51
|
Martin S, Allan KC, Pinkard O, Sweet T, Tesar PJ, Coller J. Oligodendrocyte differentiation alters tRNA modifications and codon optimality-mediated mRNA decay. Nat Commun 2022; 13:5003. [PMID: 36008413 PMCID: PMC9411196 DOI: 10.1038/s41467-022-32766-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 08/15/2022] [Indexed: 11/08/2022] Open
Abstract
Oligodendrocytes are specialized cells that confer neuronal myelination in the central nervous system. Leukodystrophies associated with oligodendrocyte deficits and hypomyelination are known to result when a number of tRNA metabolism genes are mutated. Thus, for unknown reasons, oligodendrocytes may be hypersensitive to perturbations in tRNA biology. In this study, we survey the tRNA transcriptome in the murine oligodendrocyte cell lineage and find that specific tRNAs are hypomodified in oligodendrocytes within or near the anticodon compared to oligodendrocyte progenitor cells (OPCs). This hypomodified state may be the result of differential expression of key modification enzymes during oligodendrocyte differentiation. Moreover, we observe a concomitant relationship between tRNA hypomodification and tRNA decoding potential; observing oligodendrocyte specific alterations in codon optimality-mediated mRNA decay and ribosome transit. Our results reveal that oligodendrocytes naturally maintain a delicate, hypersensitized tRNA/mRNA axis. We suggest this axis is a potential mediator of pathology in leukodystrophies and white matter disease when further insult to tRNA metabolism is introduced.
Collapse
Affiliation(s)
- Sophie Martin
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kevin C Allan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Otis Pinkard
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Thomas Sweet
- Center for Proteomics and Bioinformatics, Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Jeff Coller
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
52
|
Adang L. Leukodystrophies. Continuum (Minneap Minn) 2022; 28:1194-1216. [PMID: 35938662 PMCID: PMC11320896 DOI: 10.1212/con.0000000000001130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE OF REVIEW This article reviews the most common leukodystrophies and is focused on diagnosis, clinical features, and emerging therapeutic options. RECENT FINDINGS In the past decade, the recognition of leukodystrophies has exponentially increased, and now this class includes more than 30 distinct disorders. Classically recognized as progressive and fatal disorders affecting young children, it is now understood that leukodystrophies are associated with an increasing spectrum of neurologic trajectories and can affect all ages. Next-generation sequencing and newborn screening allow the opportunity for the recognition of presymptomatic and atypical cases. These new testing opportunities, in combination with growing numbers of natural history studies and clinical consensus guidelines, have helped improve diagnosis and clinical care. Additionally, a more granular understanding of disease outcomes informs clinical trial design and has led to several recent therapeutic advances. This review summarizes the current understanding of the clinical manifestations of disease and treatment options for the most common leukodystrophies. SUMMARY As early testing becomes more readily available through next-generation sequencing and newborn screening, neurologists will better understand the true incidence of the leukodystrophies and be able to diagnose children within the therapeutic window. As targeted therapies are developed, it becomes increasingly imperative that this broad spectrum of disorders is recognized and diagnosed. This work summarizes key advances in the leukodystrophy field.
Collapse
|
53
|
Yoon Han J, Gon Cho Y, Park J, Jang W. A novel variant of the POLR3A gene in a patient with hypomyelinating POLR3-related leukodystrophy. Clin Chim Acta 2022; 533:15-21. [PMID: 35691411 DOI: 10.1016/j.cca.2022.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/04/2022] [Accepted: 06/05/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Hypomyelinating POLR3-related leukodystrophy is a group of rare neurological diseases characterized by degeneration of the white matter of the brain with different combinations of major clinical findings. Here we report the first Korean POLR3-related leukodystrophy caused by bi-allelic POLR3A c.1771-6C > G and novel c.1650_1661del variants. METHODS An 18-month-old girl was admitted for evaluation of a seizure-like activity with spasticity that affected her entire body. She showed dental abnormalities, but not suspicious facial dysmorphism. She was in a bed-ridden state with severe cognitive impairments and episodes of dystonic posturing for 1-2 min. Trio exome sequencing (ES) was performed to determine the potential genetic cause of severe developmental delay with leukodystrophy in our proband. RESULTS Trio ES revealed that bi-allelic POLR3A deleterious variants, c.1650_1661del of the exon 13, and c.1771-6C > G of the intron 13 were best candidate as causes of hypomyelinating POLR3-related leukodystrophy. Sanger sequencing confirmed the genetic origin of these POLR3A deleterious variants as autosomal recessive hereditary transmission. CONCLUSION Our report provides additional evidence for a phenotypic continuum of hypomyelinating POLR3-related leukodystrophy caused by bi-allelic POLR3A variants. Further genetic studies are required to understand underlying pleiotropic effects of different POLR3A variants.
Collapse
Affiliation(s)
- Ji Yoon Han
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong Gon Cho
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Hospital, Jeonju, Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Joonhong Park
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Hospital, Jeonju, Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea.
| | - Woori Jang
- Department of Laboratory Medicine, Inha University School of Medicine, Incheon, Korea.
| |
Collapse
|
54
|
Costei C, Barbarosie M, Bernard G, Brais B, La Piana R. Adult Hereditary White Matter Diseases With Psychiatric Presentation: Clinical Pointers and MRI Algorithm to Guide the Diagnostic Process. J Neuropsychiatry Clin Neurosci 2022; 33:180-193. [PMID: 33951919 DOI: 10.1176/appi.neuropsych.20110294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The investigators aimed to provide clinical and MRI guidelines for determining when genetic workup should be considered in order to exclude hereditary leukoencephalopathies in affected patients with a psychiatric presentation. METHODS A systematic literature review was conducted, and clinical cases are provided. Given the central role of MRI pattern recognition in the diagnosis of white matter disorders, the investigators adapted an MRI algorithm that guides the interpretation of MRI findings and thus directs further investigations, such as genetic testing. RESULTS Twelve genetic leukoencephalopathies that can present with psychiatric symptoms were identified. As examples of presentations that can occur in clinical practice, five clinical vignettes from patients assessed at a referral center for adult genetic leukoencephalopathies are provided. CONCLUSIONS Features such as drug-resistant symptoms, presence of long-standing somatic features, trigger events, consanguinity, and positive family history should orient the clinician toward diagnostic workup to exclude the presence of a genetic white matter disorder. The identification of MRI white matter abnormalities, especially when presenting a specific pattern of involvement, should prompt genetic testing for known forms of genetic leukoencephalopathies.
Collapse
Affiliation(s)
- Catalina Costei
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal (Costei, Brais, La Piana); Department of Psychiatry, McGill University (Barbarosie); Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University (Bernard); Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal (Bernard); Child Health and Human Development Program, Research Institute of the McGill University Health Center (Bernard); and Department of Diagnostic Radiology, McGill University (La Piana)
| | - Michaela Barbarosie
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal (Costei, Brais, La Piana); Department of Psychiatry, McGill University (Barbarosie); Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University (Bernard); Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal (Bernard); Child Health and Human Development Program, Research Institute of the McGill University Health Center (Bernard); and Department of Diagnostic Radiology, McGill University (La Piana)
| | - Geneviève Bernard
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal (Costei, Brais, La Piana); Department of Psychiatry, McGill University (Barbarosie); Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University (Bernard); Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal (Bernard); Child Health and Human Development Program, Research Institute of the McGill University Health Center (Bernard); and Department of Diagnostic Radiology, McGill University (La Piana)
| | - Bernard Brais
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal (Costei, Brais, La Piana); Department of Psychiatry, McGill University (Barbarosie); Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University (Bernard); Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal (Bernard); Child Health and Human Development Program, Research Institute of the McGill University Health Center (Bernard); and Department of Diagnostic Radiology, McGill University (La Piana)
| | - Roberta La Piana
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal (Costei, Brais, La Piana); Department of Psychiatry, McGill University (Barbarosie); Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University (Bernard); Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal (Bernard); Child Health and Human Development Program, Research Institute of the McGill University Health Center (Bernard); and Department of Diagnostic Radiology, McGill University (La Piana)
| |
Collapse
|
55
|
Yan H, Yang S, Hou Y, Ali S, Escobar A, Gao K, Duan R, Kubisiak T, Wang J, Zhang Y, Xiao J, Jiang Y, Zhang T, Wu Y, Burmeister M, Wang Q, Cuajungco MP, Wang J. Functional Study of TMEM163 Gene Variants Associated with Hypomyelination Leukodystrophy. Cells 2022; 11:cells11081285. [PMID: 35455965 PMCID: PMC9031525 DOI: 10.3390/cells11081285] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/19/2022] [Accepted: 04/06/2022] [Indexed: 02/06/2023] Open
Abstract
Hypomyelinating leukodystrophies (HLDs) are a rare group of heterogeneously genetic disorders characterized by persistent deficit of myelin observed on magnetic resonance imaging (MRI). To identify a new disease-associated gene of HLD, trio-based whole exome sequencing was performed for unexplained patients with HLD. Functional studies were performed to confirm the phenotypic effect of candidate protein variants. Two de novo heterozygous variants, c.227T>G p.(L76R) or c.227T>C p.(L76P) in TMEM163 were identified in two unrelated HLD patients. TMEM163 protein is a zinc efflux transporter localized within the plasma membrane, lysosomes, early endosomes, and other vesicular compartments. It has not been associated with hypomyelination. Functional zinc flux assays in HeLa cells stably-expressing TMEM163 protein variants, L76R and L76P, revealed distinct attenuation or enhancement of zinc efflux, respectively. Experiments using a zebrafish model with knockdown of tmem163a and tmem163b (morphants) showed that loss of tmem163 causes dysplasia of the larvae, locomotor disability and myelin deficit. Expression of human wild type TMEM163 mRNAs in morphants rescues the phenotype, while the TMEM163 L76P and L76R mutants aggravated the condition. Moreover, poor proliferation, elevated apoptosis of oligodendrocytes, and reduced oligodendrocytes and neurons were also observed in zebrafish morphants. Our findings suggest an unappreciated role for TMEM163 protein in myelin development and add TMEM163 to a growing list of genes associated with hypomyelination leukodystrophy.
