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Steven D Mitchell
- Department of Neurology, Michigan State University, East Lansing, Michigan, USA
| | - Roger L Albin
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,VAAAHS GRECC, Ann Arbor, Michigan, USA
| | - William T Dauer
- Department of Neurology and Neurotherapeutics, O'Donnell Brain Institute, Dallas, Texas, USA.,Department of Neuroscience, University of Texas Southwestern, Dallas, Texas, USA
| | - John L Goudreau
- Department of Neurology, Michigan State University, East Lansing, Michigan, USA
| | | |
Collapse
|
21
|
Genetic Dissection of Vps13 Regulation in Yeast Using Disease Mutations from Human Orthologs. Int J Mol Sci 2021; 22:ijms22126200. [PMID: 34201352 PMCID: PMC8229349 DOI: 10.3390/ijms22126200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/03/2021] [Accepted: 06/06/2021] [Indexed: 01/03/2023] Open
Abstract
The VPS13 family of proteins have emerged as key players in intracellular lipid transport and human health. Humans have four different VPS13 orthologs, the dysfunction of which leads to different diseases. Yeast has a single VPS13 gene, which encodes a protein that localizes to multiple different membrane contact sites. The yeast vps13Δ mutant is pleiotropic, exhibiting defects in sporulation, protein trafficking, endoplasmic reticulum (ER)-phagy and mitochondrial function. Non-null alleles resulting from missense mutations can be useful reagents for understanding the multiple functions of a gene. The exceptionally large size of Vps13 makes the identification of key residues challenging. As a means to identify critical residues in yeast Vps13, amino acid substitution mutations from VPS13A, B, C and D, associated with human disease, were introduced at the cognate positions of yeast VPS13, some of which created separation-of-function alleles. Phenotypic analyses of these mutants have revealed that the promotion of ER-phagy is a fourth, genetically separable role of VPS13 and provide evidence that co-adaptors at the endosome mediate the activity of VPS13 in vacuolar sorting.
Collapse
|
22
|
Dziurdzik SK, Bean BDM, Davey M, Conibear E. A VPS13D spastic ataxia mutation disrupts the conserved adaptor-binding site in yeast Vps13. Hum Mol Genet 2021; 29:635-648. [PMID: 31943017 PMCID: PMC7068118 DOI: 10.1093/hmg/ddz318] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/19/2019] [Accepted: 12/27/2019] [Indexed: 12/12/2022] Open
Abstract
Mutations in each of the four human VPS13 (VPS13A-D) proteins are associated with distinct neurological disorders: chorea-acanthocytosis, Cohen syndrome, early-onset Parkinson's disease and spastic ataxia. Recent evidence suggests that the different VPS13 paralogs transport lipids between organelles at different membrane contact sites. How each VPS13 isoform is targeted to organelles is not known. We have shown that the localization of yeast Vps13 protein to membranes requires a conserved six-repeat region, the Vps13 Adaptor Binding (VAB) domain, which binds to organelle-specific adaptors. Here, we use a systematic mutagenesis strategy to determine the role of each repeat in recognizing each known adaptor. Our results show that mutation of invariant asparagines in repeats 1 and 6 strongly impacts the binding of all adaptors and blocks Vps13 membrane recruitment. However, we find that repeats 5-6 are sufficient for localization and interaction with adaptors. This supports a model where a single adaptor-binding site is found in the last two repeats of the VAB domain, while VAB domain repeat 1 may influence domain conformation. Importantly, a disease-causing mutation in VPS13D, which maps to the highly conserved asparagine residue in repeat 6, blocks adaptor binding and Vps13 membrane recruitment when modeled in yeast. Our findings are consistent with a conserved adaptor binding role for the VAB domain and suggest the presence of as-yet-unidentified adaptors in both yeast and humans.
Collapse
Affiliation(s)
- Samantha K Dziurdzik
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Björn D M Bean
- Department of Biology, Concordia University, Montreal, QC H3G 1M8, Canada
| | - Michael Davey
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Elizabeth Conibear
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| |
Collapse
|
23
|
Targeting Lyn Kinase in Chorea-Acanthocytosis: A Translational Treatment Approach in a Rare Disease. J Pers Med 2021; 11:jpm11050392. [PMID: 34068769 PMCID: PMC8150322 DOI: 10.3390/jpm11050392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Chorea-acanthocytosis (ChAc) is a neurodegenerative disease caused by mutations in the VPS13A gene. It is characterized by several neurological symptoms and the appearance of acanthocytes. Elevated tyrosine kinase Lyn activity has been recently identified as one of the key pathophysiological mechanisms in this disease, and therefore represents a promising drug target. Methods: We evaluated an individual off-label treatment with the tyrosine kinase inhibitor dasatinib (100 mg/d, 25.8–50.4 weeks) of three ChAc patients. Alongside thorough safety monitoring, we assessed motor and non-motor scales (e.g., MDS-UPDRS, UHDRS, quality of life) as well as routine and experimental laboratory parameters (e.g., serum neurofilament, Lyn kinase activity, actin cytoskeleton in red blood cells). Results: Dasatinib appeared to be reasonably safe. The clinical parameters remained stable without significant improvement or deterioration. Regain of deep tendon reflexes was observed in one patient. Creatine kinase, serum neurofilament levels, and acanthocyte count did not reveal consistent effects. However, a reduction of initially elevated Lyn kinase activity and accumulated autophagy markers, as well as a partial restoration of the actin cytoskeleton, was found in red blood cells. Conclusions: We report on the first treatment approach with disease-modifying intention in ChAc. The experimental parameters indicate target engagement in red blood cells, while clinical effects on the central nervous system could not be proven within a rather short treatment time. Limited knowledge on the natural history of ChAc and the lack of appropriate biomarkers remain major barriers for “clinical trial readiness”. We suggest a panel of outcome parameters for future clinical trials in ChAc.
Collapse
|
24
|
Peikert K, Federti E, Matte A, Constantin G, Pietronigro EC, Fabene PF, Defilippi P, Turco E, Del Gallo F, Pucci P, Amoresano A, Illiano A, Cozzolino F, Monti M, Garello F, Terreno E, Alper SL, Glaß H, Pelzl L, Akgün K, Ziemssen T, Ordemann R, Lang F, Brunati AM, Tibaldi E, Andolfo I, Iolascon A, Bertini G, Buffelli M, Zancanaro C, Lorenzetto E, Siciliano A, Bonifacio M, Danek A, Walker RH, Hermann A, De Franceschi L. Therapeutic targeting of Lyn kinase to treat chorea-acanthocytosis. Acta Neuropathol Commun 2021; 9:81. [PMID: 33941276 PMCID: PMC8091687 DOI: 10.1186/s40478-021-01181-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/14/2021] [Indexed: 11/18/2022] Open
Abstract
Chorea-Acanthocytosis (ChAc) is a devastating, little understood, and currently untreatable neurodegenerative disease caused by VPS13A mutations. Based on our recent demonstration that accumulation of activated Lyn tyrosine kinase is a key pathophysiological event in human ChAc cells, we took advantage of Vps13a−/− mice, which phenocopied human ChAc. Using proteomic approach, we found accumulation of active Lyn, γ-synuclein and phospho-tau proteins in Vps13a−/− basal ganglia secondary to impaired autophagy leading to neuroinflammation. Mice double knockout Vps13a−/− Lyn−/− showed normalization of red cell morphology and improvement of autophagy in basal ganglia. We then in vivo tested pharmacologic inhibitors of Lyn: dasatinib and nilotinib. Dasatinib failed to cross the mouse brain blood barrier (BBB), but the more specific Lyn kinase inhibitor nilotinib, crosses the BBB. Nilotinib ameliorates both Vps13a−/− hematological and neurological phenotypes, improving autophagy and preventing neuroinflammation. Our data support the proposal to repurpose nilotinib as new therapeutic option for ChAc patients.
