1
|
Neuman SD, Thakur RS, Gratz SJ, O'Connor-Giles KM, Bashirullah A. Neurodegenerative and Neurodevelopmental Roles for Bulk Lipid Transporters VPS13A and BLTP2. Mov Disord 2025. [PMID: 40152532 DOI: 10.1002/mds.30178] [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: 12/19/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Bridge-like lipid transfer proteins (BLTPs) mediate bulk lipid transport at membrane contact sites. Mutations in BLTPs are linked to both early-onset neurodevelopmental and later-onset neurodegenerative diseases, including movement disorders. The tissue specificity and temporal requirements of BLTPs in disease pathogenesis remain poorly understood. OBJECTIVE The objective of this study was to determine tissue-specific and aging-dependent roles for VPS13A and BLTP2 using Drosophila models. METHODS We generated tissue-specific knockdowns of the VPS13A ortholog (Vps13) and the BLTP2 ortholog (hobbit) in neurons and muscles of Drosophila. We analyzed age-dependent locomotor behavior, neurodegeneration, and synapse development and function. RESULTS Neuron-specific loss of the VPS13A ortholog caused neurodegeneration followed by aging-dependent movement deficits and reduced lifespan, whereas muscle-specific loss affected only lifespan. In contrast, neuronal loss of the BLTP2 ortholog resulted in severe early-onset locomotor defects without neurodegeneration, whereas muscle loss impaired synaptogenesis and neurotransmission at the neuromuscular junction. CONCLUSIONS VPS13A maintains neuronal survival, whereas BLTP2 orchestrates synaptic development. The phenotypic specificity of BLTP function provides mechanistic insights into distinct disease trajectories for BLTP-associated disorders. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sarah D Neuman
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Rajan S Thakur
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| | - Scott J Gratz
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| | - Kate M O'Connor-Giles
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island, USA
| | - Arash Bashirullah
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
2
|
Neuman SD, Thakur RS, Gratz SJ, O'Connor-Giles KM, Bashirullah A. Neurodegenerative and neurodevelopmental roles for bulk lipid transporters VPS13A and BLTP2 in movement disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.30.630795. [PMID: 39803515 PMCID: PMC11722402 DOI: 10.1101/2024.12.30.630795] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Background Bridge-like lipid transfer proteins (BLTPs) mediate bulk lipid transport at membrane contact sites. Mutations in BLTPs are linked to both early-onset neurodevelopmental and later-onset neurodegenerative diseases, including movement disorders. The tissue specificity and temporal requirements of BLTPs in disease pathogenesis remain poorly understood. Objectives To determine the age-of-onset and tissue-specific roles of VPS13A and BLTP2 in movement disorder pathogenesis using Drosophila models. Methods We generated tissue-specific knockdowns of the VPS13A ortholog ( Vps13 ) and the BLTP2 ortholog ( hobbit ) in neurons and muscles of Drosophila . We analyzed age-dependent locomotor behavior, neurodegeneration, and synapse development and function. Results Neuron-specific loss of the VPS13A ortholog caused neurodegeneration followed by age- onset movement deficits and reduced lifespan, while muscle-specific loss affected only lifespan, revealing neurodegeneration and myopathy as independent comorbidities in VPS13A disease. In contrast, neuronal loss of the BLTP2 ortholog resulted in severe early-onset locomotor defects without neurodegeneration, while muscle loss impaired synaptogenesis and neurotransmission at the neuromuscular junction (NMJ). Conclusions VPS13A maintains neuronal survival, while BLTP2 orchestrates synaptic development. VPS13A function in muscle does not play a role in movement defects. The phenotypic specificity of BLTP function provides mechanistic insights into distinct disease trajectories for BLTP-associated movement disorders.
Collapse
|
3
|
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
|
4
|
García-García E, Carreras-Caballé M, Coll-Manzano A, Ramón-Lainez A, Besa-Selva G, Pérez-Navarro E, Malagelada C, Alberch J, Masana M, Rodríguez MJ. Preserved VPS13A distribution and expression in Huntington's disease: divergent mechanisms of action for similar movement disorders? Front Neurosci 2024; 18:1394478. [PMID: 38903599 PMCID: PMC11188336 DOI: 10.3389/fnins.2024.1394478] [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: 03/01/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
VPS13A disease and Huntington's disease (HD) are two basal ganglia disorders that may be difficult to distinguish clinically because they have similar symptoms, neuropathological features, and cellular dysfunctions with selective degeneration of the medium spiny neurons of the striatum. However, their etiology is different. VPS13A disease is caused by a mutation in the VPS13A gene leading to a lack of protein in the cells, while HD is due to an expansion of CAG repeat in the huntingtin (Htt) gene, leading to aberrant accumulation of mutant Htt. Considering the similarities of both diseases regarding the selective degeneration of striatal medium spiny neurons, the involvement of VPS13A in the molecular mechanisms of HD pathophysiology cannot be discarded. We analyzed the VPS13A distribution in the striatum, cortex, hippocampus, and cerebellum of a transgenic mouse model of HD. We also quantified the VPS13A levels in the human cortex and putamen nucleus; and compared data on mutant Htt-induced changes in VPS13A expression from differential expression datasets. We found that VPS13A brain distribution or expression was unaltered in most situations with a decrease in the putamen of HD patients and small mRNA changes in the striatum and cerebellum of HD mice. We concluded that the selective susceptibility of the striatum in VPS13A disease and HD may be a consequence of disturbances in different cellular processes with convergent molecular mechanisms already to be elucidated.
Collapse
Affiliation(s)
- Esther García-García
- Department of Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Barcelona, Spain
| | - Maria Carreras-Caballé
- Department of Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Albert Coll-Manzano
- Department of Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Alba Ramón-Lainez
- Department of Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Barcelona, Spain
| | - Gisela Besa-Selva
- Department of Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Barcelona, Spain
| | - Esther Pérez-Navarro
- Department of Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Barcelona, Spain
| | - Cristina Malagelada
- Department of Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Barcelona, Spain
| | - Jordi Alberch
- Department of Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Mercè Masana
- Department of Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Barcelona, Spain
| | - Manuel J. Rodríguez
- Department of Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
5
|
Chen X, Zhang P, Wang L, Zhang Y. Novel heterozygous VPS13A pathogenic variants in chorea-neuroacanthocytosis: a case report. BMC Neurol 2023; 23:350. [PMID: 37794323 PMCID: PMC10548615 DOI: 10.1186/s12883-023-03398-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 09/20/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Chorea-acanthocytosis (ChAc) is a rare hereditary autosomal recessive neurodegenerative disorder caused by pathogenic variants of the Vacuolar Protein Sorting 13 homolog A (VPS13A) gene. The variant spectrum of VPS13A has not been completely elucidated. This study reports two novel heterozygous VPS13A pathogenic variants in ChAc that expand the variant spectrum of VPS13A. CASE PRESENTATION We described a case of a 29-year-old man with typical clinical manifestations of ChAc, including chorea, orofacial lingual dyskinesia, vocal tics, elevated serum biochemical indicators, increased acanthocytes in peripheral blood, and caudate nucleus atrophy. Next-generation sequencing revealed two heterozygous variants of VPS13A: a nonsense variant (NM_033305.2: c.8215G > T, p. Glu2739Ter) and a deletion variant in the exons 25-31. CONCLUSION The identified nonsense variant gives rise to premature translation termination, while the deletion variant is expected to cause a significant in-frame deletion of amino acid residues in the encoded protein. Both variants are considered to be pathogenic and result in loss-of-function proteins. These findings have implications for the genetic counseling of patients with VPS13A variants.
Collapse
Affiliation(s)
- Xi Chen
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, No. 106 Zhongshan Er Road, Guangzhou, 510080, China
- Shantou University Medical College, Shantou, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Piao Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, No. 106 Zhongshan Er Road, Guangzhou, 510080, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Lijuan Wang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, No. 106 Zhongshan Er Road, Guangzhou, 510080, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China
| | - Yuhu Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, No. 106 Zhongshan Er Road, Guangzhou, 510080, China.