Collapse
Affiliation(s)
- Huifang Yan
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China; (H.Y.); (K.G.); (R.D.); (J.W.); (Y.Z.); (Y.J.); (Y.W.)
- Joint International Research Center of Translational and Clinical Research, Beijing 100191, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing 100034, China
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (T.K.); (M.B.)
| | - Shuyan Yang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China; (S.Y.); (T.Z.)
| | - Yiming Hou
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China; (Y.H.); (Q.W.)
| | - Saima Ali
- Department of Biological Science, California State University, Fullerton, CA 92831, USA; (S.A.); (A.E.)
| | - Adrian Escobar
- Department of Biological Science, California State University, Fullerton, CA 92831, USA; (S.A.); (A.E.)
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China; (H.Y.); (K.G.); (R.D.); (J.W.); (Y.Z.); (Y.J.); (Y.W.)
| | - Ruoyu Duan
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China; (H.Y.); (K.G.); (R.D.); (J.W.); (Y.Z.); (Y.J.); (Y.W.)
| | - Thomas Kubisiak
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (T.K.); (M.B.)
| | - Junyu Wang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China; (H.Y.); (K.G.); (R.D.); (J.W.); (Y.Z.); (Y.J.); (Y.W.)
| | - Yu Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China; (H.Y.); (K.G.); (R.D.); (J.W.); (Y.Z.); (Y.J.); (Y.W.)
| | - Jiangxi Xiao
- Department of Radiology, Peking University First Hospital, Beijing 100034, China;
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China; (H.Y.); (K.G.); (R.D.); (J.W.); (Y.Z.); (Y.J.); (Y.W.)
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing 100034, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing 100191, China
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China; (S.Y.); (T.Z.)
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China; (H.Y.); (K.G.); (R.D.); (J.W.); (Y.Z.); (Y.J.); (Y.W.)
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing 100034, China
| | - Margit Burmeister
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (T.K.); (M.B.)
- Departments of Computational Medicine & Bioinformatics, Psychiatry and Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China; (Y.H.); (Q.W.)
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Math P. Cuajungco
- Department of Biological Science, California State University, Fullerton, CA 92831, USA; (S.A.); (A.E.)
- Center for Applied Biotechnology Studies, California State University, Fullerton, CA 92831, USA
- Correspondence: (M.P.C.); (J.W.)
| | - Jingmin Wang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China; (H.Y.); (K.G.); (R.D.); (J.W.); (Y.Z.); (Y.J.); (Y.W.)
- Joint International Research Center of Translational and Clinical Research, Beijing 100191, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing 100034, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing 100191, China
- Correspondence: (M.P.C.); (J.W.)
| |
Collapse
|
56
|
Naseer MI, Abdulkareem AA, Pushparaj PN, Saharti S, Muthaffar OY. Next-Generation Sequencing Reveals Novel Homozygous Missense Variant c.934T > C in POLR1C Gene Causing Leukodystrophy and Hypomyelinating Disease. Front Pediatr 2022; 10:862722. [PMID: 35685919 PMCID: PMC9171136 DOI: 10.3389/fped.2022.862722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Leukodystrophies are a diverse group of genetically established disorders categorized by unusual white matter changes on brain imaging. Hypomyelinating leukodystrophies (HLDs) are a group of neurodevelopmental disorders that affect myelin sheath development in the brain. These disorders are categorized as developmental delay, spasticity, hypotonia, and intellectual disabilities. We describe a patient with developmental delay, cerebellar ataxia, spasticity, hypotonia, and intellectual disability from a healthy family member. Whole exome sequencing (WES) was performed to identify causative variants, which were further analyzed by bioinformatic analysis. WES was performed, and Sanger sequencing-based segregation analysis confirmed the presence of the homozygous missense variants of NM_203290.3 c.934T > C p.Ser312Pro of RNA polymerase I and III subunit C (POLR1C) gene in this patient and heterozygous variant in the unaffected carrier father and mother, supporting the pathogenicity and inheritance pattern of this variant. Furthermore, the variant identified by WES was validated in healthy controls (n = 100) using Sanger sequencing analysis. Finally, our study explained the important use of WES in disease diagnosis and provided further evidence that the variant in the POLR1C gene may play an important role in the development of hypomyelinating leukodystrophy in Saudi families.
Collapse
Affiliation(s)
- Muhammad Imran Naseer
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Angham Abdulrahman Abdulkareem
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Pharmacology, Center for Transdisciplinary Research, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Samah Saharti
- Department of Pathology and Microbiology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Osama Y Muthaffar
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
57
|
Wang C, Wang S, Xie H, Yang S. Clinical and imaging characteristics of 4H syndrome: A case report. CNS Neurosci Ther 2021; 28:458-460. [PMID: 34953043 PMCID: PMC8841286 DOI: 10.1111/cns.13790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/02/2021] [Accepted: 12/12/2021] [Indexed: 11/29/2022] Open
Affiliation(s)
- Chao Wang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, NO.88 Jiefang Road, Hangzhou, China
| | - Shan Wang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, NO.88 Jiefang Road, Hangzhou, China
| | - Huanhuan Xie
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, NO.88 Jiefang Road, Hangzhou, China
| | - Siyu Yang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, NO.88 Jiefang Road, Hangzhou, China
| |
Collapse
|
58
|
Muirhead KJ, Clause AR, Schlachetzki Z, Dubbs H, Perry DL, Hagelstrom RT, Taft RJ, Vanderver A. Genome sequencing identifies three molecular diagnoses including a mosaic variant in the COL2A1 gene in an individual with Pol III-related leukodystrophy and Feingold syndrome. Cold Spring Harb Mol Case Stud 2021; 7:a006143. [PMID: 34737199 PMCID: PMC8751417 DOI: 10.1101/mcs.a006143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Undiagnosed genetic disease imposes a significant burden on families and health-care resources, especially in cases with a complex phenotype. Here we present a child with suspected leukodystrophy in the context of additional features, including hearing loss, clinodactyly, rotated thumbs, tapered fingers, and simplified palmar crease. Trio genome sequencing (GS) identified three molecular diagnoses in this individual: compound heterozygous missense variants associated with polymerase III (Pol III)-related leukodystrophy, a 4-Mb de novo copy-number loss including the MYCN gene associated with Feingold syndrome, and a mosaic single-nucleotide variant associated with COL2A1-related disorders. These variants fully account for the individual's features, but also illustrate the potential for superimposed and unclear contributions of multiple diagnoses to an individual's overall presentation. This report demonstrates the advantage of GS in detection of multiple variant types, including low-level mosaic variants, and emphasizes the need for comprehensive genetic analysis and detailed clinical phenotyping to provide individuals and their families with the maximum benefit for clinical care and genetic counseling.
Collapse
Affiliation(s)
- Kayla J Muirhead
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Amanda R Clause
- Illumina Clinical Services Laboratory, Illumina Inc., San Diego, California 92122, USA
| | - Zinayida Schlachetzki
- Illumina Clinical Services Laboratory, Illumina Inc., San Diego, California 92122, USA
| | - Holly Dubbs
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Denise L Perry
- Illumina Clinical Services Laboratory, Illumina Inc., San Diego, California 92122, USA
| | - R Tanner Hagelstrom
- Illumina Clinical Services Laboratory, Illumina Inc., San Diego, California 92122, USA
| | - Ryan J Taft
- Illumina Clinical Services Laboratory, Illumina Inc., San Diego, California 92122, USA
| | - Adeline Vanderver
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
59
|
Wilson D, Hallett M, Anderson T. An Eye on Movement Disorders. Mov Disord Clin Pract 2021; 8:1168-1180. [PMID: 34765682 DOI: 10.1002/mdc3.13317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/28/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
Eye disorders spanning a range of ocular tissue are common in patients with movement disorders. Highlighting these ocular manifestations will benefit patients and may even aid in diagnosis. In this educational review we outline the anatomy and function of the ocular tissues with a focus on the tissues most affected in movement disorders. We review the movement disorders associated with ocular pathology and where possible explore the underlying cellular basis thought to be driving the pathology and provide a brief overview of ophthalmic investigations available to the neurologist. This review does not cover intracranial primary visual pathways, higher visual function, or the ocular motor system.
Collapse
Affiliation(s)
- Duncan Wilson
- Department of Neurology Christchurch Hospital Christchurch New Zealand.,New Zealand Brain Research Institute Christchurch New Zealand
| | - Mark Hallett
- Human Motor Control Section, NINDS, NIH Bethesda Maryland USA
| | - Tim Anderson
- Department of Neurology Christchurch Hospital Christchurch New Zealand.,New Zealand Brain Research Institute Christchurch New Zealand.,Department of Medicine Otago University Dunedin New Zealand
| |
Collapse
|
60
|
Sytsma TM, Chen DH, Rolf B, Dorschner M, Jayadev S, Keene CD, Zhang J, Bird TD, Latimer CS. Spinal cord-predominant neuropathology in an adult-onset case of POLR3A-related spastic ataxia. Neuropathology 2021; 42:58-65. [PMID: 34753215 PMCID: PMC8810698 DOI: 10.1111/neup.12775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 12/30/2022]
Abstract
Biallelic mutations in POLR3A have been associated with childhood‐onset hypomyelinating leukodystrophies and adolescent‐to‐adult‐onset spastic ataxia, the latter of which has been linked to the intronic variant c.1909 + 22G>A. We report a case of adult‐onset spastic ataxia in a 75‐year‐old man, being a compound heterozygous carrier of this variant, whose brain and spinal cord were for the first time investigated by neuropathological examination. We describe prominent degeneration of the posterior columns, spinocerebellar tracts, and anterior corticospinal tracts of the spinal cord in a pattern resembling Friedreich's ataxia, with a notable lack of significant white matter pathology throughout the brain, in marked contrast with childhood‐onset cases. Immunohistochemical examination for the POLR3A protein demonstrated no apparent differences in localization or staining intensity between the proband and an age‐matched control subject. We demonstrate the clinicopathologic description of POLR3A‐related neurodegenerative disease and also mention the differential diagnosis of the childhood‐onset hypomyelinating leukodystrophy and late‐onset spastic ataxia phenotypes.