Collapse
|
25
|
Dziurdzik SK, Conibear E. The Vps13 Family of Lipid Transporters and Its Role at Membrane Contact Sites. Int J Mol Sci 2021; 22:ijms22062905. [PMID: 33809364 PMCID: PMC7999203 DOI: 10.3390/ijms22062905] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/27/2022] Open
Abstract
The conserved VPS13 proteins constitute a new family of lipid transporters at membrane contact sites. These large proteins are suspected to bridge membranes and form a direct channel for lipid transport between organelles. Mutations in the 4 human homologs (VPS13A–D) are associated with a number of neurological disorders, but little is known about their precise functions or the relevant contact sites affected in disease. In contrast, yeast has a single Vps13 protein which is recruited to multiple organelles and contact sites. The yeast model system has proved useful for studying the function of Vps13 at different organelles and identifying the localization determinants responsible for its membrane targeting. In this review we describe recent advances in our understanding of VPS13 proteins with a focus on yeast research.
Collapse
Affiliation(s)
- Samantha Katarzyna Dziurdzik
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada;
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Elizabeth Conibear
- Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada;
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Correspondence:
| |
Collapse
|
26
|
Vaisfeld A, Bruno G, Petracca M, Bentivoglio AR, Servidei S, Vita MG, Bove F, Straccia G, Dato C, Di Iorio G, Sampaolo S, Peluso S, De Rosa A, De Michele G, Barghigiani M, Galatolo D, Tessa A, Santorelli F, Chiurazzi P, Melone MAB. Neuroacanthocytosis Syndromes in an Italian Cohort: Clinical Spectrum, High Genetic Variability and Muscle Involvement. Genes (Basel) 2021; 12:genes12030344. [PMID: 33652783 PMCID: PMC7996727 DOI: 10.3390/genes12030344] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 01/12/2023] Open
Abstract
Neuroacanthocytosis (NA) syndromes are a group of genetically defined diseases characterized by the association of red blood cell acanthocytosis, progressive degeneration of the basal ganglia and neuromuscular features with characteristic persistent hyperCKemia. The main NA syndromes include autosomal recessive chorea-acanthocytosis (ChAc) and X-linked McLeod syndrome (MLS). A series of Italian patients selected through a multicenter study for these specific neurological phenotypes underwent DNA sequencing of the VPS13A and XK genes to search for causative mutations. Where it has been possible, muscle biopsies were obtained and thoroughly investigated with histochemical assays. A total of nine patients from five different families were diagnosed with ChAC and had mostly biallelic changes in the VPS13A gene (three nonsense, two frameshift, three splicing), while three patients from a single X-linked family were diagnosed with McLeod syndrome and had a deletion in the XK gene. Despite a very low incidence (only one thousand cases of ChAc and a few hundred MLS cases reported worldwide), none of the 8 VPS13A variants identified in our patients is shared by two families, suggesting the high genetic variability of ChAc in the Italian population. In our series, in line with epidemiological data, McLeod syndrome occurs less frequently than ChAc, although it can be easily suspected because of its X-linked mode of inheritance. Finally, histochemical studies strongly suggest that muscle pathology is not simply secondary to the axonal neuropathy, frequently seen in these patients, but primary myopathic alterations can be detected in both NA syndromes.
Collapse
Affiliation(s)
- Alessandro Vaisfeld
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Giorgia Bruno
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Interuniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (G.B.); (G.S.); (C.D.); (G.D.I.); (S.S.); (M.A.B.M.)
| | - Martina Petracca
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, UOC di Neurologia, 00168 Roma, Italy; (M.P.); (A.R.B.); (M.G.V.); (F.B.)
| | - Anna Rita Bentivoglio
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, UOC di Neurologia, 00168 Roma, Italy; (M.P.); (A.R.B.); (M.G.V.); (F.B.)
- Dipartimento Universitario di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Serenella Servidei
- Dipartimento Universitario di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, UOC di Neurofisiopatologia, 00168 Rome, Italy
| | - Maria Gabriella Vita
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, UOC di Neurologia, 00168 Roma, Italy; (M.P.); (A.R.B.); (M.G.V.); (F.B.)
| | - Francesco Bove
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, UOC di Neurologia, 00168 Roma, Italy; (M.P.); (A.R.B.); (M.G.V.); (F.B.)
- Dipartimento Universitario di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Giulia Straccia
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Interuniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (G.B.); (G.S.); (C.D.); (G.D.I.); (S.S.); (M.A.B.M.)
| | - Clemente Dato
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Interuniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (G.B.); (G.S.); (C.D.); (G.D.I.); (S.S.); (M.A.B.M.)
| | - Giuseppe Di Iorio
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Interuniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (G.B.); (G.S.); (C.D.); (G.D.I.); (S.S.); (M.A.B.M.)
| | - Simone Sampaolo
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Interuniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (G.B.); (G.S.); (C.D.); (G.D.I.); (S.S.); (M.A.B.M.)
| | - Silvio Peluso
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Federico II University, 80138 Naples, Italy; (S.P.); (A.D.R.); (G.D.M.)
| | - Anna De Rosa
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Federico II University, 80138 Naples, Italy; (S.P.); (A.D.R.); (G.D.M.)
| | - Giuseppe De Michele
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, Federico II University, 80138 Naples, Italy; (S.P.); (A.D.R.); (G.D.M.)
| | - Melissa Barghigiani
- Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy; (M.B.); (D.G.); (A.T.); (F.S.)
| | - Daniele Galatolo
- Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy; (M.B.); (D.G.); (A.T.); (F.S.)
| | - Alessandra Tessa
- Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy; (M.B.); (D.G.); (A.T.); (F.S.)
| | - Filippo Santorelli
- Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy; (M.B.); (D.G.); (A.T.); (F.S.)
| | - Pietro Chiurazzi
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, UOC Genetica Medica, 00168 Roma, Italy
- Correspondence: ; Tel.: +39-338-8361006
| | - Mariarosa Anna Beatrice Melone
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Interuniversity Center for Research in Neurosciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (G.B.); (G.S.); (C.D.); (G.D.I.); (S.S.); (M.A.B.M.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA 19122-6078, USA
| |
Collapse
|
27
|
Park JS, Neiman AM. XK is a partner for VPS13A: a molecular link between Chorea-Acanthocytosis and McLeod Syndrome. Mol Biol Cell 2020; 31:2425-2436. [PMID: 32845802 PMCID: PMC7851852 DOI: 10.1091/mbc.e19-08-0439-t] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Vps13 is a highly conserved lipid transfer protein found at multiple interorganelle membrane contact sites where it mediates distinct processes. In yeast, recruitment of Vps13 to different contact sites occurs via various partner proteins. In humans, four VPS13 family members, A-D, are associated with different diseases. In particular, vps13A mutants result in the neurodegenerative disorder Chorea-Acanthocytosis (ChAc). ChAc phenotypes resemble those of McLeod Syndrome, caused by mutations in the XK gene, suggesting that XK could be a partner protein for VPS13A. XK does, in fact, exhibit hallmarks of a VPS13A partner: it forms a complex with VPS13A in human cells and, when overexpressed, relocalizes VPS13A from lipid droplets to subdomains of the endoplasmic reticulum. Introduction of two different ChAc disease-linked missense mutations into VPS13A prevents this XK-induced relocalization. These results suggest that dysregulation of a VPS13A-XK complex is the common basis for ChAc and McLeod Syndrome.