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, 510080, China.
| |
Collapse
|
6
|
Walker RH, Peikert K, Jung HH, Hermann A, Danek A. Neuroacanthocytosis Syndromes: The Clinical Perspective. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2023; 6:25152564231210339. [PMID: 38090146 PMCID: PMC10714877 DOI: 10.1177/25152564231210339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/01/2023] [Accepted: 10/11/2023] [Indexed: 09/05/2024]
Abstract
The two very rare neurodegenerative diseases historically known as the "neuroacanthocytosis syndromes" are due to mutations of either VPS13A or XK. These are phenotypically similar disorders that affect primarily the basal ganglia and hence result in involuntary abnormal movements as well as neuropsychiatric and cognitive alterations. There are other shared features such as abnormalities of red cell membranes which result in acanthocytes, whose relationship to neurodegeneration is not yet known. Recent insights into the functions of these two proteins suggest dysfunction of lipid processing and trafficking at the subcellular level and may provide a mechanism for neuronal dysfunction and death, and potentially a target for therapeutic interventions.
Collapse
Affiliation(s)
- Ruth H. Walker
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
- Department of Neurology, Mount Sinai School of Medicine, New York City, NY, USA
| | - 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, Germany
| | - Hans H. Jung
- Department of Neurology, University and University Hospital Zürich, Zürich, Switzerland
| | - Andreas Hermann
- 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
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Adrian Danek
- Neurologische Klinik, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
7
|
Molina-Ruiz RM, Looi JCL, Walterfang M, García-Saiz T, Wilkes FA, Liu LL, Velakoulis D, Perera JLC, Diaz-Marsa M. Striatal volumes as potential biomarkers in Eating Disorders: A pilot study. REVISTA DE PSIQUIATRIA Y SALUD MENTAL 2022; 15:65-73. [PMID: 35840286 DOI: 10.1016/j.rpsmen.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 01/20/2020] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Differences in bulimic and impulsive behaviours in Eating Disorders (ED) have been associated with cortico-striatal circuit dysfunction at a neurobiological level. We sought to investigate neo-striatal volume as a biomarker in ED subgroups as well as the possible relationship with trauma history. MATERIAL AND METHODS We studied 24 female patients: Anorexia Nervosa AN (n=8), Bulimia Nervosa BN (n=9), comorbid ED with borderline personality disorder (EDc; n=7), and a group of Healthy Controls (n=19). Binge eating behaviours and impulsivity scales were used to characterize our sample as well as Trauma Questionnaires and Magnetic resonance imaging (MRI) volumetric manual measurements of caudate and putamen nuclei (striatum). RESULTS Our preliminary results showed a significantly larger left putaminal volume in AN compared to the other three groups [C (p=0.008), BN (p<.001) and EDc (p=.001)] and a smaller right putaminal volume in EDc compared to controls (p=.045) and AN (p=.039). Some negative correlations were found between bilateral putaminal volumes and self-reported general and early traumatization scores. CONCLUSION This pilot study suggested that striatal volumes might differentiate AN from BN and EDc at a neurobiological level with implications for treatment strategies. Larger scale studies should be carried out that allow replication of these data.
Collapse
Affiliation(s)
- Rosa M Molina-Ruiz
- Complutense University Medical School, Hospital Clínico San Carlos, Madrid, Spain.
| | - Jeffrey C L Looi
- Research Centre for Neurosciences of Ageing, Academic Unit of Psychiatry and Addiction Medicine, Australian National University Medical School, Canberra. Hospital, Canberra, Australia
| | - Mark Walterfang
- Neuropsychiatry Unit, Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre, University of Melbourne and Northwestern Mental Health, Melbourne, Australia
| | | | - Fiona A Wilkes
- Research Centre for Neurosciences of Ageing, Academic Unit of Psychiatry and Addiction Medicine, Australian National University Medical School, Canberra. Hospital, Canberra, Australia
| | - Lena L Liu
- Research Centre for Neurosciences of Ageing, Academic Unit of Psychiatry and Addiction Medicine, Australian National University Medical School, Canberra. Hospital, Canberra, Australia
| | - Dennis Velakoulis
- Neuropsychiatry Unit, Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre, University of Melbourne and Northwestern Mental Health, Melbourne, Australia
| | | | - Marina Diaz-Marsa
- Complutense University Medical School, Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
8
|
Unraveling the Spatiotemporal Distribution of VPS13A in the Mouse Brain. Int J Mol Sci 2021; 22:ijms222313018. [PMID: 34884823 PMCID: PMC8657609 DOI: 10.3390/ijms222313018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 11/27/2022] Open
Abstract
Loss-of-function mutations in the human vacuolar protein sorting the 13 homolog A (VPS13A) gene cause Chorea-acanthocytosis (ChAc), with selective degeneration of the striatum as the main neuropathologic feature. Very little is known about the VPS13A expression in the brain. The main objective of this work was to assess, for the first time, the spatiotemporal distribution of VPS13A in the mouse brain. We found VPS13A expression present in neurons already in the embryonic stage, with stable levels until adulthood. VPS13A mRNA and protein distributions were similar in the adult mouse brain. We found a widespread VPS13A distribution, with the strongest expression profiles in the pons, hippocampus, and cerebellum. Interestingly, expression was weak in the basal ganglia. VPS13A staining was positive in glutamatergic, GABAergic, and cholinergic neurons, but rarely in glial cells. At the cellular level, VPS13A was mainly located in the soma and neurites, co-localizing with both the endoplasmic reticulum and mitochondria. However, it was not enriched in dendritic spines or the synaptosomal fraction of cortical neurons. In vivo pharmacological modulation of the glutamatergic, dopaminergic or cholinergic systems did not modulate VPS13A concentration in the hippocampus, cerebral cortex, or striatum. These results indicate that VPS13A has remarkable stability in neuronal cells. Understanding the distinct expression pattern of VPS13A can provide relevant information to unravel pathophysiological hallmarks of ChAc.
Collapse
|
9
|
Translational study of the whole transcriptome in rats and genetic polymorphisms in humans identifies LRP1B and VPS13A as key genes involved in tolerance to cocaine-induced motor disturbances. Transl Psychiatry 2020; 10:381. [PMID: 33159041 PMCID: PMC7648099 DOI: 10.1038/s41398-020-01050-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/18/2020] [Accepted: 10/05/2020] [Indexed: 12/04/2022] Open
Abstract
Motor disturbances strongly increase the burden of cocaine use disorder (CUDs). The objective of our translational study was to identify the genes and biological pathways underlying the tolerance to cocaine-induced motor effects. In a 5-day protocol measuring motor tolerance to cocaine in rats (N = 40), modeling the motor response to cocaine in patients, whole-genome RNA sequencing was conducted on the ventral and dorsal striatum to prioritize a genetic association study in 225 patients with severe CUD who underwent thorough phenotypic (cocaine-induced hyperlocomotion, CIH; and cocaine-induced stereotypies, CIS) and genotypic [571,000 polymorphisms (SNPs)] characterization. We provide a comprehensive description of the rat striatal transcriptomic response to cocaine in our paradigm. Repeated vs. acute cocaine binge administration elicited 27 differentially expressed genes in the ventral striatum and two in the dorsal striatum. One gene, Lrp1b, was differentially expressed in both regions. In patients, LRP1B was significantly associated with both CIS and CIH. CIH was also associated with VPS13A, a gene involved in a severe neurological disorder characterized by hyperkinetic movements. The LRP1B minor allele rs7568970 had a significant protective effect against CIS (558 SNPs, Bonferroni-corrected p = 0.02) that resisted adjustment for confounding factors, including the amount of cocaine use (adjusted beta = -0.965 and -2.35 for heterozygotes and homozygotes, respectively, p < 0.01). Using hypothesis-free prioritization of candidate genes along with thorough methodology in both the preclinical and human analysis pipelines, we provide reliable evidence that LRP1B and VPS13A are involved in the motor tolerance to cocaine in CUD patients, in line with their known pathophysiology.
Collapse
|
10
|
Molina-Ruiz RM, Looi JCL, Walterfang M, García-Saiz T, Wilkes FA, Liu LL, Velakoulis D, Perera JLC, Diaz-Marsa M. Striatal volumes as potential biomarkers in Eating Disorders: A pilot study. REVISTA DE PSIQUIATRIA Y SALUD MENTAL 2020; 15:S1888-9891(20)30022-7. [PMID: 32513624 DOI: 10.1016/j.rpsm.2020.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 10/02/2019] [Accepted: 01/20/2020] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Differences in bulimic and impulsive behaviours in Eating Disorders (ED) have been associated with cortico-striatal circuit dysfunction at a neurobiological level. We sought to investigate neo-striatal volume as a biomarker in ED subgroups as well as the possible relationship with trauma history. MATERIAL AND METHODS We studied 24 female patients: Anorexia Nervosa AN (n=8), Bulimia Nervosa BN (n=9), comorbid ED with borderline personality disorder (EDc; n=7), and a group of Healthy Controls (n=19). Binge eating behaviours and impulsivity scales were used to characterize our sample as well as Trauma Questionnaires and Magnetic resonance imaging (MRI) volumetric manual measurements of caudate and putamen nuclei (striatum). RESULTS Our preliminary results showed a significantly larger left putaminal volume in AN compared to the other three groups [C (p=0.008), BN (p<.001) and EDc (p=.001)] and a smaller right putaminal volume in EDc compared to controls (p=.045) and AN (p=.039). Some negative correlations were found between bilateral putaminal volumes and self-reported general and early traumatization scores. CONCLUSION This pilot study suggested that striatal volumes might differentiate AN from BN and EDc at a neurobiological level with implications for treatment strategies. Larger scale studies should be carried out that allow replication of these data.