Collapse
Affiliation(s)
- Trevor M Sytsma
- Neuropathology Division, Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Dong-Hui Chen
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Bradley Rolf
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Michael Dorschner
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Suman Jayadev
- Neuropathology Division, Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA.,Department of Neurology, University of Washington School of Medicine, Seattle, Washington, USA.,Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - C Dirk Keene
- Neuropathology Division, Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jing Zhang
- Neuropathology Division, Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Thomas D Bird
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington, USA.,Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA.,Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
| | - Caitlin S Latimer
- Neuropathology Division, Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
61
|
Xue YY, Cheng HL, Dong HL, Yin HM, Yuan Y, Meng LC, Wu ZY, Yu H. A de novo variant of POLR3B causes demyelinating Charcot-Marie-Tooth disease in a Chinese patient: a case report. BMC Neurol 2021; 21:402. [PMID: 34666706 PMCID: PMC8527611 DOI: 10.1186/s12883-021-02399-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Charcot-Marie-Tooth (CMT) disease is a group of inherited peripheral neuropathies, which are subdivided into demyelinating and axonal forms. Biallelic mutations in POLR3B are the well-established cause of hypomyelinating leukodystrophy, which is characterized by hypomyelination, hypodontia, and hypogonadotropic hypogonadism. To date, only one study has reported the demyelinating peripheral neuropathy phenotype caused by heterozygous POLR3B variants. CASE PRESENTATION A 19-year-old male patient was referred to our hospital for progressive muscle weakness of the lower extremities. Physical examination showed muscle atrophy, sensory loss and deformities of the extremities. Nerve conduction studies and electromyography tests revealed sensorimotor demyelinating polyneuropathy with secondary axonal loss. Trio whole-exome sequencing revealed a de novo variant in POLR3B (c.3137G > A). CONCLUSIONS In this study, we report the case of a Chinese patient with a de novo variant in POLR3B (c.3137G > A), who manifested demyelinating CMT phenotype without additional neurological or extra-neurological involvement. This work is the second report on POLR3B-related CMT.
Collapse
Affiliation(s)
- Yan-Yan Xue
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, China
| | - Hao-Ling Cheng
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, China.,Department of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hai-Lin Dong
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, China
| | - Hou-Min Yin
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Ling-Chao Meng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, China.
| | - Hao Yu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, China.
| |
Collapse
|
62
|
Gonzalez-Latapi P, Sousa M, Lang AE. Movement Disorders Associated with Hypogonadism. Mov Disord Clin Pract 2021; 8:997-1011. [PMID: 34631935 DOI: 10.1002/mdc3.13308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/25/2021] [Accepted: 07/03/2021] [Indexed: 11/10/2022] Open
Abstract
A variety of movement disorders can be associated with hypogonadism. Identification of this association may aid in guiding workup and reaching an accurate diagnosis. We conducted a comprehensive and structured search to identify the most common movement disorders associated with hypogonadism. Only Case Reports and Case Series articles were included. Ataxia was the most common movement disorder associated with hypogonadism, including entities such as Gordon-Holmes syndrome, Boucher-Neuhäuser, Marinesco-Sjögren and Perrault syndrome. Tremor was also commonly described, particularly with aneuploidies such as Klinefelter syndrome and Jacob's syndrome. Other rare conditions including mitochondrial disorders and Woodhouse-Sakati syndrome are associated with dystonia and parkinsonism and either hypo or hypergonadotropic hypogonadism. We also highlight those entities where a combination of movement disorders is present. Hypogonadism may be more commonly associated with movement disorders than previously appreciated. It is important for the clinician to be aware of this association, as well as accompanying symptoms in order to reach a precise diagnosis.
Collapse
Affiliation(s)
- Paulina Gonzalez-Latapi
- The Edmond J. Safra Program for Parkinson Disease, Movement Disorder Clinic Toronto Western Hospital, University Health Network Toronto Ontario Canada
| | - Mario Sousa
- The Edmond J. Safra Program for Parkinson Disease, Movement Disorder Clinic Toronto Western Hospital, University Health Network Toronto Ontario Canada
| | - Anthony E Lang
- The Edmond J. Safra Program for Parkinson Disease, Movement Disorder Clinic Toronto Western Hospital, University Health Network Toronto Ontario Canada.,Division of Neurology, Department of Medicine University of Toronto Toronto Ontario Canada
| |
Collapse
|
63
|
Mishra S, Hasan SH, Sakhawala RM, Chaudhry S, Maraia RJ. Mechanism of RNA polymerase III termination-associated reinitiation-recycling conferred by the essential function of the N terminal-and-linker domain of the C11 subunit. Nat Commun 2021; 12:5900. [PMID: 34625550 PMCID: PMC8501072 DOI: 10.1038/s41467-021-26080-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/17/2021] [Indexed: 01/06/2023] Open
Abstract
RNA polymerase III achieves high level tRNA synthesis by termination-associated reinitiation-recycling that involves the essential C11 subunit and heterodimeric C37/53. The C11-CTD (C-terminal domain) promotes Pol III active center-intrinsic RNA 3'-cleavage although deciphering function for this activity has been complicated. We show that the isolated NTD (N-terminal domain) of C11 stimulates Pol III termination by C37/53 but not reinitiation-recycling which requires the NTD-linker (NTD-L). By an approach different from what led to current belief that RNA 3'-cleavage activity is essential, we show that NTD-L can provide the essential function of Saccharomyces cerevisiae C11 whereas classic point mutations that block cleavage, interfere with active site function and are toxic to growth. Biochemical and in vivo analysis including of the C11 invariant central linker led to a model for Pol III termination-associated reinitiation-recycling. The C11 NTD and CTD stimulate termination and RNA 3'-cleavage, respectively, whereas reinitiation-recycling activity unique to Pol III requires only the NTD-linker. RNA 3'-cleavage activity increases growth rate but is nonessential.
Collapse
Affiliation(s)
- Saurabh Mishra
- Intramural Research Program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- Department of Biochemistry, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Shaina H Hasan
- Intramural Research Program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- Mayo Clinic Alix School of Medicine, Scottsdale, AZ, USA
| | - Rima M Sakhawala
- Intramural Research Program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- Section on Regulatory RNA, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Shereen Chaudhry
- Intramural Research Program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
- Pfizer (Pearl River Site), 401 N Middletown Rd, Pearl River, NY, USA
| | - Richard J Maraia
- Intramural Research Program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
64
|
Lessel D, Rading K, Campbell SE, Thiele H, Altmüller J, Gordon LB, Kubisch C. A novel homozygous synonymous variant further expands the phenotypic spectrum of POLR3A-related pathologies. Am J Med Genet A 2021; 188:216-223. [PMID: 34611991 DOI: 10.1002/ajmg.a.62525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/21/2021] [Accepted: 09/06/2021] [Indexed: 12/18/2022]
Abstract
Pathogenic biallelic variants in POL3RA have been associated with different disorders characterized by progressive neurological deterioration. These include the 4H leukodystrophy syndrome (hypomyelination, hypogonadotropic hypogonadism, and hypodontia) and adolescent-onset progressive spastic ataxia, as well as Wiedemann-Rautenstrauch syndrome (WRS), a recognizable neonatal progeroid syndrome. The phenotypic differences between these disorders are thought to occur mainly due to different functional effects of underlying POLR3A variants. Here we present the detailed clinical course of a 37-year-old woman in whom we identified a homozygous synonymous POLR3A variant c.3336G>A resulting in leaky splicing r.[3336ins192, =, 3243_3336del94]. She presented at birth with intrauterine growth retardation, lipodystrophy, muscular hypotonia, and several WRS-like facial features, albeit without sparse hair and prominent scalp veins. She had no signs of developmental delay or intellectual disability. Over the years, above characteristic facial features, she showed severe postnatal growth retardation, global lipodystrophy, joint contractures, thoracic hypoplasia, scoliosis, anodontia, spastic quadriplegia, bilateral hearing loss, aphonia, hypogonadotropic hypogonadism, and cerebellar peduncles hyperintensities in brain imaging. These manifestations partially overlap the clinical features of the previously reported POLR3A-associated disorders, mostly mimicking the WRS. Thus, our study expands the POLR3A-mediated phenotypic spectrum and suggests existence of a phenotypic continuum underlying biallelic POLR3A variants.