Collapse
Affiliation(s)
- Jae-Sook Park
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Aaron M Neiman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| |
Collapse
|
28
|
Hosseinzadeh Z, Hauser S, Singh Y, Pelzl L, Schuster S, Sharma Y, Höflinger P, Zacharopoulou N, Stournaras C, Rathbun DL, Zrenner E, Schöls L, Lang F. Decreased Na +/K + ATPase Expression and Depolarized Cell Membrane in Neurons Differentiated from Chorea-Acanthocytosis Patients. Sci Rep 2020; 10:8391. [PMID: 32439941 PMCID: PMC7242441 DOI: 10.1038/s41598-020-64845-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 04/17/2020] [Indexed: 02/03/2023] Open
Abstract
Loss of function mutations of the chorein-encoding gene VPS13A lead to chorea-acanthocytosis (ChAc), a neurodegenerative disorder with accelerated suicidal neuronal cell death, which could be reversed by lithium. Chorein upregulates the serum and glucocorticoid inducible kinase SGK1. Targets of SGK1 include the Na+/K+-ATPase, a pump required for cell survival. To explore whether chorein-deficiency affects Na+/K+ pump capacity, cortical neurons were differentiated from iPSCs generated from fibroblasts of ChAc patients and healthy volunteers. Na+/K+ pump capacity was estimated from K+-induced whole cell outward current (pump capacity). As a result, the pump capacity was completely abolished in the presence of Na+/K+ pump-inhibitor ouabain (100 µM), was significantly smaller in ChAc neurons than in control neurons, and was significantly increased in ChAc neurons by lithium treatment (24 hours 2 mM). The effect of lithium was reversed by SGK1-inhibitor GSK650394 (24 h 10 µM). Transmembrane potential (Vm) was significantly less negative in ChAc neurons than in control neurons, and was significantly increased in ChAc neurons by lithium treatment (2 mM, 24 hours). The effect of lithium on Vm was virtually abrogated by ouabain. Na+/K+ α1-subunit transcript levels and protein abundance were significantly lower in ChAc neurons than in control neurons, an effect reversed by lithium treatment (2 mM, 24 hours). In conclusion, consequences of chorein deficiency in ChAc include impaired Na+/K+ pump capacity.
Collapse
Affiliation(s)
- Zohreh Hosseinzadeh
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany.,Department of Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Stefan Hauser
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Yogesh Singh
- Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Lisann Pelzl
- Transfusion Medicine, Medical Faculty, Eberhard Karl University, Tübingen, Germany
| | - Stefanie Schuster
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Yamini Sharma
- Department of Internal Medicine III, University of Tübingen, Tübingen, Germany
| | - Philip Höflinger
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Nefeli Zacharopoulou
- Department of Biochemistry, University of Crete Medical School, Heraklion, Greece.,Department of Vegetative and Clinical Physiology, University of Tübingen, Tübingen, Germany
| | - Christos Stournaras
- Department of Biochemistry, University of Crete Medical School, Heraklion, Greece
| | - Daniel L Rathbun
- Department of Ophthalmology, University of Tübingen, Tübingen, Germany.,Department Ophthalmology, Bionics and Vision, Henry Ford Hospital, Henry Ford, United States
| | - Eberhart Zrenner
- Department of Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Ludger Schöls
- German Center for Neurodegenerative Diseases, Tübingen, Germany.,Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Florian Lang
- Department of Vegetative and Clinical Physiology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
29
|
Tada Y, Hamaguchi T, Ikeda Y, Iwasa K, Nishida Y, Nakamura M, Sano A, Yamada M. Chorea-acanthocytosis with a novel mutation in the vacuolar protein sorting 13 homolog a gene: A case report. J Neurol Sci 2020; 412:116731. [DOI: 10.1016/j.jns.2020.116731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/06/2020] [Accepted: 02/11/2020] [Indexed: 11/27/2022]
|
30
|
Spieler D, Velayos-Baeza A, Mühlbäck A, Castrop F, Maegerlein C, Slotta-Huspenina J, Bader B, Haslinger B, Danek A. Identification of two compound heterozygous VPS13A large deletions in chorea-acanthocytosis only by protein and quantitative DNA analysis. Mol Genet Genomic Med 2020; 8:e1179. [PMID: 32056394 PMCID: PMC7507471 DOI: 10.1002/mgg3.1179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 01/09/2020] [Accepted: 01/30/2020] [Indexed: 01/04/2023] Open
Abstract
Background Chorea‐acanthocytosis (ChAc; OMIM #200150) is a rare autosomal recessive condition with onset in early adulthood that is caused by mutations in the vacuolar protein sorting 13A (VPS13A) gene encoding chorein. Several diagnostic genomic DNA (gDNA) sequencing approaches are widely used. However, their limitations appear not to be acknowledged thoroughly enough. Methods Clinically, we deployed magnetic resonance imaging, blood smear analysis, and clinical chemistry for the index patient's characterization. The molecular analysis of the index patient next to his parents covered genomic DNA (gDNA) sequencing approaches, RNA/cDNA sequencing, and chorein specific Western blot. Results We report a 33‐year‐old male patient without functional protein due to compound heterozygosity for two VPS13A large deletions of 1168 and 1823 base pairs (bp) affecting, respectively, exons 8 and 9, and exon 13. To our knowledge, this represents the first ChAc case with two compound heterozygous large deletions identified so far. Of note, standard genomic DNA (gDNA) Sanger sequencing approaches alone yielded false negative findings. Conclusion Our case demonstrates the need to carry out detection of chorein in patients suspected of having ChAc as a helpful and potentially decisive tool to establish diagnosis. Furthermore, the course of the molecular analysis in this case discloses diagnostic pitfalls in detecting some variations, such as deletions, using only standard genomic DNA (gDNA) Sanger sequencing approaches and exemplifies alternative methods, such as RNA/cDNA sequencing or qRT‐PCR analysis, necessary to avoid false negative results.
Collapse
Affiliation(s)
- Derek Spieler
- Department of Psychosomatic Medicine and Psychotherapy, Center for Mental Health, Faculty of Medicine, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany.,Institute of Epidemiology, Mental Health Research Unit, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | | - Alžbeta Mühlbäck
- kbo-Isar-Amper-Klinikum Taufkirchen (Vils), Taufkirchen (Vils), Germany.,Department of Neurology and Centre of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital Prague, Prague, Czech Republic
| | - Florian Castrop
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christian Maegerlein
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Julia Slotta-Huspenina
- Institut für Allgemeine Pathologie und Pathologische Anatomie der Technischen Universität München, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Benedikt Bader
- Neurologische Klinik und Poliklinik, Ludwigs-Maximilians Universität München, Munich, Germany
| | - Bernhard Haslinger
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwigs-Maximilians Universität München, Munich, Germany
| |
Collapse
|
31
|
Discriminating chorea-acanthocytosis from Huntington's disease with single-case voxel-based morphometry analysis. J Neurol Sci 2020; 408:116545. [DOI: 10.1016/j.jns.2019.116545] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 01/22/2023]
|
32
|
Nishida Y, Nakamura M, Urata Y, Kasamo K, Hiwatashi H, Yokoyama I, Mizobuchi M, Sakurai K, Osaki Y, Morita Y, Watanabe M, Yoshida K, Yamane K, Miyakoshi N, Okiyama R, Ueda T, Wakasugi N, Saitoh Y, Sakamoto T, Takahashi Y, Shibano K, Tokuoka H, Hara A, Monma K, Ogata K, Kakuda K, Mochizuki H, Arai T, Araki M, Fujii T, Tsukita K, Sakamaki-Tsukita H, Sano A. Novel pathogenic VPS13A gene mutations in Japanese patients with chorea-acanthocytosis. NEUROLOGY-GENETICS 2019; 5:e332. [PMID: 31192303 PMCID: PMC6515943 DOI: 10.1212/nxg.0000000000000332] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/25/2019] [Indexed: 11/15/2022]
Abstract
Objective To identify mutations in vacuolar protein sorting 13A (VPS13A) for Japanese patients with suspected chorea-acanthocytosis (ChAc). Methods We performed a comprehensive mutation screen, including sequencing and copy number variation (CNV) analysis of the VPS13A gene, and chorein Western blotting of erythrocyte ghosts. As the results of the analysis, 17 patients were molecularly diagnosed with ChAc. In addition, we investigated the distribution of VPS13A gene mutations and clinical symptoms in a total of 39 molecularly diagnosed Japanese patients with ChAc, including 22 previously reported cases. Results We identified 11 novel pathogenic mutations, including 1 novel CNV. Excluding 5 patients with the unknown symptoms, 97.1% of patients displayed various neuropsychiatric symptoms or forms of cognitive dysfunction during the course of disease. The patients carrying the 2 major mutations representing over half of the mutations, exon 60–61 deletion and exon 37 c.4411C>T (R1471X), were localized in western Japan. Conclusions We identified 13 different mutations in VPS13A, including 11 novel mutations, and verified the clinical manifestations in 39 Japanese patients with ChAc.