Collapse
Affiliation(s)
- Rosa M Molina-Ruiz
- Complutense University Medical School, Hospital Clínico San Carlos, Madrid, Spain.
| | - Jeffrey C L Looi
- Research Centre for Neurosciences of Ageing, Academic Unit of Psychiatry and Addiction Medicine, Australian National University Medical School, Canberra. Hospital, Canberra, Australia
| | - Mark Walterfang
- Neuropsychiatry Unit, Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre, University of Melbourne and Northwestern Mental Health, Melbourne, Australia
| | | | - Fiona A Wilkes
- Research Centre for Neurosciences of Ageing, Academic Unit of Psychiatry and Addiction Medicine, Australian National University Medical School, Canberra. Hospital, Canberra, Australia
| | - Lena L Liu
- Research Centre for Neurosciences of Ageing, Academic Unit of Psychiatry and Addiction Medicine, Australian National University Medical School, Canberra. Hospital, Canberra, Australia
| | - Dennis Velakoulis
- Neuropsychiatry Unit, Royal Melbourne Hospital; Melbourne Neuropsychiatry Centre, University of Melbourne and Northwestern Mental Health, Melbourne, Australia
| | | | - Marina Diaz-Marsa
- Complutense University Medical School, Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
11
|
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
|
12
|
Niemelä V, Salih A, Solea D, Lindvall B, Weinberg J, Miltenberger G, Granberg T, Tzovla A, Nordin L, Danfors T, Savitcheva I, Dahl N, Paucar M. Phenotypic variability in chorea-acanthocytosis associated with novel VPS13A mutations. NEUROLOGY-GENETICS 2020; 6:e426. [PMID: 32494755 PMCID: PMC7217656 DOI: 10.1212/nxg.0000000000000426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/27/2020] [Indexed: 11/20/2022]
Abstract
Objective To perform a comprehensive characterization of a cohort of patients with chorea-acanthocytosis (ChAc) in Sweden. Methods Clinical assessments, targeted genetic studies, neuroimaging with MRI, [18F]-fluorodeoxyglucose (FDG) PET, and dopamine transporter with 123I FP-CIT (DaTscan) SPECT. One patient underwent magnetic resonance spectroscopy (MRS). Results Four patients living in Sweden but with different ethnical backgrounds were included. Their clinical features were variable. Biallelic VPS13A mutations were confirmed in all patients, including 3 novel mutations. All tested patients had either low or absent chorein levels. One patient had progressive caudate atrophy. Investigation using FDG-PET revealed severe bilateral striatal hypometabolism, and DaTscan SPECT displayed presynaptic dopaminergic deficiency in 3 patients. MRS demonstrated reduced N-acetylaspartate/creatine (Cr) ratio and mild elevation of both choline/Cr and combined glutamate and glutamine/Cr in the striatum in 1 case. One patient died during sleep, and another was treated with deep brain stimulation, which transiently attenuated feeding dystonia but not his gait disorder or chorea. Conclusions Larger longitudinal neuroimaging studies with different modalities, particularly MRS, are needed to determine their potential role as biomarkers for ChAc.
Collapse
Affiliation(s)
- Valter Niemelä
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Ammar Salih
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Daniela Solea
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Björn Lindvall
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Jan Weinberg
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Gabriel Miltenberger
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Tobias Granberg
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Aikaterini Tzovla
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Love Nordin
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Torsten Danfors
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Irina Savitcheva
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Niklas Dahl
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| | - Martin Paucar
- Department of Neurology (V.N.), Uppsala University Hospital; Department of Neurology (A.S.), Västerås Hospital, Sweden; Department of Neurology (D.S.), Gävle Hospital; Department of Neurology (B.L.), University Hospital in Örebro; Department of Neurology (J.W., M.P.), Karolinska University Hospital, Stockholm, Sweden; Department of Neurology (G.M.), Ludwig-Maximilians-Universität München, Munich, Germany; Department of Clinical Neuroscience (T.G., M.P.), Karolinska Institutet, Stockholm; Department of Radiology (T.G., A.T.), Karolinska University Hospital, Stockholm; Department of Diagnostic Medical Physics (L.N.), Karolinska University Hospital Solna, Stockholm; Division of Clinical Geriatrics (L.N.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm; Department of Surgical Sciences (T.D.), Section for Nuclear Medicine and PET, Uppsala University Hospital; Department of Medical Radiation Physics and Nuclear Medicine (I.S.), Karolinska University Hospital, Stockholm; and Department of Immunology, Genetics and Pathology (N.D.), Science for Life Laboratory, Uppsala University, Sweden
| |
Collapse
|
13
|
Suzuki F, Sato N, Sugiyama A, Iijima K, Shigemoto Y, Morimoto E, Kimura Y, Fujii H, Takahashi Y, Nakata Y, Matsuda H, Abe O. Chorea-acanthocytosis: Time-dependent changes of symptoms and imaging findings. J Neuroradiol 2020; 48:419-424. [PMID: 31889551 DOI: 10.1016/j.neurad.2019.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/10/2019] [Accepted: 11/28/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND PURPOSE Chorea-acanthocytosis, a rare neurodegenerative disease, affects both the striatum and the medial temporal lobe which may cause involuntary movements and epilepsy, respectively. We examined the imaging changes of the hippocampus/amygdala and the striatum as well as clinical symptoms. MATERIALS AND METHODS We retrospectively reviewed 29 MRI and 13 SPECT studies and the clinical findings of seven genetically confirmed chorea-acanthocytosis patients. We evaluated the time-dependent imaging changes of the hippocampus/amygdala and striatum and examined the relationships among these images and symptoms. RESULTS The initial symptom was epilepsy in four patients and involuntary movements in three patients. These symptoms were eventually noted in five and all seven patients, respectively. On MRI, most patients showed striatum atrophy before a hippocampus/amygdala abnormality emerged, but one patient showed a hippocampus/amygdala abnormality before striatum atrophy. Abnormal MRI findings of hippocampus/amygdala were noted in five patients and atrophy of striatum in all seven patients. SPECT demonstrated hypoperfusion of hippocampus/amygdala in three patients and that of striatum in all five available patients. Four patients demonstrated hypoperfusion of striatum earlier than that of hippocampus/amygdala and one patient showed hypoperfusion of both simultaneously. Many imaging abnormal lesions were accompanied by their corresponding symptoms, but not always so. CONCLUSION Striatum abnormalities were the initial imaging findings in many chorea-acanthocytosis patients, but epilepsy or hippocampus/amygdala imaging abnormalities may be the only findings at the early stage. It is important to understand the detailed clinical and imaging time courses for the diagnosis of chorea-acanthocytosis.
Collapse
Affiliation(s)
- Fumio Suzuki
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, 187-8551 Kodaira, Tokyo, Japan; Department of Radiology, Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, 113-8655 Bunkyo-ku, Tokyo, Japan
| | - Noriko Sato
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, 187-8551 Kodaira, Tokyo, Japan.
| | - Atsuhiko Sugiyama
- Department of Neurology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, 260-8670 Chiba, Chuo-ku, Japan
| | - Keiya Iijima
- Department of Neurosurgery, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, 187-8551 Kodaira, Tokyo, Japan
| | - Yoko Shigemoto
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, 187-8551 Kodaira, Tokyo, Japan
| | - Emiko Morimoto
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, 187-8551 Kodaira, Tokyo, Japan
| | - Yukio Kimura
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, 187-8551 Kodaira, Tokyo, Japan
| | - Hiroyuki Fujii
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, 187-8551 Kodaira, Tokyo, Japan
| | - Yuji Takahashi
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, 187-8551 Kodaira, Tokyo, Japan
| | - Yasuhiro Nakata
- Department of Neuroradiology, Tokyo Metropolitan Neurological Hospital, 2-6-1, Musashidai, 183-0042 Fuchu, Tokyo, Japan
| | - Hiroshi Matsuda
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, 4-1-1, Ogawahigashi, 187-8551 Kodaira, Tokyo, Japan
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, 113-8655 Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
14
|
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]
|
15
|
Criminal Behaviour Associated with a Novel Mutation in the VPS13A-Gene Causing Chorea-Acanthocytosis. Case Rep Psychiatry 2019; 2019:5947416. [PMID: 31139485 PMCID: PMC6500623 DOI: 10.1155/2019/5947416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/01/2019] [Indexed: 11/29/2022] Open
Abstract
Heralded by obsessive-compulsive disorder and anxiety, chorea-acanthocytosis may initially present in a psychiatric setting. As insidious onset of involuntary movements is commonly precipitated by dopamine blocking agents, this may not prompt further neurological investigation until symptoms progress after withdrawal of the suspected offending drug. Oromandibular dystonia and frontal disinhibition should call for early neurologic evaluation.