Collapse
Affiliation(s)
- Davor Lessel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Rading
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susan E Campbell
- Center for Gerontology and Healthcare Research, Brown University, Providence, Rhode Island, USA
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, Cologne, Germany.,Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Core Facility Genomics, Berlin, Germany.,The Genomics unit, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Leslie B Gordon
- Department of Pediatrics, Division of Genetics, Hasbro Children's Hospital, Providence, Rhode Island, USA.,Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA.,Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian Kubisch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
65
|
Defective myelination in an RNA polymerase III mutant leukodystrophic mouse. Proc Natl Acad Sci U S A 2021; 118:2024378118. [PMID: 34583988 DOI: 10.1073/pnas.2024378118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2021] [Indexed: 01/06/2023] Open
Abstract
RNA polymerase (Pol) III synthesizes abundant short noncoding RNAs that have essential functions in protein synthesis, secretion, and other processes. Despite the ubiquitous functions of these RNAs, mutations in Pol III subunits cause Pol III-related leukodystrophy, an early-onset neurodegenerative disease. The basis of this neural sensitivity and the mechanisms of disease pathogenesis are unknown. Here we show that mice expressing pathogenic mutations in the largest Pol III subunit, Polr3a, specifically in Olig2-expressing cells, have impaired growth and developmental delay, deficits in cognitive, sensory, and fine sensorimotor function, and hypomyelination in multiple regions of the cerebrum and spinal cord. These phenotypes reflect a subset of clinical features seen in patients. In contrast, the gross motor defects and cerebellar hypomyelination that are common features of severely affected patients are absent in the mice, suggesting a relatively mild form of the disease in this conditional model. Our results show that disease pathogenesis in the mice involves defects that reduce both the number of mature myelinating oligodendrocytes and the ability of these cells to produce a myelin sheath of normal thickness. The findings suggest unique sensitivities of oligodendrogenesis and myelination to perturbations of Pol III transcription.
Collapse
|
66
|
Lata E, Choquet K, Sagliocco F, Brais B, Bernard G, Teichmann M. RNA Polymerase III Subunit Mutations in Genetic Diseases. Front Mol Biosci 2021; 8:696438. [PMID: 34395528 PMCID: PMC8362101 DOI: 10.3389/fmolb.2021.696438] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/21/2021] [Indexed: 12/24/2022] Open
Abstract
RNA polymerase (Pol) III transcribes small untranslated RNAs such as 5S ribosomal RNA, transfer RNAs, and U6 small nuclear RNA. Because of the functions of these RNAs, Pol III transcription is best known for its essential contribution to RNA maturation and translation. Surprisingly, it was discovered in the last decade that various inherited mutations in genes encoding nine distinct subunits of Pol III cause tissue-specific diseases rather than a general failure of all vital functions. Mutations in the POLR3A, POLR3C, POLR3E and POLR3F subunits are associated with susceptibility to varicella zoster virus-induced encephalitis and pneumonitis. In addition, an ever-increasing number of distinct mutations in the POLR3A, POLR3B, POLR1C and POLR3K subunits cause a spectrum of neurodegenerative diseases, which includes most notably hypomyelinating leukodystrophy. Furthermore, other rare diseases are also associated with mutations in genes encoding subunits of Pol III (POLR3H, POLR3GL) and the BRF1 component of the TFIIIB transcription initiation factor. Although the causal relationship between these mutations and disease development is widely accepted, the exact molecular mechanisms underlying disease pathogenesis remain enigmatic. Here, we review the current knowledge on the functional impact of specific mutations, possible Pol III-related disease-causing mechanisms, and animal models that may help to better understand the links between Pol III mutations and disease.
Collapse
Affiliation(s)
- Elisabeth Lata
- Bordeaux University, Inserm U 1212, CNRS UMR 5320, ARNA laboratory, Bordeaux, France
| | - Karine Choquet
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - Francis Sagliocco
- Bordeaux University, Inserm U 1212, CNRS UMR 5320, ARNA laboratory, Bordeaux, France
| | - Bernard Brais
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Geneviève Bernard
- Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University, Montreal, QC, Canada
- Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Martin Teichmann
- Bordeaux University, Inserm U 1212, CNRS UMR 5320, ARNA laboratory, Bordeaux, France
| |
Collapse
|
67
|
Di Donato I, Gallo A, Ricca I, Fini N, Silvestri G, Gurrieri F, Cirillo M, Cerase A, Natale G, Matrone F, Riso V, Melone MAB, Tessa A, De Michele G, Federico A, Filla A, Dotti MT, Santorelli FM. POLR3A variants in hereditary spastic paraparesis and ataxia: clinical, genetic, and neuroradiological findings in a cohort of Italian patients. Neurol Sci 2021; 43:1071-1077. [PMID: 34296356 PMCID: PMC8789690 DOI: 10.1007/s10072-021-05462-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/03/2021] [Indexed: 11/01/2022]
Abstract
Mutations in POLR3A are characterized by high phenotypic heterogeneity, with manifestations ranging from severe childhood-onset hypomyelinating leukodystrophic syndromes to milder and later-onset gait disorders with central hypomyelination, with or without additional non-neurological signs. Recently, a milder phenotype consisting of late-onset spastic ataxia without hypomyelinating leukodystrophy has been suggested to be specific to the intronic c.1909 + 22G > A mutation in POLR3A. Here, we present 10 patients from 8 unrelated families with POLR3A-related late-onset spastic ataxia, all harboring the c.1909 + 22G > A variant. Most of them showed an ataxic-spastic picture, two a "pure" cerebellar phenotype, and one a "pure" spastic presentation. The non-neurological findings typically associated with POLR3A mutations were absent in all the patients. The main findings on brain MRI were bilateral hyperintensity along the superior cerebellar peduncles on FLAIR sequences, observed in most of the patients, and cerebellar and/or spinal cord atrophy, found in half of the patients. Only one patient exhibited central hypomyelination. The POLR3A mutations present in this cohort were the c.1909 + 22G > A splice site variant found in compound heterozygosity with six additional variants (three missense, two nonsense, one splice) and, in one patient, with a novel large deletion involving exons 14-18. Interestingly, this patient had the most "complex" presentation among those observed in our cohort; it included some neurological and non-neurological features, such as seizures, neurosensory deafness, and lipomas, that have not previously been reported in association with late-onset POLR3A-related disorders, and therefore further expand the phenotype.
Collapse
Affiliation(s)
- Ilaria Di Donato
- Department of Medicine, Surgery and Neurosciences, University of Siena, viale Bracci, 16, 50055, Siena, Italy.
| | - Antonio Gallo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Ivana Ricca
- Molecular Medicine and Neurogenetics, IRCCS Fondazione Stella Maris, via dei Giacinti 2, Calambrone, 56128, Pisa, Italy
| | - Nicola Fini
- Neurology Unit, Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Gabriella Silvestri
- Fondazione Policlinico Universitario 'A. Gemelli' IRCCS, UOC Neurologia, Rome, Italy
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Mario Cirillo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alfonso Cerase
- Department of Medicine, Surgery and Neurosciences, University of Siena, viale Bracci, 16, 50055, Siena, Italy
| | - Gemma Natale
- Molecular Medicine and Neurogenetics, IRCCS Fondazione Stella Maris, via dei Giacinti 2, Calambrone, 56128, Pisa, Italy
| | - Federica Matrone
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vittorio Riso
- Fondazione Policlinico Universitario 'A. Gemelli' IRCCS, UOC Neurologia, Rome, Italy
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mariarosa Anna Beatrice Melone
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA, USA
| | - Alessandra Tessa
- Molecular Medicine and Neurogenetics, IRCCS Fondazione Stella Maris, via dei Giacinti 2, Calambrone, 56128, Pisa, Italy
| | - Giovanna De Michele
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, Naples, Italy
| | - Antonio Federico
- Department of Medicine, Surgery and Neurosciences, University of Siena, viale Bracci, 16, 50055, Siena, Italy
| | - Alessandro Filla
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, Naples, Italy
| | - Maria Teresa Dotti
- Department of Medicine, Surgery and Neurosciences, University of Siena, viale Bracci, 16, 50055, Siena, Italy
| | - Filippo Maria Santorelli
- Molecular Medicine and Neurogenetics, IRCCS Fondazione Stella Maris, via dei Giacinti 2, Calambrone, 56128, Pisa, Italy.
| |
Collapse
|
68
|
Wu SW, Li L, Feng F, Wang L, Kong YY, Liu XW, Yin C. Whole-exome sequencing reveals POLR3B variants associated with progeria-related Wiedemann-Rautenstrauch syndrome. Ital J Pediatr 2021; 47:160. [PMID: 34289880 PMCID: PMC8296688 DOI: 10.1186/s13052-021-01112-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/04/2021] [Indexed: 11/10/2022] Open
Abstract
Background Wiedemann-Rautenstrauch syndrome (WRS) is a rare autosomal recessive neonatal progeroid disorder characterized by prenatal and postnatal growth retardation, short stature, a progeroid appearance, hypotonia, and mental impairment. Case presentation A 6-year-old patient, who initially presented with multiple postnatal abnormalities, facial dysplasia, micrognathia, skull appearance, hallux valgus, and congenital dislocation of the hip, was recruited in this study. The patient was initially diagnosed with progeria. The mother of the patient had abnormal fetal development during her second pregnancy check-up, and the clinical phenotype of the fetus was similar to that of the patient. Whole-exome sequencing (WES) of the patient was performed, and POLR3B compound heterozygous variants—c.2191G > C:p.E731Q and c.3046G > A:p.V1016M—were identified in the patient. Using Sanger sequencing, we found that the phenotypes and genotypes were segregated within the pedigree. These two variants are novel and not found in the gnomAD and 1000 Genomes databases. The two mutation sites are highly conserved between humans and zebrafish. Conclusions Our study not only identified a novel WRS-associated gene, POLR3B, but also broadened the mutational and phenotypic spectra of POLR3B. Furthermore, WES may be useful for identifying rare disease-related genetic variants. Supplementary Information The online version contains supplementary material available at 10.1186/s13052-021-01112-6.