Collapse
Affiliation(s)
- Yoshiaki Nishida
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Masayuki Nakamura
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Yuka Urata
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Kei Kasamo
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Hanae Hiwatashi
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Izumi Yokoyama
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Masahiro Mizobuchi
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Kotaro Sakurai
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Yasushi Osaki
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Yukari Morita
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Masako Watanabe
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Kenji Yoshida
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Kiyomi Yamane
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Natsuki Miyakoshi
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Ryouichi Okiyama
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Takehiro Ueda
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Noritaka Wakasugi
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Yuji Saitoh
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Takashi Sakamoto
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Yuji Takahashi
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Ken Shibano
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Hideki Tokuoka
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Atsushi Hara
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Kazunari Monma
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Katsuhisa Ogata
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Keita Kakuda
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Hideki Mochizuki
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Takeo Arai
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Manabu Araki
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Takeshi Fujii
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Kazuto Tsukita
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Haruhi Sakamaki-Tsukita
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| | - Akira Sano
- Kagoshima University Graduate School of Medical and Dental Sciences (Y.N., M.N., Y.U., K. Kasamo, H.H., I.Y., A.S.), Department of Psychiatry, Kagoshima, Japan; Epilepsy Center (M.M.), Department of Neurology, Nakamura Memorial Hospital, Hokkaido, Japan; Department of Psychiatry and Neurology (K. Sakurai.), Hokkaido University Graduate School of Medicine, Hokkaido, Japan; Department of Neurology (Y.O., Y.M.), Kochi Medical School, Kochi, Japan; Shinjyuku Neuro Clinic (M.W.), Tokyo, Japan; Department of Neurology (K. Yoshida and K. Yamane), Neurological Institute, Ohta-Atami Hospital, Fukushima, Japan; Department of Neurology (N.M., R.O.), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Division of Neurology (T.U., H.T.), Kobe University Graduate School of Medicine, Hyogo, Japan; Department of Neurology (N.W., Y.S., T.S., Y.T., M.A.), National Center of Neurology and Psychiatry Hospital, Tokyo, Japan; Department of Neurology (K. Shibano), Akita Red Cross Hospital, Japan; Amagasaki General Medical Center (A.H.), Hyogo, Japan; Department of Neurology (K.M., K.O.), National Hospital Organization Higashisaitama National Hospital, Saitama, Japan; Department of Neurology (K. Kakuda, H.M.), Graduate School of Medicine, Osaka University, Japan; Ikebe Clinic (T.A.), Shizuoka, Japan; Department of Psychiatry (T.F.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (K.T., H.S.-T.), Tenri Hospital, Nara, Japan; and Department of Neurology (K.T., H.S.-T.), Graduate School of Medicine, Kyoto University, Japan
| |
Collapse
|
33
|
Muñoz-Braceras S, Tornero-Écija AR, Vincent O, Escalante R. VPS13A is closely associated with mitochondria and is required for efficient lysosomal degradation. Dis Model Mech 2019; 12:dmm036681. [PMID: 30709847 PMCID: PMC6398486 DOI: 10.1242/dmm.036681] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/22/2019] [Indexed: 12/11/2022] Open
Abstract
Members of the VPS13 family are associated with various human diseases. In particular, the loss of function of VPS13A leads to chorea-acanthocytosis (ChAc), a rare neurodegenerative disease without available curative treatments. Autophagy has been considered a promising therapeutic target because the absence of VPS13A causes a defective autophagy flux. However, the mechanistic details of this deficiency are unknown. Here, we identified Rab7A as an interactor of one of the VPS13 family members in Dictyostelium discoideum and showed that this interaction is conserved between the human homologs VPS13A and RAB7A in HeLa cells. As RAB7A is a key player in endosome trafficking, we addressed the possible function of VPS13A in endosome dynamics and lysosome degradation. Our results suggest that the decrease in autophagy observed in the absence of VPS13A may be the result of a more general defect in endocytic trafficking and lysosomal degradation. Unexpectedly, we found that VPS13A is closely localized to mitochondria, suggesting that the role of VPS13A in the endolysosomal pathway might be related to inter-organelle communication. We show that VPS13A localizes at the interface between mitochondria-endosomes and mitochondria-endoplasmic reticulum and that the presence of membrane contact sites is altered in the absence of VPS13A. Based on these findings, we propose that therapeutic strategies aimed at modulating the endolysosomal pathway could be beneficial in the treatment of ChAc.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sandra Muñoz-Braceras
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| | - Alba R Tornero-Écija
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| | - Olivier Vincent
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| | - Ricardo Escalante
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| |
Collapse
|
34
|
Mente K, Kim SA, Grunseich C, Hefti MM, Crary JF, Danek A, Karp BI, Walker RH. Hippocampal sclerosis and mesial temporal lobe epilepsy in chorea-acanthocytosis: a case with clinical, pathologic and genetic evaluation. Neuropathol Appl Neurobiol 2019; 43:542-546. [PMID: 28398599 DOI: 10.1111/nan.12403] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- K Mente
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - S A Kim
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - C Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - M M Hefti
- Departments of Pathology and Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J F Crary
- Departments of Pathology and Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A Danek
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - B I Karp
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - R H Walker
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
35
|
Liu J, Heinsen H, Grinberg LT, Alho E, Amaro E, Pasqualucci CA, Rüb U, Seidel K, den Dunnen W, Arzberger T, Schmitz C, Kiessling MC, Bader B, Danek A. Pathoarchitectonics of the cerebral cortex in chorea-acanthocytosis and Huntington's disease. Neuropathol Appl Neurobiol 2018; 45:230-243. [PMID: 29722054 DOI: 10.1111/nan.12495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 02/27/2018] [Indexed: 11/29/2022]
Abstract
AIMS Quantitative estimation of cortical neurone loss in cases with chorea-acanthocytosis (ChAc) and its impact on laminar composition. METHODS We used unbiased stereological tools to estimate the degree of cortical pathology in serial gallocyanin-stained brain sections through the complete hemispheres of three subjects with genetically verified ChAc and a range of disease durations. We compared these results with our previous data of five Huntington's disease (HD) and five control cases. Pathoarchitectonic changes were exemplarily documented in TE1 of a 61-year-old female HD-, a 60-year-old female control case, and ChAc3. RESULTS Macroscopically, the cortical volume of our ChAc cases (ChAc1-3) remained close to normal. However, the average number of neurones was reduced by 46% in ChAc and by 33% in HD (P = 0.03 for ChAc & HD vs. controls; P = 0.64 for ChAc vs. HD). Terminal HD cases featured selective laminar neurone loss with pallor of layers III, V and VIa, a high density of small, pale, closely packed radial fibres in deep cortical layers VI and V, shrinkage, and chromophilia of subcortical white matter. In ChAc, pronounced diffuse astrogliosis blurred the laminar borders, thus masking the complete and partial loss of pyramidal cells in layer IIIc and of neurones in layers III, V and VI. CONCLUSION ChAc is a neurodegenerative disease with distinct cortical neurodegeneration. The hypertrophy of the peripheral neuropil space of minicolumns with coarse vertical striation was characteristic of ChAc. The role of astroglia in the pathogenesis of this disorder remains to be elucidated.