Collapse
|
16
|
Walker RH, Miranda M, Jung HH, Danek A. Life expectancy and mortality in chorea-acanthocytosis and McLeod syndrome. Parkinsonism Relat Disord 2019; 60:158-161. [DOI: 10.1016/j.parkreldis.2018.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 10/28/2022]
|
17
|
Fazio P, Paucar M, Svenningsson P, Varrone A. Novel Imaging Biomarkers for Huntington's Disease and Other Hereditary Choreas. Curr Neurol Neurosci Rep 2018; 18:85. [PMID: 30291526 PMCID: PMC6182636 DOI: 10.1007/s11910-018-0890-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF THE REVIEW Imaging biomarkers for neurodegenerative disorders are primarily developed with the goal to aid diagnosis, to monitor disease progression, and to assess the efficacy of disease-modifying therapies in support to clinical outcomes that may either show limited sensitivity or need extended time for their evaluation. This article will review the most recent concepts and findings in the field of neuroimaging applied to Huntington's disease and Huntington-like syndromes. Emphasis will be given to the discussion of potential pharmacodynamic biomarkers for clinical trials in Huntington's disease (HD) and of neuroimaging tools that can be used as diagnostic biomarkers in HD-like syndromes. RECENT FINDINGS Several magnetic resonance (MR) and positron emission tomography (PET) molecular imaging tools have been identified as potential pharmacodynamic biomarkers and others are in the pipeline after preclinical validation. MRI and 18F-fluorodeoxyglucose PET can be considered useful supportive diagnostic tools for the differentiation of other HD-like syndromes. New trials in HD have the primary goal to lower mutant huntingtin (mHTT) protein levels in the brain in order to reduce or alter the progression of the disease. MR and PET molecular imaging markers have been developed as tools to monitor disease progression and to evaluate treatment outcomes of disease-modifying trials in HD. These markers could be used alone or in combination for detecting structural and pharmacodynamic changes potentially associated with the lowering of mHTT.
Collapse
Affiliation(s)
- Patrik Fazio
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, R5:02 Karolinska University Hospital, SE-171 76, Stockholm, Sweden.
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.
| | - Martin Paucar
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Per Svenningsson
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Varrone
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, R5:02 Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Ortega MCP, Skármeta NP, Diaz YJ. Management of oromandibular dystonia on a chorea acanthocytosis: a brief review of the literature and a clinical case. Cranio 2016; 34:332-7. [DOI: 10.1179/2151090315y.0000000027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
21
|
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
|
22
|
Walker RH. Management of Neuroacanthocytosis Syndromes. Tremor Other Hyperkinet Mov (N Y) 2015; 5:346. [PMID: 26504667 PMCID: PMC4613733 DOI: 10.7916/d8w66k48] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 09/24/2015] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND The two core neuroacanthocytosis (NA) syndromes, chorea-acanthocytosis (ChAc) and McLeod syndrome, are progressive neurodegenerative disorders that primarily affect the basal ganglia. The characteristic phenotype comprises a variety of movement disorders including chorea, dystonia, and parkinsonism, as well as psychiatric and cognitive symptoms attributable to basal ganglia dysfunction. These disorders are symptomatically managed on a case-by-case basis, with very few practitioners seeing more than a single case in their careers. METHODS A literature search was performed on PubMed utilizing the terms neuroacanthocytosis, chorea-acanthocytosis, and McLeod syndrome, and articles were reviewed for mentions of therapies, successful or otherwise. RESULTS There have been no blinded, controlled trials and only one retrospective case series describing ChAc. The various therapies that have been used in patients with NA syndromes are summarized. DISCUSSION Management remains at present purely symptomatic, which is similar in principle to other more common basal ganglia neurodegenerative disorders such as Huntington's disease (HD) and Parkinson's disease (PD). However, there are some specific issues particular to NA syndromes that merit attention. An integrated multidisciplinary approach is the ideal management strategy for these complex and multifaceted neurodegenerative disorders.
Collapse
Affiliation(s)
- Ruth H. Walker
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
- Department of Neurology, Mount Sinai School of Medicine, New York City, NY, USA
| |
Collapse
|
23
|
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
|
24
|
Walker RH. Untangling the Thorns: Advances in the Neuroacanthocytosis Syndromes. J Mov Disord 2015; 8:41-54. [PMID: 26090076 PMCID: PMC4460540 DOI: 10.14802/jmd.15009] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 12/25/2022] Open
Abstract
There have been significant advances in neuroacanthocytosis (NA) syndromes in the past 20 years, however, confusion still exists regarding the precise nature of these disorders and the correct nomenclature. This article seeks to clarify these issues and to summarise the recent literature in the field. The four key NA syndromes are described here-chorea-acanthocytosis, McLeod syndrome, Huntington's disease-like 2, and pantothenate kinase- associated neurodegeneration. In the first two, acanthocytosis is a frequent, although not invariable, finding; in the second two, it occurs in approximately 10% of patients. Degeneration affecting the basal ganglia is the key neuropathologic finding, thus the clinical presentations can be remarkably similar. The characteristic phenotype comprises a variety of movement disorders, including chorea, dystonia, and parkinsonism, and also psychiatric and cognitive symptoms attributable to basal ganglia dysfunction. The age of onset, inheritance patterns, and ethnic background differ in each condition, providing diagnostic clues. Other investigations, including routine blood testing and neuroimaging can be informative. Genetic diagnosis, if available, provides a definitive diagnosis, and is important for genetic counseling, and hopefully molecular therapies in the future. In this article I provide a historical perspective on each NA syndrome. The first 3 disorders, chorea-acanthocytosis, McLeod syndrome, Huntington's disease-like 2, are discussed in detail, with a comprehensive review of the literature to date for each, while pantothenate kinase-associated neurodegeneration is presented in summary, as this disorder has recently been reviewed in this journal. Therapy for all of these diseases is, at present, purely symptomatic.
Collapse
Affiliation(s)
- Ruth H. Walker
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
- Department of Neurology, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
25
|
Macfarlane MD, Looi JC, Walterfang M, Spulber G, Velakoulis D, Styner M, Crisby M, Örndahl E, Erkinjuntti T, Waldemar G, Hennerici MG, Bäzner H, Blahak C, Wallin A, Wahlund LO. Shape abnormalities of the caudate nucleus correlate with poorer gait and balance: results from a subset of the LADIS study. Am J Geriatr Psychiatry 2015; 23:59-71.e1. [PMID: 23916546 PMCID: PMC4234689 DOI: 10.1016/j.jagp.2013.04.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 04/18/2013] [Accepted: 04/24/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Functional deficits seen in several neurodegenerative disorders have been linked with dysfunction in frontostriatal circuits and with associated shape alterations in striatal structures. The severity of visible white matter hyperintensities (WMHs) on magnetic resonance imaging has been found to correlate with poorer performance on measures of gait and balance. This study aimed to determine whether striatal volume and shape changes were correlated with gait dysfunction. METHODS Magnetic resonance imaging scans and clinical gait/balance data (scores from the Short Physical Performance Battery [SPPB]) were sourced from 66 subjects in the previously published LADIS trial, performed in nondisabled individuals older than age 65 years with WMHs at study entry. Data were obtained at study entry and at 3-year follow-up. Caudate nuclei and putamina were manually traced using a previously published method and volumes calculated. The relationships between volume and physical performance on the SPPB were investigated with shape analysis using the spherical harmonic shape description toolkit. RESULTS There was no correlation between the severity of WMHs and striatal volumes. Caudate nuclei volume correlated with performance on the SPPB at baseline but not at follow-up, with subsequent shape analysis showing left caudate changes occurred in areas corresponding to inputs of the dorsolateral prefrontal, premotor, and motor cortex. There was no correlation between putamen volumes and performance on the SPPB. CONCLUSION Disruption in frontostriatal circuits may play a role in mediating poorer physical performance in individuals with WMHs. Striatal volume and shape changes may be suitable biomarkers for functional changes in this population.