Collapse
Affiliation(s)
- Shao-Wen Wu
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, Chaoyang, China.,Beijing Maternal and Child Health Care Hospital, Beijing, 100026, Chaoyang, China
| | - Lin Li
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, Chaoyang, China.,Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, Chaoyang, China
| | - Fan Feng
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Haidian, Beijing, 100084, China
| | - Li Wang
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, Chaoyang, China.,Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, Chaoyang, China
| | - Yuan-Yuan Kong
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, Chaoyang, China.,Department of Newborn Screening, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, Chaoyang, China
| | - Xiao-Wei Liu
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, Chaoyang, China. .,Beijing Maternal and Child Health Care Hospital, Beijing, 100026, Chaoyang, China.
| | - Chenghong Yin
- Beijing Maternal and Child Health Care Hospital, Beijing, 100026, Chaoyang, China. .,Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, Chaoyang, China.
| |
Collapse
|
69
|
Dinov D, Vorona G, Harper A. Child Neurology: Hypotonia and Delayed Teeth Eruption in a 2-Year-Old Female. Neurology 2021; 97:875-878. [PMID: 34187860 DOI: 10.1212/wnl.0000000000012445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
POLR3- related disorders are rare hypo-myelinating leukodystrophies associated with hypodontia. We present a female patient, who was referred to pediatric neurology at 2 years of age for tremor, low tone, and motor delays. Additionally, she was noted to have a delay in her teeth eruption and myopia. Neurological examination was significant for ataxic features and global developmental delay. Laboratory workup was unrevealing. MRI imaging was significant for hypomyelination. Genetic testing confirmed a pathogenic variant of POLR3B. POLR3- related leukodystrophies should be considered in patients who present with hypotonia, ataxia and hypodontia. There are many different subtypes of POLR-related leukodystrophies each with distinguishing phenotypic and radiographic features. Although, MRI can be helpful in initial evaluation genetic testing is needed for confirmatory diagnosis and to guide prognosis.
Collapse
Affiliation(s)
- Darina Dinov
- Children's Hospital of Richmond, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Gregory Vorona
- Department of Radiology, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Amy Harper
- Children's Hospital of Richmond, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| |
Collapse
|
70
|
von Jonquieres G, Rae CD, Housley GD. Emerging Concepts in Vector Development for Glial Gene Therapy: Implications for Leukodystrophies. Front Cell Neurosci 2021; 15:661857. [PMID: 34239416 PMCID: PMC8258421 DOI: 10.3389/fncel.2021.661857] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Central Nervous System (CNS) homeostasis and function rely on intercellular synchronization of metabolic pathways. Developmental and neurochemical imbalances arising from mutations are frequently associated with devastating and often intractable neurological dysfunction. In the absence of pharmacological treatment options, but with knowledge of the genetic cause underlying the pathophysiology, gene therapy holds promise for disease control. Consideration of leukodystrophies provide a case in point; we review cell type – specific expression pattern of the disease – causing genes and reflect on genetic and cellular treatment approaches including ex vivo hematopoietic stem cell gene therapies and in vivo approaches using adeno-associated virus (AAV) vectors. We link recent advances in vectorology to glial targeting directed towards gene therapies for specific leukodystrophies and related developmental or neurometabolic disorders affecting the CNS white matter and frame strategies for therapy development in future.
Collapse
Affiliation(s)
- Georg von Jonquieres
- Translational Neuroscience Facility, Department of Physiology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Caroline D Rae
- Neuroscience Research Australia, Randwick, NSW, Australia
| | - Gary D Housley
- Translational Neuroscience Facility, Department of Physiology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
71
|
Coulombe B, Derksen A, La Piana R, Brais B, Gauthier MS, Bernard G. POLR3-related leukodystrophy: How do mutations affecting RNA polymerase III subunits cause hypomyelination? Fac Rev 2021; 10:12. [PMID: 33659930 PMCID: PMC7894263 DOI: 10.12703/r/10-12] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Hypomyelinating leukodystrophies are a group of genetic disorders characterized by insufficient myelin deposition during development. A subset of hypomyelinating leukodystrophies, named RNA polymerase III (Pol III or POLR3)-related leukodystrophy or 4H (Hypomyelination, Hypodontia and Hypogonadotropic Hypogonadism) leukodystrophy, was found to be caused by biallelic variants in genes encoding subunits of the enzyme Pol III, including POLR3A, POLR3B, POLR3K, and POLR1C. Pol III is one of the three nuclear RNA polymerases that synthesizes small non-coding RNAs, such as tRNAs, 5S RNA, and others, that are involved in the regulation of essential cellular processes, including transcription, translation and RNA maturation. Affinity purification coupled with mass spectrometry (AP-MS) revealed that a number of mutations causing POLR3-related leukodystrophy impair normal assembly or biogenesis of Pol III, often causing a retention of the unassembled subunits in the cytoplasm. Even though these proteomic studies have helped to understand the molecular defects associated with leukodystrophy, how these mutations cause hypomyelination has yet to be defined. In this review we propose two main hypotheses to explain how mutations affecting Pol III subunits can cause hypomyelination.
Collapse
Affiliation(s)
- Benoit Coulombe
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Alexa Derksen
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Roberta La Piana
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
- Department of Diagnostic Radiology, McGill University, Montréal, QC, Canada
| | - Bernard Brais
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Marie-Soleil Gauthier
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC, Canada
| | - Geneviève Bernard
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Department of Pediatrics, McGill University, Montréal, QC, Canada
- Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montréal, QC, Canada
| |
Collapse
|
72
|
Kyle K, Mason X, Bordelon Y, Pouratian N, Bronstein J. Adult onset POLR3A leukodystrophy presenting with parkinsonism treated with pallidal deep brain stimulation. Parkinsonism Relat Disord 2021; 85:23-25. [PMID: 33652360 DOI: 10.1016/j.parkreldis.2021.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/10/2021] [Accepted: 02/14/2021] [Indexed: 11/17/2022]
Affiliation(s)
- Kevin Kyle
- Department of Neurology, Massachusetts General Hospital, United States.
| | - Xenos Mason
- Department of Neurology, UCLA David Geffen School of Medicine, United States
| | - Yvette Bordelon
- Department of Neurology, UCLA David Geffen School of Medicine, United States
| | - Nader Pouratian
- Department of Neurosurgery, UCLA David Geffen School of Medicine, United States
| | - Jeff Bronstein
- Department of Neurology, UCLA David Geffen School of Medicine, United States
| |
Collapse
|
73
|
Furukawa S, Kunii M, Doi H, Kondo N, Ogura A, Hirabuki K, Itoh T, Matsumoto N, Tanaka F, Katsuno M, Ito Y. Case Report: Severe Osteoporosis and Preventive Therapy in RNA Polymerase III-Related Leukodystrophy. Front Neurol 2021; 12:622355. [PMID: 33716926 PMCID: PMC7952608 DOI: 10.3389/fneur.2021.622355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/25/2021] [Indexed: 11/18/2022] Open
Abstract
RNA polymerase III (POLR3)-related leukodystrophy is an autosomal recessive form of leukodystrophy caused by homozygous or compound heterozygous mutations of the RNA polymerase III subunit genes, including subunit A (POLR3A). With respect to the manifestation triad, hypomyelination, hypodontia, and hypogonadotropic hypogonadism, it is also known as 4H leukodystrophy. Here, we report a 41-year-old woman of POLR3-related leukodystrophy by carrying compound heterozygous pathogenic variants of c.2554A>G (p.M852V) and c.2668G>T (p.V890F) in the POLR3A gene. She was amenorrheic and became a wheelchair user from the age of 15 years and suffered from multiple episodes of pathologic fractures, starting with a subtrochanteric fracture of the right femur after a tonic seizure at age 30 years. Head magnetic resonance imaging demonstrated hypomyelination and atrophies of the cerebellum, brainstem, and corpus callosum. Laboratory examination revealed a marked decrease of gonadotropins and estrogen, low bone density, and high bone resorption markers. Administration of anti-receptor activator of nuclear factor kappa-B ligand monoclonal antibody restored bone resorption markers to a normal level and prevented further pathological bone fractures. Our case emphasizes that osteoporosis should be recognized as a potential but serious complication in POLR3-related leukodystrophy. It may be feasible to prevent pathologic fractures by intensive osteoporosis therapy after endocrinological examinations and evaluation of bone metabolism.
Collapse
Affiliation(s)
- Soma Furukawa
- Department of Neurology, Toyota Memorial Hospital, Toyota, Japan.,Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Misako Kunii
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroshi Doi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naohide Kondo
- Department of Neurology, Toyota Memorial Hospital, Toyota, Japan.,Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Wellness Promotion Center, Corporate Human Resource, Fuji Xerox Co., Ltd, Ebina, Japan
| | - Aya Ogura
- Department of Neurology, Toyota Memorial Hospital, Toyota, Japan.,Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Takayuki Itoh
- Faculty of Psychological and Physical Science, Aichi-Gakuin University, Nissin, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Ito
- Department of Neurology, Toyota Memorial Hospital, Toyota, Japan
| |
Collapse
|
74
|
Fellner A, Lossos A, Kogan E, Argov Z, Gonzaga-Jauregui C, Shuldiner AR, Darawshe M, Bazak L, Lidzbarsky G, Shomron N, Basel-Salmon L, Goldberg Y. Two intronic cis-acting variants in both alleles of the POLR3A gene cause progressive spastic ataxia with hypodontia. Clin Genet 2021; 99:713-718. [PMID: 33491183 DOI: 10.1111/cge.13929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/02/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022]
Abstract
POLR3A encodes the largest subunit of the DNA-dependent RNA polymerase III. Pathogenic variants in this gene are associated with dysregulation of tRNA production and other non-coding RNAs. POLR3A-related disorders include variable phenotypes. The genotype-phenotype correlation is still unclear. Phenotypic analysis and exome sequencing were performed in four affected siblings diagnosed clinically with hereditary spastic ataxia, two healthy siblings and their unaffected mother. All four affected siblings (ages 46-55) had similar clinical features of early childhood-onset hypodontia and adolescent-onset progressive spastic ataxia. None had progeria, gonadal dysfunction or dysmorphism. All affected individuals had biallelic POLR3A pathogenic variants composed by two cis-acting intronic splicing-altering variants, c.1909 + 22G > A and c.3337-11 T > C. The two healthy siblings had wild-type alleles. The mother and another unaffected sibling were heterozygous for the allele containing both variants. This is the first report addressing the clinical consequence associated with homozygosity for a unique pathogenic intronic allele in the POLR3A gene. This allele was previously reported in compound heterozygous combinations in patients with Wiedemann-Rautenstrauch syndrome, a severe progeroid POLR3A-associated phenotype. We show that homozygosity for this allele is associated with spastic ataxia with hypodontia, and not with progeroid features. These findings contribute to the characterization of genotype-phenotype correlation in POLR3A-related disorders.