Collapse
Affiliation(s)
- J Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, München, Germany
| | - H Heinsen
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany.,Ageing Brain Study Group, Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - L T Grinberg
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - E Alho
- Praça Amadeu Amaral, São Paulo, Brazil
| | - E Amaro
- Department of Radiology, University of São Paulo Medical School, São Paulo, Brazil
| | - C A Pasqualucci
- Ageing Brain Study Group, Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - U Rüb
- Experimental Neurobiology (Anatomical Institute II), Goethe-University, Frankfurt/Main, Germany
| | - K Seidel
- Experimental Neurobiology (Anatomical Institute II), Goethe-University, Frankfurt/Main, Germany.,Anatomy & Cell Biology, Medical Faculty, Anatomical Institute, University of Bonn, Bonn, Germany
| | - W den Dunnen
- Department of Pathology and Medical Biology, University Medical Center Groningen University of Groningen, Groningen, The Netherlands
| | - T Arzberger
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany.,Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - C Schmitz
- Department of Neuroanatomy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - M C Kiessling
- Department of Neuroanatomy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - B Bader
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, München, Germany.,Clienia Privatklinik für Psychiatrie und Psychotherapie, Oetwil am See, Switzerland
| | - A Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, München, Germany
| |
Collapse
|
36
|
Liu J, Heinsen H, Grinberg LT, Alho E, Amaro E, Pasqualucci CA, Rüb U, den Dunnen W, Arzberger T, Schmitz C, Kiessling M, Bader B, Danek A. Subcortical neurodegeneration in chorea: Similarities and differences between chorea-acanthocytosis and Huntington's disease. Parkinsonism Relat Disord 2018; 49:54-59. [PMID: 29402698 DOI: 10.1016/j.parkreldis.2018.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 12/05/2017] [Accepted: 01/07/2018] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Chorea-acanthocytosis (ChAc) and Huntington's disease (HD) are neurodegenerative conditions that share clinical and neuropathological features, despite their distinct genetic etiologies. METHODS In order to compare these neuropathologies, serial gallocyanin-stained brain sections from three subjects with ChAc were analyzed and compared with our previous studies of eight HD cases, in addition to three hemispheres from two male controls. RESULTS Astrogliosis was much greater in the ChAc striatum, as compared to that found in HD, with dramatic increase in total striatal glia numbers and the number of glia per striatal neuron. Striatal astrocytes are most likely derived from the striatal subependymal layer in ChAc, which showed massive proliferation. The thalamic centromedian-parafascicular complex is reciprocally connected to the striatum and is more heavily affected in HD than in ChAc. CONCLUSION The distinct patterns of selective vulnerability and gliosis observed in HD and ChAc challenge simplistic views on the pathogenesis of these two diseases with rather similar clinical signs. The particular roles played by astroglia in ChAc and in HD clearly need to be elucidated in more detail.
Collapse
Affiliation(s)
- Jia Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China; Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Germany.
| | - Helmut Heinsen
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Germany; Departments of Pathology and Radiology, University of São Paulo Medical School, São Paulo, Brazil.
| | - Lea T Grinberg
- Department of Neurology, University of California, San Francisco, USA
| | - Eduardo Alho
- Praça Amadeu Amaral, 27, cj 31, Bela Vista, São Paulo, Brazil
| | - Edson Amaro
- Department of Radiology, University of São Paulo Medical School, São Paulo, Brazil
| | - Carlos A Pasqualucci
- Departments of Pathology and Radiology, University of São Paulo Medical School, São Paulo, Brazil
| | - Udo Rüb
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe-Universität, Frankfurt/Main, Germany
| | - Wilfred den Dunnen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Germany; Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Germany
| | - Christoph Schmitz
- Department of Neuroanatomy, Ludwig-Maximilians-Universität München, Germany
| | - Maren Kiessling
- Department of Neuroanatomy, Ludwig-Maximilians-Universität München, Germany
| | - Benedikt Bader
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Germany; Clienia Privatklinik für Psychiatrie und Psychotherapie, Oetwil am See, Switzerland
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Germany
| |
Collapse
|
37
|
Neuronal Dysfunction in iPSC-Derived Medium Spiny Neurons from Chorea-Acanthocytosis Patients Is Reversed by Src Kinase Inhibition and F-Actin Stabilization. J Neurosci 2017; 36:12027-12043. [PMID: 27881786 DOI: 10.1523/jneurosci.0456-16.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 09/07/2016] [Accepted: 09/26/2016] [Indexed: 11/21/2022] Open
Abstract
Chorea-acanthocytosis (ChAc) is a fatal neurological disorder characterized by red blood cell acanthocytes and striatal neurodegeneration. Recently, severe cell membrane disturbances based on depolymerized cortical actin and an elevated Lyn kinase activity in erythrocytes from ChAc patients were identified. How this contributes to the mechanism of neurodegeneration is still unknown. To gain insight into the pathophysiology, we established a ChAc patient-derived induced pluripotent stem cell model and an efficient differentiation protocol providing a large population of human striatal medium spiny neurons (MSNs), the main target of neurodegeneration in ChAc. Patient-derived MSNs displayed enhanced neurite outgrowth and ramification, whereas synaptic density was similar to controls. Electrophysiological analysis revealed a pathologically elevated synaptic activity in ChAc MSNs. Treatment with the F-actin stabilizer phallacidin or the Src kinase inhibitor PP2 resulted in the significant reduction of disinhibited synaptic currents to healthy control levels, suggesting a Src kinase- and actin-dependent mechanism. This was underlined by increased G/F-actin ratios and elevated Lyn kinase activity in patient-derived MSNs. These data indicate that F-actin stabilization and Src kinase inhibition represent potential therapeutic targets in ChAc that may restore neuronal function. SIGNIFICANCE STATEMENT Chorea-acanthocytosis (ChAc) is a fatal neurodegenerative disease without a known cure. To gain pathophysiological insight, we newly established a human in vitro model using skin biopsies from ChAc patients to generate disease-specific induced pluripotent stem cells (iPSCs) and developed an efficient iPSC differentiation protocol providing striatal medium spiny neurons. Using patch-clamp electrophysiology, we detected a pathologically enhanced synaptic activity in ChAc neurons. Healthy control levels of synaptic activity could be restored by treatment of ChAc neurons with the F-actin stabilizer phallacidin and the Src kinase inhibitor PP2. Because Src kinases are involved in bridging the membrane to the actin cytoskeleton by membrane protein phosphorylation, our data suggest an actin-dependent mechanism of this dysfunctional phenotype and potential treatment targets in ChAc.
Collapse
|
38
|
Lithium Sensitive ORAI1 Expression, Store Operated Ca 2+ Entry and Suicidal Death of Neurons in Chorea-Acanthocytosis. Sci Rep 2017; 7:6457. [PMID: 28743945 PMCID: PMC5526875 DOI: 10.1038/s41598-017-06451-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/13/2017] [Indexed: 12/11/2022] Open
Abstract
Chorea-Acanthocytosis (ChAc), a neurodegenerative disorder, results from loss-of-function-mutations of chorein-encoding gene VPS13A. In tumour cells chorein up-regulates ORAI1, a Ca2+-channel accomplishing store operated Ca2+-entry (SOCE) upon stimulation by STIM1. Furthermore SOCE could be up-regulated by lithium. The present study explored whether SOCE impacts on neuron apoptosis. Cortical neurons were differentiated from induced pluripotent stem cells generated from fibroblasts of ChAc patients and healthy volunteers. ORAI1 and STIM1 transcript levels and protein abundance were estimated from qRT-PCR and Western blotting, respectively, cytosolic Ca2+-activity ([Ca2+]i) from Fura-2-fluorescence, as well as apoptosis from annexin-V-binding and propidium-iodide uptake determined by flow cytometry. As a result, ORAI1 and STIM1 transcript levels and protein abundance and SOCE were significantly smaller and the percentage apoptotic cells significantly higher in ChAc neurons than in control neurons. Lithium treatment (2 mM, 24 hours) increased significantly ORAI1 and STIM1 transcript levels and protein abundance, an effect reversed by inhibition of Serum & Glucocorticoid inducible Kinase 1. ORAI1 blocker 2-APB (50 µM, 24 hours) significantly decreased SOCE, markedly increased apoptosis and abrogated the anti-apoptotic effect of lithium. In conclusion, enhanced neuronal apoptosis in ChAc at least partially results from decreased ORAI1 expression and SOCE, which could be reversed by lithium treatment.