Collapse
Affiliation(s)
- Matthew D. Macfarlane
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychological and Addiction Medicine, Australian National University Medical School, Canberra Hospital, Canberra, ACT, Australia
| | - Jeffrey C.L. Looi
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychological and Addiction Medicine, Australian National University Medical School, Canberra Hospital, Canberra, ACT, Australia, Karolinska Institute, Department of Neurobiology, Care Science and Society, Division of Clinical Geriatrics, Stockholm, Sweden
| | - Mark Walterfang
- Melbourne Neuropsychiatry Centre, Royal Melbourne Hospital and University of Melbourne, Melbourne, VIC, Australia
| | - Gabriela Spulber
- Karolinska Institute, Department of Neurobiology, Care Science and Society, Division of Clinical Geriatrics, Stockholm, Sweden
| | - Dennis Velakoulis
- Melbourne Neuropsychiatry Centre, Royal Melbourne Hospital and University of Melbourne, Melbourne, VIC, Australia
| | - Martin Styner
- Neuroimaging Research and Analysis Laboratories, Carolina Institute of Developmental Disabilities, Departments of Psychiatry and Computer Science, University of North Carolina, Chapel Hill, NC
| | - Milita Crisby
- Karolinska Institute, Department of Neurobiology, Care Science and Society, Division of Clinical Geriatrics, Stockholm, Sweden
| | - Eva Örndahl
- Department of Clinical Science, Intervention and Technology at Karolinska Institute, Division of Medical Imaging and Technology, Stockholm, Sweden and Department of Radiology, Karolinska University Hospital in Huddinge, Stockholm, Sweden
| | - Timo Erkinjuntti
- Department of Neurological Sciences, University of Helsinki, Finland and Department of Neurology, Helsinki University Central Hospital, Finland
| | - Gunhild Waldemar
- Memory Disorders Research Group, Dept. of Neurology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Michael G. Hennerici
- Department of Neurology, Universitäts Medizin Mannheim UMM, University of Heidelberg, Mannheim, Germany
| | - Hansjörg Bäzner
- Department of Neurology, Universitäts Medizin Mannheim UMM, University of Heidelberg, Mannheim, Germany
| | - Christian Blahak
- Department of Neurology, Universitäts Medizin Mannheim UMM, University of Heidelberg, Mannheim, Germany
| | - Anders Wallin
- Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lars-Olof Wahlund
- Karolinska Institute, Department of Neurobiology, Care Science and Society, Division of Clinical Geriatrics, Stockholm, Sweden
| | | | | |
Collapse
|
26
|
Looi JC, Velakoulis D, Walterfang M, Georgiou-Karistianis N, Macfarlane MD, Power BD, Nilsson C, Styner M, Thompson PM, Van Westen D, Wilkes FA, Wahlund LO. The Australian, US, Scandinavian Imaging Exchange (AUSSIE): an innovative, virtually-integrated health research network embedded in health care. Australas Psychiatry 2014; 22:260-265. [PMID: 24554532 PMCID: PMC4276529 DOI: 10.1177/1039856214523494] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To describe the development, design and function of an innovative international clinical research network for neuroimaging research, based in Australia, within a joint state health service/medical school. This Australian, US, Scandinavian Imaging Exchange (AUSSIE) network focuses upon identifying neuroimaging biomarkers for neuropsychiatric and neurodegenerative disease. METHODS We describe a case study of the iterative development of the network, identifying characteristic features and methods which may serve as potential models for virtual clinical research networks. This network was established to analyse clinically-derived neuroimaging data relevant to neuropsychiatric and neurodegenerative disease, specifically in relation to subcortical brain structures. RESULTS The AUSSIE network has harnessed synergies from the individual expertise of the component groups, primarily clinical neuroscience researchers, to analyse a variety of clinical data. CONCLUSION AUSSIE is an active virtual clinical research network, analogous to a connectome, which is embedded in health care and has produced significant research, advancing our understanding of neuropsychiatric and neurodegenerative disease through the lens of neuroimaging.
Collapse
Affiliation(s)
- Jeffrey Cl Looi
- Academic Unit of Psychiatry and Addiction Medicine, Australian National University Medical School, Canberra, and; Mental Health Service, Australian Capital Territory Health Directorate, Canberra Hospital, Woden, ACT, Australia
| | - Dennis Velakoulis
- Neuropsychiatry Unit, Royal Melbourne Hospital, Melbourne, and; Melbourne Neuropsychiatry Centre, University of Melbourne and Northwestern Mental Health, Melbourne, VIC, Australia
| | - Mark Walterfang
- Neuropsychiatry Unit, Royal Melbourne Hospital, Melbourne, and; Melbourne Neuropsychiatry Centre, University of Melbourne and Northwestern Mental Health, Melbourne, VIC, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychology and Psychiatry, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Matthew D Macfarlane
- Academic Unit of Psychiatry and Addiction Medicine, Australian National University Medical School, Canberra, and; Mental Health Service, Australian Capital Territory Health Directorate, Canberra Hospital, Woden, ACT, Australia
| | - Brian D Power
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley; and; Peel and Rockingham Kwinana Mental Health Service, Rockingham, WA, Australia
| | - Christer Nilsson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Martin Styner
- Departments of Psychiatry and Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | - Paul M Thompson
- Imaging Genetics Center, University of Southern California (USC) Institute for Neuroimaging and Informatics, Keck School of Medicine, USC, Marina del Rey, CA, USA; Departments of Neurology and Psychiatry, University of California at Los Angeles (UCLA) School of Medicine, UCLA, CA, USA; and Departments of Neurology, Psychiatry, Engineering, Radiology and Ophthalmology, Keck School of Medicine, USC, USA
| | - Danielle Van Westen
- Center for Medical Imaging and Physiology, Skåne University Hospital, Lund, and; Diagnostic Radiology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Fiona A Wilkes
- Academic Unit of Psychiatry and Addiction Medicine, ANU Medical School, Canberra, and; Mental Health Service, ACT Health Directorate, Canberra Hospital, Woden, ACT, Australia
| | - Lars-Olof Wahlund
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
27
|
Walterfang M, van de Warrenburg BP. Cognitive impairment in “Other” movement disorders: Hidden defects and valuable clues. Mov Disord 2014; 29:694-703. [DOI: 10.1002/mds.25849] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 12/15/2022] Open
Affiliation(s)
- Mark Walterfang
- Neuropsychiatry Unit; Royal Melbourne Hospital; Melbourne Australia
- Melbourne Neuropsychiatry Center; University of Melbourne; Melbourne Australia
| | - Bart P. van de Warrenburg
- Department of Neurology; Donders Institute of Brain, Cognition, and Behavior, Radboud University Medical Center; Nijmegen the Netherlands
| |
Collapse
|
28
|
Miquel M, Spampinato U, Latxague C, Aviles-Olmos I, Bader B, Bertram K, Bhatia K, Burbaud P, Burghaus L, Cho JW, Cuny E, Danek A, Foltynie T, Garcia Ruiz PJ, Giménez-Roldán S, Guehl D, Guridi J, Hariz M, Jarman P, Kefalopoulou ZM, Limousin P, Lipsman N, Lozano AM, Moro E, Ngy D, Rodriguez-Oroz MC, Shang H, Shin H, Walker RH, Yokochi F, Zrinzo L, Tison F. Short and long term outcome of bilateral pallidal stimulation in chorea-acanthocytosis. PLoS One 2013; 8:e79241. [PMID: 24223913 PMCID: PMC3818425 DOI: 10.1371/journal.pone.0079241] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 09/19/2013] [Indexed: 11/28/2022] Open
Abstract
Background Chorea-acanthocytosis (ChAc) is a neuroacanthocytosis syndrome presenting with severe movement disorders poorly responsive to drug therapy. Case reports suggest that bilateral deep brain stimulation (DBS) of the ventro-postero-lateral internal globus pallidus (GPi) may benefit these patients. To explore this issue, the present multicentre (n=12) retrospective study collected the short and long term outcome of 15 patients who underwent DBS. Methods Data were collected in a standardized way 2-6 months preoperatively, 1-5 months (early) and 6 months or more (late) after surgery at the last follow-up visit (mean follow-up: 29.5 months). Results Motor severity, assessed by the Unified Huntington’s Disease Rating Scale-Motor Score, UHDRS-MS), was significantly reduced at both early and late post-surgery time points (mean improvement 54.3% and 44.1%, respectively). Functional capacity (UHDRS-Functional Capacity Score) was also significantly improved at both post-surgery time points (mean 75.5% and 73.3%, respectively), whereas incapacity (UHDRS-Independence Score) improvement reached significance at early post-surgery only (mean 37.3%). Long term significant improvement of motor symptom severity (≥20 % from baseline) was observed in 61.5 % of the patients. Chorea and dystonia improved, whereas effects on dysarthria and swallowing were variable. Parkinsonism did not improve. Linear regression analysis showed that preoperative motor severity predicted motor improvement at both post-surgery time points. The most serious adverse event was device infection and cerebral abscess, and one patient died suddenly of unclear cause, 4 years after surgery. Conclusion This study shows that bilateral DBS of the GPi effectively reduces the severity of drug-resistant hyperkinetic movement disorders such as present in ChAc.