Collapse
Affiliation(s)
- Avi Fellner
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel.,Department of Neurology, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
| | - Alexander Lossos
- Department of Neurology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Elena Kogan
- Department of Neurology, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
| | - Zohar Argov
- Department of Neurology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | - Malak Darawshe
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Lily Bazak
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
| | - Gabriel Lidzbarsky
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel
| | - Noam Shomron
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Lina Basel-Salmon
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Felsenstein Medical Research Center, Petah Tikva, Israel
| | - Yael Goldberg
- Raphael Recanati Genetics Institute, Rabin Medical Center, Beilinson Campus, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
75
|
Majethia P, Girisha KM. Wiedemann-Rautenstrauch syndrome in an Indian patient with biallelic pathogenic variants in POLR3A. Am J Med Genet A 2021; 185:1602-1605. [PMID: 33559318 DOI: 10.1002/ajmg.a.62115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/02/2021] [Accepted: 01/12/2021] [Indexed: 11/11/2022]
Abstract
Wiedemann-Rautenstrauch syndrome (WRS; MIM# 264090) is a rare neonatal progeroid disorder resulting from biallelic pathogenic variants in the POLR3A. It is an autosomal recessive condition characterized by growth retardation, lipoatrophy, a distinctive face, sparse scalp hair, and dental anomalies. Till date, 19 families are reported with WRS due to variants in POLR3A. Here, we describe an 18 months old male child with biallelic c.2005C>T p.(Arg669Ter) and c.1771-7C>G variant in heterozygous state identified by exome sequencing in POLR3A leading to WRS phenotype. The variant c.1771-7C>G was earlier found to be associated with hereditary spastic ataxia. We emphasize on the phenotype in an Indian patient with WRS.
Collapse
Affiliation(s)
- Purvi Majethia
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Katta Mohan Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
76
|
Cryo-EM structures of human RNA polymerase III in its unbound and transcribing states. Nat Struct Mol Biol 2021; 28:210-219. [PMID: 33558764 PMCID: PMC7610652 DOI: 10.1038/s41594-020-00555-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 12/21/2020] [Indexed: 01/30/2023]
Abstract
RNA polymerase III (Pol III) synthesizes transfer RNAs and other short, essential RNAs. Human Pol III misregulation is linked to tumor transformation, neurodegenerative and developmental disorders, and increased sensitivity to viral infections. Here, we present cryo-electron microscopy structures at 2.8 to 3.3 Å resolution of transcribing and unbound human Pol III. We observe insertion of the TFIIS-like subunit RPC10 into the polymerase funnel, providing insights into how RPC10 triggers transcription termination. Our structures resolve elements absent from Saccharomyces cerevisiae Pol III such as the winged-helix domains of RPC5 and an iron-sulfur cluster, which tethers the heterotrimer subcomplex to the core. The cancer-associated RPC7α isoform binds the polymerase clamp, potentially interfering with Pol III inhibition by tumor suppressor MAF1, which may explain why overexpressed RPC7α enhances tumor transformation. Finally, the human Pol III structure allows mapping of disease-related mutations and may contribute to the development of inhibitors that selectively target Pol III for therapeutic interventions.
Collapse
|
77
|
Perrier S, Michell-Robinson MA, Bernard G. POLR3-Related Leukodystrophy: Exploring Potential Therapeutic Approaches. Front Cell Neurosci 2021; 14:631802. [PMID: 33633543 PMCID: PMC7902007 DOI: 10.3389/fncel.2020.631802] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 12/28/2020] [Indexed: 12/19/2022] Open
Abstract
Leukodystrophies are a class of rare inherited central nervous system (CNS) disorders that affect the white matter of the brain, typically leading to progressive neurodegeneration and early death. Hypomyelinating leukodystrophies are characterized by the abnormal formation of the myelin sheath during development. POLR3-related or 4H (hypomyelination, hypodontia, and hypogonadotropic hypogonadism) leukodystrophy is one of the most common types of hypomyelinating leukodystrophy for which no curative treatment or disease-modifying therapy is available. This review aims to describe potential therapies that could be further studied for effectiveness in pre-clinical studies, for an eventual translation to the clinic to treat the neurological manifestations associated with POLR3-related leukodystrophy. Here, we discuss the therapeutic approaches that have shown promise in other leukodystrophies, as well as other genetic diseases, and consider their use in treating POLR3-related leukodystrophy. More specifically, we explore the approaches of using stem cell transplantation, gene replacement therapy, and gene editing as potential treatment options, and discuss their possible benefits and limitations as future therapeutic directions.
Collapse
Affiliation(s)
- Stefanie Perrier
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Mackenzie A. Michell-Robinson
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Geneviève Bernard
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Pediatrics, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Department of Specialized Medicine, Division of Medical Genetics, Montréal Children’s Hospital and McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
78
|
Pelletier F, Perrier S, Cayami FK, Mirchi A, Saikali S, Tran LT, Ulrick N, Guerrero K, Rampakakis E, van Spaendonk RML, Naidu S, Pohl D, Gibson WT, Demos M, Goizet C, Tejera-Martin I, Potic A, Fogel BL, Brais B, Sylvain M, Sébire G, Lourenço CM, Bonkowsky JL, Catsman-Berrevoets C, Pinto PS, Tirupathi S, Strømme P, de Grauw T, Gieruszczak-Bialek D, Krägeloh-Mann I, Mierzewska H, Philippi H, Rankin J, Atik T, Banwell B, Benko WS, Blaschek A, Bley A, Boltshauser E, Bratkovic D, Brozova K, Cimas I, Clough C, Corenblum B, Dinopoulos A, Dolan G, Faletra F, Fernandez R, Fletcher J, Garcia Garcia ME, Gasparini P, Gburek-Augustat J, Gonzalez Moron D, Hamati A, Harting I, Hertzberg C, Hill A, Hobson GM, Innes AM, Kauffman M, Kirwin SM, Kluger G, Kolditz P, Kotzaeridou U, La Piana R, Liston E, McClintock W, McEntagart M, McKenzie F, Melançon S, Misbahuddin A, Suri M, Monton FI, Moutton S, Murphy RPJ, Nickel M, Onay H, Orcesi S, Özkınay F, Patzer S, Pedro H, Pekic S, Pineda Marfa M, Pizzino A, Plecko B, Poll-The BT, Popovic V, Rating D, Rioux MF, Rodriguez Espinosa N, Ronan A, Ostergaard JR, Rossignol E, Sanchez-Carpintero R, Schossig A, Senbil N, Sønderberg Roos LK, Stevens CA, Synofzik M, Sztriha L, et alPelletier F, Perrier S, Cayami FK, Mirchi A, Saikali S, Tran LT, Ulrick N, Guerrero K, Rampakakis E, van Spaendonk RML, Naidu S, Pohl D, Gibson WT, Demos M, Goizet C, Tejera-Martin I, Potic A, Fogel BL, Brais B, Sylvain M, Sébire G, Lourenço CM, Bonkowsky JL, Catsman-Berrevoets C, Pinto PS, Tirupathi S, Strømme P, de Grauw T, Gieruszczak-Bialek D, Krägeloh-Mann I, Mierzewska H, Philippi H, Rankin J, Atik T, Banwell B, Benko WS, Blaschek A, Bley A, Boltshauser E, Bratkovic D, Brozova K, Cimas I, Clough C, Corenblum B, Dinopoulos A, Dolan G, Faletra F, Fernandez R, Fletcher J, Garcia Garcia ME, Gasparini P, Gburek-Augustat J, Gonzalez Moron D, Hamati A, Harting I, Hertzberg C, Hill A, Hobson GM, Innes AM, Kauffman M, Kirwin SM, Kluger G, Kolditz P, Kotzaeridou U, La Piana R, Liston E, McClintock W, McEntagart M, McKenzie F, Melançon S, Misbahuddin A, Suri M, Monton FI, Moutton S, Murphy RPJ, Nickel M, Onay H, Orcesi S, Özkınay F, Patzer S, Pedro H, Pekic S, Pineda Marfa M, Pizzino A, Plecko B, Poll-The BT, Popovic V, Rating D, Rioux MF, Rodriguez Espinosa N, Ronan A, Ostergaard JR, Rossignol E, Sanchez-Carpintero R, Schossig A, Senbil N, Sønderberg Roos LK, Stevens CA, Synofzik M, Sztriha L, Tibussek D, Timmann D, Tonduti D, van de Warrenburg BP, Vázquez-López M, Venkateswaran S, Wasling P, Wassmer E, Webster RI, Wiegand G, Yoon G, Rotteveel J, Schiffmann R, van der Knaap MS, Vanderver A, Martos-Moreno GÁ, Polychronakos C, Wolf NI, Bernard G. Endocrine and Growth Abnormalities in 4H Leukodystrophy Caused by Variants in POLR3A, POLR3B, and POLR1C. J Clin Endocrinol Metab 2021; 106:e660-e674. [PMID: 33005949 PMCID: PMC7823228 DOI: 10.1210/clinem/dgaa700] [Show More Authors] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Indexed: 12/22/2022]
Abstract
CONTEXT 4H or POLR3-related leukodystrophy is an autosomal recessive disorder typically characterized by hypomyelination, hypodontia, and hypogonadotropic hypogonadism, caused by biallelic pathogenic variants in POLR3A, POLR3B, POLR1C, and POLR3K. The endocrine and growth abnormalities associated with this disorder have not been thoroughly investigated to date. OBJECTIVE To systematically characterize endocrine abnormalities of patients with 4H leukodystrophy. DESIGN An international cross-sectional study was performed on 150 patients with genetically confirmed 4H leukodystrophy between 2015 and 2016. Endocrine and growth abnormalities were evaluated, and neurological and other non-neurological features were reviewed. Potential genotype/phenotype associations were also investigated. SETTING This was a multicenter retrospective study using information collected from 3 predominant centers. PATIENTS A total of 150 patients with 4H leukodystrophy and pathogenic variants in POLR3A, POLR3B, or POLR1C were included. MAIN OUTCOME MEASURES Variables used to evaluate endocrine and growth abnormalities included pubertal history, hormone levels (estradiol, testosterone, stimulated LH and FSH, stimulated GH, IGF-I, prolactin, ACTH, cortisol, TSH, and T4), and height and head circumference charts. RESULTS The most common endocrine abnormalities were delayed puberty (57/74; 77% overall, 64% in males, 89% in females) and short stature (57/93; 61%), when evaluated according to physician assessment. Abnormal thyroid function was reported in 22% (13/59) of patients. CONCLUSIONS Our results confirm pubertal abnormalities and short stature are the most common endocrine features seen in 4H leukodystrophy. However, we noted that endocrine abnormalities are typically underinvestigated in this patient population. A prospective study is required to formulate evidence-based recommendations for management of the endocrine manifestations of this disorder.