Collapse
|
39
|
Shen Y, Liu X, Long X, Han C, Wan F, Fan W, Guo X, Ma K, Guo S, Wang L, Xia Y, Liu L, Huang J, Lin Z, Xiong N, Wang T. Novel VPS13A Gene Mutations Identified in Patients Diagnosed with Chorea-acanthocytosis (ChAc): Case Presentation and Literature Review. Front Aging Neurosci 2017; 9:95. [PMID: 28446873 PMCID: PMC5388735 DOI: 10.3389/fnagi.2017.00095] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/27/2017] [Indexed: 11/13/2022] Open
Abstract
Chorea-acanthocytosis (ChAc) is a rare autosomal recessive inherited syndrome characterized by hyperkinetic movements, seizures, cognitive impairment, neuropsychiatric symptoms, elevated serum biochemical indicators and acanthocytes detection in peripheral blood smear. Vacuolar protein sorting 13A (VPS13A) gene mutations have been proven to be genetically responsible for the pathogenesis of ChAc. Herein, based on the typical clinical symptoms and neuroimaging features, we present two suspected ChAc cases which are further genetically confirmed by four novel VPS13A gene mutations. Nevertheless, the sharp contrast between the population base and published ChAc reports implies that ChAc is considerably underdiagnosed in China. Therefore, we conclude several suggestive features and propose a diagnostic path of ChAc from a clinical, genetic and neuroimaging perspective, aiming to facilitate the diagnosis and management of ChAc in China.
Collapse
Affiliation(s)
- Yan Shen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Xiaoming Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Xi Long
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Chao Han
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Fang Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Wenliang Fan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Xingfang Guo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Kai Ma
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Shiyi Guo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Luxi Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Ling Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Zhicheng Lin
- Department of Psychiatry, Division of Alcohol and Drug Abuse, and Mailman Neuroscience Research Center, McLean Hospital, Harvard Medical School, BelmontMA, USA
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| |
Collapse
|
40
|
Pappas SS, Bonifacino J, Danek A, Dauer WT, De M, De Franceschi L, DiPaolo G, Fuller R, Haucke V, Hermann A, Kornmann B, Landwehrmeyer B, Levin J, Neiman AM, Rudnicki DD, Sibon O, Velayos-Baeza A, Vonk JJ, Walker RH, Weisman LS, Albin RL. Eighth International Chorea-Acanthocytosis Symposium: Summary of Workshop Discussion and Action Points. Tremor Other Hyperkinet Mov (N Y) 2017; 7:428. [PMID: 28224046 PMCID: PMC5313633 DOI: 10.7916/d8xd127w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/10/2017] [Indexed: 02/05/2023] Open
Abstract
Chorea-Acanthocytosis (ChAc) is a rare hereditary neurological disorder characterized by abnormal movements, red blood cell pathology, and progressive neurodegeneration. Little is understood of the pathogenesis of ChAc and related disorders (collectively Neuroacanthocytosis). The Eighth International Chorea-Acanthocytosis Symposium was held in May 2016 in Ann Arbor, MI, USA, and focused on molecular mechanisms driving ChAc pathophysiology. Accompanying the meeting, members of the neuroacanthocytosis research community and other invited scientists met in a workshop to discuss the current understanding and next steps needed to better understand ChAc pathogenesis. These discussions identified several broad and critical needs for advancing ChAc research and patient care, and led to the definition of 18 specific action points related to functional and molecular studies, animal models, and clinical research. These action points, described below, represent tractable research goals to pursue for the next several years.
Collapse
Affiliation(s)
- Samuel S. Pappas
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Juan Bonifacino
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, Germany
| | - William T. Dauer
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neurology Service, VAAAHS, University of Michigan, Ann Arbor, MI, USA
- Udall Centre, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Mithu De
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Lucia De Franceschi
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | | | - Robert Fuller
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Volker Haucke
- Department of Molecular Pharmacology and Cell Biology, Leibniz Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Andreas Hermann
- Department of Neurology, Technische Universität, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | | | | | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Aaron M. Neiman
- Department of Biochemistry and Cell Biology, Stony Brook University, New York, NY, USA
| | | | - Ody Sibon
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Gronigen, The Netherlands
| | | | - Jan J. Vonk
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Gronigen, The Netherlands
| | - Ruth H. Walker
- Department of Neurology, James J. Peters VAMC, Bronx, NY, USA
- Department of Neurology, Mount Sinai School of Medicine, New York, NY, USA
| | - Lois S. Weisman
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Roger L. Albin
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neurology Service, VAAAHS, University of Michigan, Ann Arbor, MI, USA
- Udall Centre, University of Michigan, Ann Arbor, MI, USA
- GRECC, VAAAHS, University of Michigan, Ann Arbor, MI, USA,
- Michigan Alzheimer's Disease Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
41
|
De M, Oleskie AN, Ayyash M, Dutta S, Mancour L, Abazeed ME, Brace EJ, Skiniotis G, Fuller RS. The Vps13p-Cdc31p complex is directly required for TGN late endosome transport and TGN homotypic fusion. J Cell Biol 2017; 216:425-439. [PMID: 28122955 PMCID: PMC5294781 DOI: 10.1083/jcb.201606078] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/04/2016] [Accepted: 01/11/2017] [Indexed: 01/09/2023] Open
Abstract
VPS13 proteins are widely conserved in eukaryotes and associated with human neurodegenerative and neurodevelopmental diseases. De et al. describe the lipid specificity and structure of yeast Vps13p, providing insight into its role in both TGN late endosome transport and TGN homotypic fusion. Yeast VPS13 is the founding member of a eukaryotic gene family of growing interest in cell biology and medicine. Mutations in three of four human VPS13 genes cause autosomal recessive neurodegenerative or neurodevelopmental disease, making yeast Vps13p an important structural and functional model. Using cell-free reconstitution with purified Vps13p, we show that Vps13p is directly required both for transport from the trans-Golgi network (TGN) to the late endosome/prevacuolar compartment (PVC) and for TGN homotypic fusion. Vps13p must be in complex with the small calcium-binding protein Cdc31p to be active. Single-particle electron microscopic analysis of negatively stained Vps13p indicates that this 358-kD protein is folded into a compact rod-shaped density (20 × 4 nm) with a loop structure at one end with a circular opening ∼6 nm in diameter. Vps13p exhibits ATP-stimulated binding to yeast membranes and specific interactions with phosphatidic acid and phosphorylated forms of phosphatidyl inositol at least in part through the binding affinities of conserved N- and C-terminal domains.