Collapse
Affiliation(s)
- Marie Miquel
- Service de Neurologie, CHU Bordeaux, Bordeaux, France
- Service de Neurologie, CH François Mitterrand, Pau, France
| | - Umberto Spampinato
- Service de Neurologie, CHU Bordeaux, Bordeaux, France
- Univ.Bordeaux-INSERM U862, Neurocentre Magendie, Bordeaux, France
- * E-mail:
| | | | - Iciar Aviles-Olmos
- Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Benedikt Bader
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Kelly Bertram
- Neurosciences, Alfred Hospital, Commercial Road, Melbourne, Victoria, Australia
- Van Cleef Roet Centre for Nervous Diseases, Monash University, Melbourne, Victoria, Australia
| | - Kailash Bhatia
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London, Institute of Neurology, Queen Square, London, United Kingdom
| | - Pierre Burbaud
- Service de Neurophysiologie Clinique, CHU Bordeaux, Bordeaux, France
| | - Lothar Burghaus
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Jin Whan Cho
- Department of Neurology, Samsung Medical Centre, Sungkyunkwan University, School of Medicine, Seoul, Korea
| | - Emmanuel Cuny
- Service de Neurochirurgie, CHU Bordeaux, Bordeaux, France
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Thomas Foltynie
- Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | | | | | - Dominique Guehl
- Service de Neurophysiologie Clinique, CHU Bordeaux, Bordeaux, France
| | - Jorge Guridi
- Neurology and Neurosurgical Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Marwan Hariz
- Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Paul Jarman
- National Hospital of Neurology and Neurosurgery, Queen Square, London, United Kingdom
| | - Zinovia Maria Kefalopoulou
- Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Patricia Limousin
- Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Nir Lipsman
- Division of Neurosurgery, Toronto Western Hospital, University of Toronto, University Health Network, Toronto, Ontario, Canada
| | - Andres M. Lozano
- Division of Neurosurgery, Toronto Western Hospital, University of Toronto, University Health Network, Toronto, Ontario, Canada
| | - Elena Moro
- Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, University of Toronto, University Health Network, Toronto, Ontario, Canada
- Movement Disorders Center, Department of Psychiatry and Neurology, University Hospital Centre of Grenoble, Grenoble, France
| | - Dhita Ngy
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
- Avicenna Medical Center, New York, New York, United States of America
| | - Maria Cruz Rodriguez-Oroz
- Department of Neurology, University Hospital Donostia, Neuroscience Unit, BioDonostia Research Institute, San Sebastian, Spain
| | - Huifang Shang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hyeeun Shin
- Department of Neurology, Samsung Medical Centre, Sungkyunkwan University, School of Medicine, Seoul, Korea
| | - Ruth H. Walker
- Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States of America
| | - Fusako Yokochi
- Department of Neurology, Tokyo Metropolitan Neurological Hospital, Fuchu, Tokyo, Japan
| | - Ludvic Zrinzo
- Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - François Tison
- Service de Neurologie, CHU Bordeaux, Bordeaux, France
- Université Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
Collapse
|
29
|
Executive dysfunction correlates with caudate nucleus atrophy in patients with white matter changes on MRI: a subset of LADIS. Psychiatry Res 2013; 214:16-23. [PMID: 23916538 DOI: 10.1016/j.pscychresns.2013.05.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 04/19/2013] [Accepted: 05/17/2013] [Indexed: 11/22/2022]
Abstract
White matter changes (WMC) are common magnetic resonance imaging (MRI) findings, particularly in the elderly. Recent studies such as the Leukoaraiosis and Disability Study (LADIS) have found that WMC relate to adverse outcomes including cognitive impairment, depression, disability, unsteadiness and falls in cross-sectional and follow-up studies. Frontostriatal (or frontosubcortical) brain circuits may serve many of these functions, with the caudate nuclei playing a role in convergence of cognitive functions. This study aimed to determine whether reduced caudate volume relates to cognitive functions (executive functions, memory functions and speed of processing) and WMC. We determined caudate nuclei volumes, through manual tracing, on a subgroup of the LADIS study (n=66) from four centres with baseline and 3-year follow-up MRI scans. Regression analysis was used to assess relationships between caudate volume, cognitive function and WMC. Severity of WMC did not relate to caudate volume. Smaller caudate volumes were significantly associated with poorer executive functioning at baseline and at 3 years, but were not associated with scores of memory or speed of processing. Thus, in patients with WMC, a surrogate of small vessel disease, caudate atrophy relates to the dysexecutive syndrome, supporting the role of caudate as an important part of the frontostriatal circuit.
Collapse
|
30
|
Hayhow BD, Hassan I, Looi JCL, Gaillard F, Velakoulis D, Walterfang M. The neuropsychiatry of hyperkinetic movement disorders: insights from neuroimaging into the neural circuit bases of dysfunction. Tremor Other Hyperkinet Mov (N Y) 2013; 3:tre-03-175-4242-1. [PMID: 24032090 PMCID: PMC3760049 DOI: 10.7916/d8sn07pk] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/08/2013] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Movement disorders, particularly those associated with basal ganglia disease, have a high rate of comorbid neuropsychiatric illness. METHODS We consider the pathophysiological basis of the comorbidity between movement disorders and neuropsychiatric illness by 1) reviewing the epidemiology of neuropsychiatric illness in a range of hyperkinetic movement disorders, and 2) correlating findings to evidence from studies that have utilized modern neuroimaging techniques to investigate these disorders. In addition to diseases classically associated with basal ganglia pathology, such as Huntington disease, Wilson disease, the neuroacanthocytoses, and diseases of brain iron accumulation, we include diseases associated with pathology of subcortical white matter tracts, brain stem nuclei, and the cerebellum, such as metachromatic leukodystrophy, dentatorubropallidoluysian atrophy, and the spinocerebellar ataxias. CONCLUSIONS Neuropsychiatric symptoms are integral to a thorough phenomenological account of hyperkinetic movement disorders. Drawing on modern theories of cortico-subcortical circuits, we argue that these disorders can be conceptualized as disorders of complex subcortical networks with distinct functional architectures. Damage to any component of these complex information-processing networks can have variable and often profound consequences for the function of more remote neural structures, creating a diverse but nonetheless rational pattern of clinical symptomatology.
Collapse
Affiliation(s)
- Bradleigh D. Hayhow
- Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville, Australia
- Melbourne Neuropsychiatry Centre, University of Melbourne and Melbourne Health, Parkville, Australia
| | - Islam Hassan
- Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville, Australia
| | - Jeffrey C. L. Looi
- Academic Unit of Psychiatry & Addiction Medicine, Australian National University Medical School, Canberra Hospital, Canberra, Australia
| | | | - Dennis Velakoulis
- Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville, Australia
- Melbourne Neuropsychiatry Centre, University of Melbourne and Melbourne Health, Parkville, Australia
| | - Mark Walterfang
- Neuropsychiatry Unit, Royal Melbourne Hospital, Parkville, Australia
- Melbourne Neuropsychiatry Centre, University of Melbourne and Melbourne Health, Parkville, Australia
| |
Collapse
|
31
|
Abstract
The striatum, comprising the caudate nucleus, putamen and nucleus accumbens, occupies a strategic location within cortico-striato-pallido-thalamic-cortical (corticostriatal) re-entrant neural circuits. Striatal neurodevelopment is precisely determined by phylogenetically conserved homeobox genes. Consisting primarily of medium spiny neurons, the striatum is strictly topographically organized based on cortical afferents and efferents. Particular corticostriatal neural circuits are considered to subserve certain domains of cognition, emotion and behaviour. Thus, the striatum may serve as a map of structural change in the cortical afferent pathways owing to deafferentation or neuroplasticity, and conversely, structural change in the striatum per se may structurally disrupt corticostriatal pathways. The morphology of the striatum may be quantified in vivo using advanced magnetic resonance imaging, as may cognitive functioning pertaining to corticostriatal circuits. It is proposed that striatal morphology may be a biomarker in neurodegenerative disease and potentially the basis of an endophenotype.