Collapse
Affiliation(s)
- Félixe Pelletier
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Centre, Montreal, QC, Canada
- Division of Child Neurology, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Stefanie Perrier
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Ferdy K Cayami
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Center of Biomedical Research, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Amytice Mirchi
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Centre, Montreal, QC, Canada
| | - Stephan Saikali
- Department of Pathology, Centre Hospitalier Universitaire de Québec, Québec City, QC, Canada
| | - Luan T Tran
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nicole Ulrick
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kether Guerrero
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | | | - Rosalina M L van Spaendonk
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sakkubai Naidu
- Department of Neurogenetics, Kennedy Krieger Institute, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Daniela Pohl
- Division of Neurology, Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada
| | - William T Gibson
- Department of Medical Genetics, University of British Columbia, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Michelle Demos
- Division of Neurology, Department of Pediatrics, University of British Columbia, BC Children’s Hospital, Vancouver, BC, Canada
| | - Cyril Goizet
- Centre de Référence Neurogénétique, Service de Génétique Médicale, Bordeaux University Hospital, and Laboratoire MRGM, INSERM U1211, Université de Bordeaux, Bordeaux, France
| | - Ingrid Tejera-Martin
- Department of Neurology, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Canary Islands, Spain
| | - Ana Potic
- Department of Neurology, Clinic for Child Neurology and Psychiatry, Medical Faculty University of Belgrade, Belgrade, Serbia
| | - Brent L Fogel
- Departments of Neurology and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Bernard Brais
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, Montreal, QC, Canada
| | - Michel Sylvain
- Centre Mère Enfant, CHU de Québec, Québec City, QC, Canada
| | - Guillaume Sébire
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- Department of Pediatrics, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Charles Marques Lourenço
- Faculdade de Medicina, Centro Universitario Estácio de Ribeirão Preto, Ribeirão Preto, SP, Brazil
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Coriene Catsman-Berrevoets
- Department of Paediatric Neurology, Erasmus University Hospital - Sophia Children’s Hospital, 3015 CN Rotterdam, The Netherlands
| | - Pedro S Pinto
- Neuroradiology Department, Centro Hospitalar do Porto, Porto, Portugal
| | - Sandya Tirupathi
- Department of Paediatric Neurology, Royal Belfast Hospital for Sick Children, Belfast, UK
| | - Petter Strømme
- Division of Pediatrics and Adolescent Medicine, Oslo University Hospital, Ullevål, 0450 Oslo, and University of Oslo, Oslo, Norway
| | - Ton de Grauw
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, USA
| | - Dorota Gieruszczak-Bialek
- Department of Medical Genetics, Children’s Memorial Health Institute, Warsaw, Poland
- Department of Pediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Ingeborg Krägeloh-Mann
- Department of Child Neurology, University Children’s Hospital Tübingen, Tübingen, Germany
| | - Hanna Mierzewska
- Department of Child and Adolescent Neurology, Institute of Mother and Child, Warsaw, Poland
| | - Heike Philippi
- Center of Developmental Neurology (SPZ Frankfurt Mitte), Frankfurt, Germany
| | - Julia Rankin
- Peninsula Clinical Genetics Service, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Tahir Atik
- Division of Genetics, Department of Pediatrics, School of Medicine, Ege University, Izmir, Turkey
| | - Brenda Banwell
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - William S Benko
- Division of Pediatric Neurology, Department of Neurology, UC Davis Health System, Sacramento, CA, USA
| | - Astrid Blaschek
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Annette Bley
- University Children’s Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eugen Boltshauser
- Department of Child Neurology, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Drago Bratkovic
- Metabolic Clinic, Women’s and Children’s Hospital, North Adelaide, South Australia, Australia
| | - Klara Brozova
- Department of Child Neurology, Thomayers Hospital, Prague, Czech Republic
| | - Icíar Cimas
- Department of Neurology, Povisa Hospital, Vigo, Spain
| | | | - Bernard Corenblum
- Division of Endocrinology & Metabolism, Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Argirios Dinopoulos
- Third Department of Pediatrics, National and Kapodistrian University of Athens, “Attikon” Hospital, Athens, Greece
| | | | - Flavio Faletra
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | | | - Janice Fletcher
- Genetics and Molecular Pathology, Women’s and Children’s Hospital, Adelaide, South Australia, Australia
| | | | - Paolo Gasparini
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34100 Trieste, and University of Trieste, Trieste, Italy
| | - Janina Gburek-Augustat
- Division of Neuropaediatrics, Hospital for Children and Adolescents, University Leipzig, Leipzig, Germany
| | - Dolores Gonzalez Moron
- Neurogenetics Unit, Department of Neurology, Hospital JM Ramos Mejia, ADC, Buenos Aires, Argentina
| | - Aline Hamati
- Department of Child Neurology, Indiana University, Indianapolis, IN, USA
| | - Inga Harting
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Alan Hill
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Grace M Hobson
- Nemours Biomedical Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - A Micheil Innes
- Department of Medical Genetics and Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Marcelo Kauffman
- Neurogenetics Unit, Department of Neurology, Hospital JM Ramos Mejia and CONICET, ADC, Buenos Aires, Argentina
| | - Susan M Kirwin
- Molecular Diagnostics Laboratory, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Gerhard Kluger
- PMU Salzburg, 5020 Salzburg, Austria; Clinic for Neuropediatrics and Neurorehabilitation, Epilepsy Center for Children and Adolescents, Schön Klinik Vogtareuth, Vogtareuth, Germany
| | - Petra Kolditz
- Department of Child Neurology, Kantonsspital Luzern, Luzern, Switzerland
| | - Urania Kotzaeridou
- Department of Child Neurology, University Children’s Hospital Heidelberg, Heidelberg, Germany
| | - Roberta La Piana
- Department of Neuroradiology, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Eriskay Liston
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada
| | - William McClintock
- Pediatric Specialists of Virginia, Fairfax, VA, USA
- Department of Neurology, Children’s National Medical Center, Washington, DC, USA
| | - Meriel McEntagart
- South West Thames Regional Genetics Service, St. George’s Hospital, London, UK
| | - Fiona McKenzie
- Genetic Services of Western Australia, Subiaco, WA, Australia
- School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia
| | - Serge Melançon
- Department of Medical Genetics, McGill University Health Centre, Montreal Children’s Hospital, Montreal, QC, Canada
| | - Anjum Misbahuddin
- Essex Centre for Neurological Sciences, Queen’s Hospital, Romford, UK
| | - Mohnish Suri
- Nottingham Clinical Genetics Service, City Hospital Campus, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Fernando I Monton
- Department of Neurology, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Canary Islands, Spain
| | | | - Raymond P J Murphy
- Department of Neurology, Tallaght University Hospital, Tallaght, Ireland
| | - Miriam Nickel
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hüseyin Onay
- Department of Medical Genetics, Ege University, Izmir, Turkey
| | - Simona Orcesi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Ferda Özkınay
- Department of Pediatrics, Subdivision of Pediatric Genetics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Steffi Patzer
- Children’s Hospital St. Elisabeth and St. Barbara, Halle (Saale), Germany
| | - Helio Pedro
- Department of Pediatrics, The Joseph M. Sanzari Children’s Hospital, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Sandra Pekic
- Clinic for Endocrinology, Diabetes and Diseases of Metabolism, University Clinical Center, Belgrade & School of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Amy Pizzino
- Department of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Genetics, MetroHealth Hospital, Cleveland, OH, USA
| | - Barbara Plecko
- Department of Pediatrics and Adolescent Medicine, Division of General Pediatrics, Medical University of Graz, Graz, Austria
| | - Bwee Tien Poll-The
- Department of Pediatric Neurology, Emma Children’s Hospital, 1105 Amsterdam, The Netherlands
| | - Vera Popovic
- Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Dietz Rating
- Department of Paediatric Neurology, University Children’s Hospital, Heidelberg, Germany
| | - Marie-France Rioux
- Centre Hospitalier Universitaire de Sherbrooke - Hôpital Fleurimont, Sherbrooke, QC, Canada
| | - Norberto Rodriguez Espinosa
- Department of Neurology, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Canary Islands, Spain
| | - Anne Ronan
- Hunter New England LHD, University of Newcastle, NSW, Australia
| | - John R Ostergaard
- Centre for Rare Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Elsa Rossignol
- Departments of Neurosciences and Pediatrics, CHU-Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Rocio Sanchez-Carpintero
- Pediatric Neurology Unit, Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain
| | - Anna Schossig
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, Austria
| | - Nesrin Senbil
- Department of Child Neurology, Kırıkkale University Medical Faculty, Kırıkkale, Turkey
| | - Laura K Sønderberg Roos
- Applied Human Molecular Genetics, Kennedy Center, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark
| | - Cathy A Stevens