Collapse
Affiliation(s)
- Mithu De
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Austin N Oleskie
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Mariam Ayyash
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Somnath Dutta
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Liliya Mancour
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Mohamed E Abazeed
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Eddy J Brace
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Georgios Skiniotis
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Robert S Fuller
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
42
|
Lupo F, Tibaldi E, Matte A, Sharma AK, Brunati AM, Alper SL, Zancanaro C, Benati D, Siciliano A, Bertoldi M, Zonta F, Storch A, Walker RH, Danek A, Bader B, Hermann A, De Franceschi L. A new molecular link between defective autophagy and erythroid abnormalities in chorea-acanthocytosis. Blood 2016; 128:2976-2987. [PMID: 27742708 PMCID: PMC5179337 DOI: 10.1182/blood-2016-07-727321] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 09/24/2016] [Indexed: 01/04/2023] Open
Abstract
Chorea-acanthocytosis is one of the hereditary neurodegenerative disorders known as the neuroacanthocytoses. Chorea-acanthocytosis is characterized by circulating acanthocytes deficient in chorein, a protein of unknown function. We report here for the first time that chorea-acanthocytosis red cells are characterized by impaired autophagy, with cytoplasmic accumulation of active Lyn and of autophagy-related proteins Ulk1 and Atg7. In chorea-acanthocytosis erythrocytes, active Lyn is sequestered by HSP90-70 to form high-molecular-weight complexes that stabilize and protect Lyn from its proteasomal degradation, contributing to toxic Lyn accumulation. An interplay between accumulation of active Lyn and autophagy was found in chorea-acanthocytosis based on Lyn coimmunoprecipitation with Ulk1 and Atg7 and on the presence of Ulk1 in Lyn-containing high-molecular-weight complexes. In addition, chorein associated with Atg7 in healthy but not in chorea-acanthocytosis erythrocytes. Electron microscopy detected multivesicular bodies and membrane remnants only in circulating chorea-acanthocytosis red cells. In addition, reticulocyte-enriched chorea-acanthocytosis red cell fractions exhibited delayed clearance of mitochondria and lysosomes, further supporting the impairment of authophagic flux. Because autophagy is also important in erythropoiesis, we studied in vitro CD34+-derived erythroid precursors. In chorea-acanthocytosis, we found (1) dyserythropoiesis; (2) increased active Lyn; (3) accumulation of a marker of autophagic flux and autolysososme degradation; (4) accumlation of Lamp1, a lysosmal membrane protein, and LAMP1-positive aggregates; and (5) reduced clearance of lysosomes and mitochondria. Our results uncover in chorea-acanthocytosis erythroid cells an association between accumulation of active Lyn and impaired autophagy, suggesting a link between chorein and autophagic vesicle trafficking in erythroid maturation.
Collapse
Affiliation(s)
- Francesca Lupo
- Department of Medicine, University of Verona and Azienda ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Elena Tibaldi
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Alessandro Matte
- Department of Medicine, University of Verona and Azienda ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Alok K Sharma
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | | | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Carlo Zancanaro
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Donatella Benati
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Angela Siciliano
- Department of Medicine, University of Verona and Azienda ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Mariarita Bertoldi
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Francesca Zonta
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Alexander Storch
- Center for Regenerative Therapies, and
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Dresden, Germany
- Center for Neurodegenerative Diseases, Dresden, Germany
| | - Ruth H Walker
- Department of Neurology, James J. Peters VA Medical Center, Bronx, NY
- Mount Sinai School of Medicine, New York, NY; and
| | - Adrian Danek
- Department of Neurology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Benedikt Bader
- Department of Neurology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Andreas Hermann
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Lucia De Franceschi
- Department of Medicine, University of Verona and Azienda ospedaliera Universitaria Integrata di Verona, Verona, Italy
| |
Collapse
|
43
|
Peloso GM, Lange LA, Varga TV, Nickerson DA, Smith JD, Griswold ME, Musani S, Polfus LM, Mei H, Gabriel S, Quarells RC, Altshuler D, Boerwinkle E, Daly MJ, Neale B, Correa A, Reiner AP, Wilson JG, Kathiresan S. Association of Exome Sequences With Cardiovascular Traits Among Blacks in the Jackson Heart Study. ACTA ACUST UNITED AC 2016; 9:368-74. [PMID: 27422940 DOI: 10.1161/circgenetics.116.001410] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/05/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND The correlation of null alleles with human phenotypes can provide insight into gene function in humans. In individuals of African ancestry, we set out to identify null and damaging missense variants, and test these variants for association with a range of cardiovascular phenotypes. METHODS AND RESULTS We performed whole-exome sequencing in 3223 black individuals from the Jackson Heart Study and found a total of 729 666 variant sites with minor allele frequency <5%, including 17 263 null variants and 49 929 missense variants predicted to be damaging by in silico algorithms. We tested null and damaging missense variants within each gene for association with 36 cardiovascular traits. We found 3 associations that met our prespecified level of significance (α=1.1×10(-7)). Null and damaging missense variants in PCSK9 were associated with 36 mg/dL lower low-density lipoprotein cholesterol (P=3×10(-21)). Three individuals in their 50s with complete PCSK9 deficiency (each compound heterozygote for PCSK9 p.Y142X and p.C679X) were identified, with one having a coronary artery calcification score in the 83rd percentile despite a low-density lipoprotein cholesterol of 32 mg/dL. A damaging missense variant in HBQ1 (p.G52A) was associated with a 2 pg/cell lower mean corpuscular hemoglobin (P=9×10(-13)) and rare damaging missense variants in VPS13A with higher red blood cell distribution width (P=9.9×10(-8)). CONCLUSIONS A limited number of null/damaging alleles with a large effect on cardiovascular traits were detectable in ≈3000 black individuals.
Collapse
Affiliation(s)
- Gina M Peloso
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.).
| | - Leslie A Lange
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Tibor V Varga
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Deborah A Nickerson
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Joshua D Smith
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Michael E Griswold
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Solomon Musani
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Linda M Polfus
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Hao Mei
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Stacey Gabriel
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Rakale Collins Quarells
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - David Altshuler
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Eric Boerwinkle
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Mark J Daly
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Benjamin Neale
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Adolfo Correa
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Alex P Reiner
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - James G Wilson
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.)
| | - Sekar Kathiresan
- From the Department of Biostatistics, Boston University School of Public Health, Boston, MA (G.M.P.); Center for Human Genetic Research (G.M.P., S.K.) and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA (M.J.D., B.N.); Program in Medical and Population Genetics, Broad Institute, Cambridge, MA (G.M.P., S.G., D.A., M.J.D., B.N., S.K.); Department of Genetics, University of North Carolina, Chapel Hill, NC (L.A.L.); Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden (T.V.V.); Department of Genome Sciences, University of Washington, Seattle, WA (D.A.N., J.D.S.); Center of Biostatistics & Bioinformatics (M.E.G., H.M.), Department of Medicine (S.M.), Department of Pediatrics & Medicine (A.C.), and Department of Physiology & Biophysics (J.G.W.), University of Mississippi Medical Center, Jackson, MS; Human Genetics Center, University of Texas Health Science Center, Houston, TX (L.M.P., E.B.); Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA (R.C.Q.); Department of Medicine, Harvard Medical School, Boston, MA (D.A., S.K.); and Department of Epidemiology, University of Washington School of Public Health, Seattle, WA (A.P.R.).
| |
Collapse
|
44
|
Park JS, Thorsness MK, Policastro R, McGoldrick LL, Hollingsworth NM, Thorsness PE, Neiman AM. Yeast Vps13 promotes mitochondrial function and is localized at membrane contact sites. Mol Biol Cell 2016; 27:2435-49. [PMID: 27280386 PMCID: PMC4966984 DOI: 10.1091/mbc.e16-02-0112] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/03/2016] [Indexed: 12/19/2022] Open
Abstract
Loss of VPS13 produces multiple phenotypes. This study implicates VPS13 in the function of membrane contact sites and suggests that different phenotypes of the mutant result from defects in different contact sites. In yeast, mutations found in the VPS13A gene of ChAc patients have specific defects in the mitochondrial aspect of VPS13 function. The Vps13 protein family is highly conserved in eukaryotic cells. Mutations in human VPS13 genes result in a variety of diseases, such as chorea acanthocytosis (ChAc), but the cellular functions of Vps13 proteins are not well defined. In yeast, there is a single VPS13 orthologue, which is required for at least two different processes: protein sorting to the vacuole and sporulation. This study demonstrates that VPS13 is also important for mitochondrial integrity. In addition to preventing transfer of DNA from the mitochondrion to the nucleus, VPS13 suppresses mitophagy and functions in parallel with the endoplasmic reticulum–mitochondrion encounter structure (ERMES). In different growth conditions, Vps13 localizes to endosome–mitochondrion contacts and to the nuclear–vacuole junctions, indicating that Vps13 may function at membrane contact sites. The ability of VPS13 to compensate for the absence of ERMES correlates with its intracellular distribution. We propose that Vps13 is present at multiple membrane contact sites and that separation-of-function mutants are due to loss of Vps13 at specific junctions. Introduction of VPS13A mutations identified in ChAc patients at cognate sites in yeast VPS13 are specifically defective in compensating for the lack of ERMES, suggesting that mitochondrial dysfunction might be the basis for ChAc.