Collapse
|
32
|
Looi JC, Rajagopalan P, Walterfang M, Madsen SK, Thompson PM, Macfarlane MD, Ching C, Chua P, Velakoulis D. Differential putaminal morphology in Huntington's disease, Frontotemporal dementia and Alzheimer's disease. Aust N Z J Psychiatry 2012; 46:1145-58. [PMID: 22990433 PMCID: PMC4113021 DOI: 10.1177/0004867412457224] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Direct neuronal loss or deafferentation of the putamen, a critical hub in corticostriatal circuits, may result in diverse and distinct cognitive and motoric dysfunction in neurodegenerative disease. Differential putaminal morphology, as a quantitative measure of corticostriatal integrity, may thus be evident in Huntington's disease (HD), Alzheimer's disease (AD) and frontotemporal dementia (FTD), diseases with differential clinical dysfunction. METHODS HD (n = 17), FTD (n = 33) and AD (n = 13) patients were diagnosed according to international consensus criteria and, with healthy controls (n = 17), were scanned on the same MRI scanner. Patients underwent brief cognitive testing using the Neuropsychiatry Unit Cognitive Assessment Tool (NUCOG). Ten MRI scans from this dataset were manually segmented as a training set for the Adaboost algorithm, which automatically segmented all remaining scans for the putamen, yielding the following subset of the data: 9 left and 12 right putamen segmentations for AD; 25 left and 26 right putamina for FTD; 16 left and 15 right putamina for HD; 12 left and 12 right putamina for controls. Shape analysis was performed at each point on the surface of each structure using a multiple regression controlling for age and sex to compare radial distance across diagnostic groups. RESULTS Age, but not sex and intracranial volume (ICV), were significantly different in the segmentation subgroups by diagnosis. The AD group showed significantly poorer performance on cognitive testing than FTD. Mean putaminal volumes were HD < FTD < AD ≤ controls, controlling for age and ICV. The greatest putaminal shape deflation was evident in HD, followed by FTD, in regions corresponding to the interconnections to motoric cortex. CONCLUSIONS Differential patterns of putaminal atrophy in HD, FTD and AD, with relevance to corticostriatal circuits, suggest the putamen may be a suitable clinical biomarker in neurodegenerative disease.
Collapse
Affiliation(s)
- Jeffrey C.L. Looi
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychological and Addiction Medicine, Australian National University Medical School, Canberra, A.C.T., Australia,Correspondence: Associate Professor Jeffrey Looi, Research Centre for the Neurosciences of Ageing, Academic Unit of Psychological Medicine, ANU Medical School, Building 4, Level 2, Canberra Hospital, Garran A.C.T. 2605,
| | - Priya Rajagopalan
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA, USA
| | - Mark Walterfang
- Melbourne Neuropsychiatry Centre, Royal Melbourne Hospital, & University of Melbourne, Melbourne, VIC, Australia
| | - Sarah K. Madsen
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA, USA
| | - Paul M. Thompson
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA, USA
| | - Matthew D. Macfarlane
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychological and Addiction Medicine, Australian National University Medical School, Canberra, A.C.T., Australia
| | - Chris Ching
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA, USA
| | - Phyllis Chua
- School of Psychology and Psychiatry, Monash University, Melbourne, VIC, Australia
| | - Dennis Velakoulis
- Melbourne Neuropsychiatry Centre, Royal Melbourne Hospital, & University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
33
|
Lindberg O, Walterfang M, Looi JCL, Malykhin N, Ostberg P, Zandbelt B, Styner M, Paniagua B, Velakoulis D, Orndahl E, Wahlund LO. Hippocampal shape analysis in Alzheimer's disease and frontotemporal lobar degeneration subtypes. J Alzheimers Dis 2012; 30:355-65. [PMID: 22414571 DOI: 10.3233/jad-2012-112210] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Hippocampal pathology is central to Alzheimer's disease (AD) and other forms of dementia such as frontotemporal lobar degeneration (FTLD). Autopsy studies have shown that certain hippocampal subfields are more vulnerable than others to AD and FTLD pathology, in particular the subiculum and cornu ammonis 1 (CA1). We conducted shape analysis of hippocampi segmented from structural T1 MRI images on clinically diagnosed dementia patients and controls. The subjects included 19 AD and 35 FTLD patients [13 frontotemporal dementia (FTD), 13 semantic dementia (SD), and 9 progressive nonfluent aphasia (PNFA)] and 21 controls. Compared to controls, SD displayed severe atrophy of the whole left hippocampus. PNFA and FTD also displayed atrophy on the left side, restricted to the hippocampal head in FTD. Finally, AD displayed most atrophy in left hippocampal body with relative sparing of the hippocampal head. Consistent with neuropathological studies, most atrophic deformation was found in CA1 and subiculum areas in FTLD and AD.
Collapse
Affiliation(s)
- Olof Lindberg
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Looi JCL, Walterfang M, Velakoulis D, Macfarlane MD, Svensson LA, Wahlund LO. Frontotemporal dementia as a frontostriatal disorder: neostriatal morphology as a biomarker and structural basis for an endophenotype. Aust N Z J Psychiatry 2012; 46:422-34. [PMID: 22535292 DOI: 10.1177/0004867411432076] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE This article reviews the evidence for a re-conceptualisation of a subtype of frontotemporal lobar degeneration (FTLD), frontotemporal dementia (FTD), as a frontostriatal disorder, working towards an endophenotype. METHOD We provide an overview of the role of frontostriatal circuits relevant to FTLD and FTD, as a subset of larger-scale distributed brain networks. We discuss the role of a strategic structure in these circuits, the neostriatum. Then we review the relationship of the clinical features of FTLD to frontostriatal circuits, correlating this with neuropsychological and neuropathological data. CONCLUSION The unique structure and linkages of the neostriatum make it an ideal structure for in vivo neuroimaging to understand the neuroanatomical basis of FTD. We develop a frontostriatal endophenotypic model for FTD as a platform for further investigation.
Collapse
Affiliation(s)
- Jeffrey C L Looi
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychological Medicine, Australian National University Medical School, Canberra Hospital, Canberra, Australia.
| | | | | | | | | | | |
Collapse
|
35
|
Ong D, Walterfang M, Malhi GS, Styner M, Velakoulis D, Pantelis C. Size and shape of the caudate nucleus in individuals with bipolar affective disorder. Aust N Z J Psychiatry 2012; 46:340-51. [PMID: 22368240 PMCID: PMC3328643 DOI: 10.1177/0004867412440191] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVE The caudate nucleus (CN) is a crucial component of the ventral striatum, which is part of a prefrontal-striatal-thalamic circuit that is modulated by limbic structures to subserve emotional processing. Bipolar disorder is thought to be underpinned by dysfunctional anterior limbic networks, although MRI studies examining the CN have shown equivocal results. As gross volumetric analyses may not detect subtle regional change, we aimed to clarify the role of the CN in bipolar disorder by undertaking shape analysis to detect regional reductions. METHODS The CN was manually traced on MRI scans from 27 patients with bipolar-I disorder and 24 matched controls. A non-parametric spherical harmonic shape analysis was undertaken using the SPHARM toolkit. RESULTS Whilst the left CN volume was consistently larger in the sample, there was no effect of group or gender or significant interactions between these variables. Volume did not correlate with illness duration or lithium dosage, but was larger in those with a history of psychosis at trend level. However, left caudate shape differed significantly between groups, with deflation in an area along the ventromedial surface (connecting to dorsolateral prefrontal regions) in bipolar patients. Psychotic patients showed increases in the dorsal head and body at trend level overall, in regions connecting to medial and orbitofrontal regions. CONCLUSIONS These findings suggest that subtle rather than gross structural changes occur in the CN, which may not be detectable by volumetric analysis alone, and reflect alterations in specific frontostriatal circuitry in the disorder.