Collapse
Affiliation(s)
- Jae-Sook Park
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Mary K Thorsness
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| | - Robert Policastro
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Luke L McGoldrick
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Nancy M Hollingsworth
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| | - Peter E Thorsness
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| | - Aaron M Neiman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215
| |
Collapse
|
45
|
Honisch S, Yu W, Liu G, Alesutan I, Towhid ST, Tsapara A, Schleicher S, Handgretinger R, Stournaras C, Lang F. Chorein addiction in VPS13A overexpressing rhabdomyosarcoma cells. Oncotarget 2016; 6:10309-19. [PMID: 25871399 PMCID: PMC4496357 DOI: 10.18632/oncotarget.3582] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 02/13/2015] [Indexed: 12/12/2022] Open
Abstract
Chorein encoded by VPS13A (vacuolar protein sorting-associated protein 13A) is defective in chorea-acanthocytosis. Chorein fosters neuronal cell survival, cortical actin polymerization and cell stiffness. In view of its anti-apoptotic effect in neurons, we explored whether chorein is expressed in cancer cells and influences cancer cell survival. RT-PCR was employed to determine transcript levels, specific siRNA to silence chorein, FACS analysis to follow apoptosis and Western blotting to quantify protein abundance. Chorein transcripts were detected in various cancer cell types. The mRNA coding for chorein and chorein protein were most abundant in drug resistant, poorly differentiated human rhabdomyosarcoma cells. Chorein silencing significantly reduced the ratio of phosphorylated (and thus activated) to total phosphoinositide 3 kinase (PI-3K), pointing to inactivation of this crucial pro-survival signaling molecule. Moreover, chorein silencing diminished transcript levels and protein expression of anti-apoptotic BCL-2 and enhanced transcript levels of pro-apoptotic Bax. Silencing of chorein in rhabdomyosarcoma cells was followed by mitochondrial depolarization, caspase 3 activation and stimulation of early and late apoptosis. In conclusion, chorein is expressed in various cancer cells. In cells with high chorein expression levels chorein silencing promotes apoptotic cell death, an effect paralleled by down-regulation of PI-3K activity and BCL-2/Bax expression ratio.
Collapse
Affiliation(s)
- Sabina Honisch
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Willi Yu
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Guilai Liu
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Ioana Alesutan
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Syeda T Towhid
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Anna Tsapara
- Department of Biochemistry, University of Crete Medical School, Heraklion, Greece
| | - Sabine Schleicher
- Department of Hematology and Oncology, Children's Hospital, University Hospital of Tuebingen, Tübingen, Germany
| | - Rupert Handgretinger
- Department of Hematology and Oncology, Children's Hospital, University Hospital of Tuebingen, Tübingen, Germany
| | - Christos Stournaras
- Department of Physiology, University of Tübingen, Tübingen, Germany.,Department of Biochemistry, University of Crete Medical School, Heraklion, Greece
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
46
|
Heterozygous Chorein Deficiency in Probable Tau-negative Early-onset Alzheimer Disease. Alzheimer Dis Assoc Disord 2016; 30:272-5. [PMID: 26825611 PMCID: PMC5035148 DOI: 10.1097/wad.0000000000000130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Muñoz-Braceras S, Calvo R, Escalante R. TipC and the chorea-acanthocytosis protein VPS13A regulate autophagy inDictyosteliumand human HeLa cells. Autophagy 2015; 11:918-27. [DOI: 10.1080/15548627.2015.1034413] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
48
|
Nagy A, Noyce A, Velayos-Baeza A, Lees AJ, Warner TT, Ling H. Late Emergence of Parkinsonian Phenotype and Abnormal Dopamine Transporter Scan in Chorea-Acanthocytosis. Mov Disord Clin Pract 2015; 2:182-186. [PMID: 30713892 DOI: 10.1002/mdc3.12138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/14/2014] [Accepted: 11/21/2014] [Indexed: 11/05/2022] Open
Abstract
Chorea-acanthocytosis (ChAc) is a neurodegenerative condition predominantly manifesting with chorea and often acanthocytes on peripheral blood film. Abnormal appearances with 123I-FP-CIT single-photon emission computed tomography (SPECT) have not previously been reported in ChAc. We describe 2 cases with typical presentations of ChAc and late development of parkinsonism with asymmetric reduction in presynaptic striatal uptake on 123I-FP-CIT SPECT. Case 1, a 50-year-old male, developed micrographia and limb bradykinesia 14 years after initial presentation at the age of 30. Case 2, a 42-year-old female presenting with vocal tics and generalized dystonia at the age of 25, developed tremor, bradykinesia, and rigidity 11 years into the disease course. These cases represent the best description to date of the natural history of ChAc, in which the early hyperkinetic clinical syndromes give way to a parkinsonian phenotype. This is consistent with a gradual deterioration of presynaptic nigrostriatal projections, reflected in the clinical parkinsonism and abnormal 123I FP-CIT SPECT.
Collapse
Affiliation(s)
- Anna Nagy
- Reta Lila Weston Institute of Neurological Studies UCL Institute of Neurology London United Kingdom.,Barts and the London School of Medicine and Dentistry London United Kingdom
| | - Alastair Noyce
- Reta Lila Weston Institute of Neurological Studies UCL Institute of Neurology London United Kingdom.,Queen Square Brain Bank Department of Molecular Neuroscience UCL Institute of Neurology United Kingdom
| | | | - Andrew J Lees
- Reta Lila Weston Institute of Neurological Studies UCL Institute of Neurology London United Kingdom.,Queen Square Brain Bank Department of Molecular Neuroscience UCL Institute of Neurology United Kingdom
| | - Thomas T Warner
- Reta Lila Weston Institute of Neurological Studies UCL Institute of Neurology London United Kingdom.,Queen Square Brain Bank Department of Molecular Neuroscience UCL Institute of Neurology United Kingdom
| | - Helen Ling
- Reta Lila Weston Institute of Neurological Studies UCL Institute of Neurology London United Kingdom.,Queen Square Brain Bank Department of Molecular Neuroscience UCL Institute of Neurology United Kingdom
| |
Collapse
|
49
|
|
50
|
Abnormal red cell features associated with hereditary neurodegenerative disorders: the neuroacanthocytosis syndromes. Curr Opin Hematol 2015; 21:201-9. [PMID: 24626044 DOI: 10.1097/moh.0000000000000035] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW This review discusses the mechanisms involved in the generation of thorny red blood cells (RBCs), known as acanthocytes, in patients with neuroacanthocytosis, a heterogenous group of neurodegenerative hereditary disorders that include chorea-acanthocytosis (ChAc) and McLeod syndrome (MLS). RECENT FINDINGS Although molecular defects associated with neuroacanthocytosis have been identified recently, their pathophysiology and the related RBC abnormalities are largely unknown. Studies in ChAc RBCs have shown an altered association between the cytoskeleton and the integral membrane protein compartment in the absence of major changes in RBC membrane composition. In ChAc RBCs, abnormal Lyn kinase activation in a Syk-independent fashion has been reported recently, resulting in increased band 3 tyrosine phosphorylation and perturbation of the stability of the multiprotein band 3-based complexes bridging the membrane to the spectrin-based membrane skeleton. Similarly, in MLS, the absence of XK-protein, which is associated with the spectrin-actin-4.1 junctional complex, is associated with an abnormal membrane protein phosphorylation state, with destabilization of the membrane skeletal network resulting in generation of acanthocytes. SUMMARY A novel mechanism in generation of acanthocytes involving abnormal Lyn activation, identified in ChAc, expands the acanthocytosis phenomenon toward protein-protein interactions, controlled by phosphorylation-related abnormal signaling.
Collapse
|