Collapse
Affiliation(s)
- Daniel Ong
- Melbourne Neuropsychiatry Centre, University of Melbourne and Melbourne Health, Melbourne, Australia
| | | | | | | | | | | |
Collapse
|
36
|
Walker RH. Update on the Non-Huntington's Disease Choreas with Comments on the Current Nomenclature. Tremor Other Hyperkinet Mov (N Y) 2012; 2:tre-02-49-211-1. [PMID: 23440598 PMCID: PMC3570038 DOI: 10.7916/d89p30cs] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 08/08/2011] [Indexed: 12/23/2022] Open
Abstract
CHOREA CAN BE CAUSED BY A MULTITUDE OF ETIOLOGIES: neurodegenerative, pharmacological, structural, metabolic, and others. In absence of other apparent causes, exclusion of Huntington's disease is often a first step in the diagnostic process. There are a number of neurodegenerative disorders whose genetic etiology has been identified in the past decade. Molecular diagnosis has enabled genetic identification of disorder subtypes which were previously grouped together, such as the neurodegeneration with brain iron accumulation disorders and the neuroacanthocytosis syndromes, as well as identification of phenotypic outliers for recognized disorders. Correct molecular diagnosis is essential for genetic counseling and, hopefully, ultimately genetic therapies. In addition, there has recently been recognition of other disorders which can mimic neurodegenerative disorders, including paraneoplastic and prion disorders. This article focuses upon recent developments in the field but is not intended to provide an exhaustive review of all causes of chorea, which is available elsewhere. I also discuss the nomenclature of these disorders which has become somewhat unwieldy, but may ultimately be refined by association with the causative gene.
Collapse
Affiliation(s)
- Ruth H. Walker
- Departments of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States of America
- Mount Sinai School of Medicine, New York City, New York, United States of America
| |
Collapse
|
37
|
Föller M, Hermann A, Gu S, Alesutan I, Qadri SM, Borst O, Schmidt E, Schiele F, Hagen JMV, Saft C, Schöls L, Lerche H, Stournaras C, Storch A, Lang F. Chorein‐sensitive polymerization of cortical actin and suicidal cell death in chorea‐acanthocytosis. FASEB J 2012; 26:1526-34. [DOI: 10.1096/fj.11-198317] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Michael Föller
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Andreas Hermann
- Division of Neurodegenerative DiseasesDepartment of NeurologyUniversity of TechnologyDresdenGermany
| | - Shuchen Gu
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Ioana Alesutan
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Syed M. Qadri
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | - Oliver Borst
- Department of PhysiologyUniversity of TübingenTübingenGermany
| | | | - Franziska Schiele
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Jennifer Müller vom Hagen
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | - Carsten Saft
- Department of NeurologyRuhr UniversityBochumGermany
| | - Ludger Schöls
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- DZNEResearch Site TübingenTübingenGermany
| | - Holger Lerche
- Department of NeurologyUniversity of TübingenTübingenGermany
- Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
| | | | - Alexander Storch
- Division of Neurodegenerative DiseasesDepartment of NeurologyUniversity of TechnologyDresdenGermany
- German Center for Neurodegenerative Diseases (DZNE)Research Site DresdenDresdenGermany
| | - Florian Lang
- Department of PhysiologyUniversity of TübingenTübingenGermany
| |
Collapse
|
38
|
Janssen J, Alemán-Gómez Y, Reig S, Schnack HG, Parellada M, Graell M, Moreno C, Moreno D, Mateos-Pérez JM, Udias JM, Arango C, Desco M. Regional specificity of thalamic volume deficits in male adolescents with early-onset psychosis. Br J Psychiatry 2012; 200:30-6. [PMID: 22116979 DOI: 10.1192/bjp.bp.111.093732] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Thalamic volume deficits are associated with psychosis but it is unclear whether the volume reduction is uniformly distributed or whether it is more severe in particular thalamic regions. AIMS To quantify whole and regional thalamic volume in males with early-onset psychosis and healthy male controls. METHOD Brain scans were obtained for 80 adolescents: 46 individuals with early-onset psychosis with a duration of positive symptoms less than 6 months and 34 healthy controls. All participants were younger than 19 years. Total thalamic volumes were assessed using FreeSurfer and FSL-FIRST, group comparisons of regional thalamic volumes were studied with a surface-based approach. RESULTS Total thalamic volume was smaller in participants with early-onset psychosis relative to controls. Regional thalamic volume reduction was most significant in the right anterior mediodorsal area and pulvinar. CONCLUSIONS In males with minimally treated early-onset psychosis, thalamic volume deficits may be most pronounced in the anterior mediodorsal and posterior pulvinar regions, adding strength to findings from post-mortem studies in adults with psychosis.
Collapse
Affiliation(s)
- Joost Janssen
- Unidad de Medicina y Cirugía Experimental, Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Looi JCL, Macfarlane MD, Walterfang M, Styner M, Velakoulis D, Lätt J, van Westen D, Nilsson C. Morphometric analysis of subcortical structures in progressive supranuclear palsy: In vivo evidence of neostriatal and mesencephalic atrophy. Psychiatry Res 2011; 194:163-75. [PMID: 21899988 PMCID: PMC3204393 DOI: 10.1016/j.pscychresns.2011.07.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 06/25/2011] [Accepted: 07/02/2011] [Indexed: 01/17/2023]
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disease characterized by gait and postural disturbance, gaze palsy, apathy, decreased verbal fluency and dysexecutive symptoms, with some of these clinical features potentially having origins in degeneration of frontostriatal circuits and the mesencephalon. This hypothesis was investigated by manual segmentation of the caudate and putamen on MRI scans, using previously published protocols, in 15 subjects with PSP and 15 healthy age-matched controls. Midbrain atrophy was assessed by measurement of mid-sagittal area of the midbrain and pons. Shape analysis of the caudate and putamen was performed using spherical harmonics (SPHARM-PDM, University of North Carolina). The sagittal pons area/midbrain area ratio (P/M ratio) was significantly higher in the PSP group, consistent with previous findings. Significantly smaller striatal volumes were found in the PSP group - putamina were 10% smaller and caudate volumes were 17% smaller than in controls after controlling for age and intracranial volume. Shape analysis revealed significant shape deflation in PSP in the striatum, compared to controls; with regionally significant change relevant to frontostriatal and corticostriatal circuits in the caudate. Thus, in a clinically diagnosed and biomarker-confirmed cohort with early PSP, we demonstrate that neostriatal volume and shape are significantly reduced in vivo. The findings suggest a neostriatal and mesencephalic structural basis for the clinical features of PSP leading to frontostriatal and mesocortical-striatal circuit disruption.
Collapse
Affiliation(s)
- Jeffrey Chee Leong Looi
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychological Medicine, School of Clinical Medicine, Australian National University Medical School, Canberra, Australia, Correspondence: Associate Professor Jeffrey Looi, Academic Unit of Psychological Medicine, ANU Medical School, Building 4, Level 2, Canberra Hospital, Garran A.C.T. 2605,
| | - Matthew D. Macfarlane
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychological Medicine, School of Clinical Medicine, Australian National University Medical School, Canberra, Australia
| | - Mark Walterfang
- Melbourne Neuropsychiatry Centre, Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
| | - Martin Styner
- Department of Psychiatry and Department of Computer Science, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Dennis Velakoulis
- Melbourne Neuropsychiatry Centre, Royal Melbourne Hospital and University of Melbourne, Melbourne, Australia
| | - Jimmy Lätt
- Center for Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden
| | - Danielle van Westen
- Center for Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden,Diagnostic Radiology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Christer Nilsson
- Geriatric Psychiatry, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
40
|
Kapur T, Pieper S, Whitaker R, Aylward S, Jakab M, Schroeder W, Kikinis R. The National Alliance for Medical Image Computing, a roadmap initiative to build a free and open source software infrastructure for translational research in medical image analysis. J Am Med Inform Assoc 2011; 19:176-80. [PMID: 22081219 DOI: 10.1136/amiajnl-2011-000493] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The National Alliance for Medical Image Computing (NA-MIC), is a multi-institutional, interdisciplinary community of researchers, who share the recognition that modern health care demands improved technologies to ease suffering and prolong productive life. Organized under the National Centers for Biomedical Computing 7 years ago, the mission of NA-MIC is to implement a robust and flexible open-source infrastructure for developing and applying advanced imaging technologies across a range of important biomedical research disciplines. A measure of its success, NA-MIC is now applying this technology to diseases that have immense impact on the duration and quality of life: cancer, heart disease, trauma, and degenerative genetic diseases. The targets of this technology range from group comparisons to subject-specific analysis.
Collapse
Affiliation(s)
- Tina Kapur
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | |
Collapse
|