- Department of Pediatrics, Division of Medical Genetics, University of Tennessee College of Medicine, Chattanooga, TN, USA
| | - Matthis Synofzik
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research and Centre of Neurology, German Research Center for Neurodegenerative Diseases (DZNE), University of Tübingen, Tübingen, Germany
| | - László Sztriha
- Department of Paediatrics, University of Szeged, Szeged, Hungary
| | - Daniel Tibussek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Dagmar Timmann
- Department of Neurology, Essen University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Davide Tonduti
- Child Neurology Unit, V. Buzzi Children’s Hospital, Milano, Italy
| | - Bart P van de Warrenburg
- Department of Neurology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Maria Vázquez-López
- Sección Neuropediatría. Hospital Maternoinfantil Gregorio Marañón, Madrid, Spain
| | - Sunita Venkateswaran
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Pontus Wasling
- Department of Neuroscience and Rehabilitation, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Richard I Webster
- T. Y. Nelson Department of Neurology and Neurosurgery and the Institute for Neuroscience and Muscle Research, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
| | - Gert Wiegand
- Department of Pediatric Neurology, University Hospital Kiel, Germany
- Neuropediatrics Section of the Department of Pediatrics, Asklepios Clinic Hamburg Nord-Heidberg, Hamburg, Germany
| | - Grace Yoon
- Division of Clinical and Metabolic Genetics, Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Joost Rotteveel
- Emma Children’s Hospital, Amsterdam UMC, Pediatric Endocrinology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Raphael Schiffmann
- Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Marjo S van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - Adeline Vanderver
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriel Á Martos-Moreno
- Department of Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- CIBER de Fisiopatologia de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Constantin Polychronakos
- Division of Endocrinology, Montreal Children’s Hospital and the Endocrine Genetics Lab, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nicole I Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Geneviève Bernard
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
79
|
Djordjevic D, Pinard M, Gauthier MS, Smith-Hicks C, Hoffman TL, Wolf NI, Oegema R, van Binsbergen E, Baskin B, Bernard G, Fribourg S, Coulombe B, Yoon G. De novo variants in POLR3B cause ataxia, spasticity, and demyelinating neuropathy. Am J Hum Genet 2021; 108:186-193. [PMID: 33417887 DOI: 10.1016/j.ajhg.2020.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022] Open
Abstract
POLR3B encodes the second-largest catalytic subunit of RNA polymerase III, an enzyme involved in transcription. Bi-allelic pathogenic variants in POLR3B are a well-established cause of hypomyelinating leukodystrophy. We describe six unrelated individuals with de novo missense variants in POLR3B and a clinical presentation substantially different from POLR3-related leukodystrophy. These individuals had afferent ataxia, spasticity, variable intellectual disability and epilepsy, and predominantly demyelinating sensory motor peripheral neuropathy. Protein modeling and proteomic analysis revealed a distinct mechanism of pathogenicity; the de novo POLR3B variants caused aberrant association of individual enzyme subunits rather than affecting overall enzyme assembly or stability. We expand the spectrum of disorders associated with pathogenic variants in POLR3B to include a de novo heterozygous POLR3B-related disorder.
Collapse
|
80
|
Rosenfield RL, Cooke DW, Radovick S. Puberty in the Female and Its Disorders. SPERLING PEDIATRIC ENDOCRINOLOGY 2021:528-626. [DOI: 10.1016/b978-0-323-62520-3.00016-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
81
|
Abstract
Hypomyelinating leukodystrophies constitute a subset of genetic white matter disorders characterized by a primary lack of myelin deposition. Most patients with severe hypomyelination present in infancy or early childhood and develop severe neurological deficits, but the clinical presentation can also be mild with onset of symptoms in adolescence or adulthood. MRI can be used to visualize the process of myelination in detail, and MRI pattern recognition can provide a clinical diagnosis in many patients. Next-generation sequencing provides a definitive diagnosis in 80-90% of patients. Genes associated with hypomyelination include those that encode structural myelin proteins but also many that encode proteins involved in RNA translation and some lysosomal proteins. The precise pathomechanisms remain to be elucidated. Improved understanding of the process of myelination, the metabolic axonal support functions of myelin and the proposed contribution of myelin to CNS plasticity provide possible explanations as to why almost all patients with hypomyelination experience slow clinical decline after a long phase of stability. In this Review, we provide an overview of the hypomyelinating leukodystrophies, the advances in our understanding of myelin biology and of the genes involved in these disorders, and the insights these advances have provided into their clinical presentations and evolution.
Collapse
|
82
|
Di Bella D, Magri S, Benzoni C, Farina L, Maccagnano C, Sarto E, Moscatelli M, Baratta S, Ciano C, Piacentini SHMJ, Draghi L, Mauro E, Pareyson D, Gellera C, Taroni F, Salsano E. Hypomyelinating leukodystrophies in adults: Clinical and genetic features. Eur J Neurol 2020; 28:934-944. [PMID: 33190326 DOI: 10.1111/ene.14646] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND PURPOSE Little is known about hypomyelinating leukodystrophies (HLDs) in adults. The aim of this study was to investigate HLD occurrence, clinical features, and etiology among undefined leukoencephalopathies in adulthood. METHODS We recruited the patients with cerebral hypomyelinating magnetic resonance imaging pattern (mild T2 hyperintensity with normal or near-normal T1 signal) from our cohort of 62 adult index cases with undefined leukoencephalopathies, reviewed their clinical features, and used a leukoencephalopathy-targeted next generation sequencing panel. RESULTS We identified 25/62 patients (~40%) with hypomyelination. Cardinal manifestations were spastic gait and varying degree of cognitive impairment. Etiology was determined in 44% (definite, 10/25; likely, 1/25). Specifically, we found pathogenic variants in the POLR3A (n = 2), POLR1C (n = 1), RARS1 (n = 1), and TUBB4A (n = 1) genes, which are typically associated with severe early-onset HLDs, and in the GJA1 gene (n = 1), which is associated with oculodentodigital dysplasia. Duplication of a large chromosome X region encompassing PLP1 and a pathogenic GJC2 variant were found in two patients, both females, with early-onset HLDs persisting into adulthood. Finally, we found likely pathogenic variants in PEX3 (n = 1) and PEX13 (n = 1) and potentially relevant variants of unknown significance in TBCD (n = 1), which are genes associated with severe, early-onset diseases with central hypomyelination/dysmyelination. CONCLUSIONS A hypomyelinating pattern characterizes a relevant number of undefined leukoencephalopathies in adulthood. A comprehensive genetic screening allows definite diagnosis in about half of patients, and demonstrates the involvement of many disease-causing genes, including genes associated with severe early-onset HLDs, and genes causing peroxisome biogenesis disorders.
Collapse
Affiliation(s)
- Daniela Di Bella
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefania Magri
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Benzoni
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Laura Farina
- Unit of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Neuroimaging Laboratory, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Carmelo Maccagnano
- Unit of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elisa Sarto
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Marco Moscatelli
- Unit of Neuroradiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Silvia Baratta
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudia Ciano
- Unit of Neurophysiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Lara Draghi
- Unit of Neuropsychology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elena Mauro
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Davide Pareyson
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ettore Salsano
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Neuroscience PhD Program, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
83
|
Adult-onset 4H leukodystrophy: a case presentation and review of the literature. Acta Neurol Belg 2020; 120:1461-1462. [PMID: 32052360 DOI: 10.1007/s13760-020-01297-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/30/2020] [Indexed: 10/25/2022]
|
84
|
Moir RD, Lavados C, Lee J, Willis IM. Functional characterization of Polr3a hypomyelinating leukodystrophy mutations in the S. cerevisiae homolog, RPC160. Gene 2020; 768:145259. [PMID: 33148458 DOI: 10.1016/j.gene.2020.145259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/23/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
Mutations in RNA polymerase III (Pol III) cause hypomeylinating leukodystrophy (HLD) and neurodegeneration in humans. POLR3A and POLR3B, the two largest Pol III subunits, together form the catalytic center and carry the majority of disease alleles. Disease-causing mutations include invariant and highly conserved residues that are predicted to negatively affect Pol III activity and decrease transcriptional output. A subset of HLD missense mutations in POLR3A cluster in the pore region that provides nucleotide access to the Pol III active site. These mutations were engineered at the corresponding positions in the Saccharomyces cerevisiae homolog, Rpc160, to evaluate their functional deficits. None of the mutations caused a growth or transcription phenotype in yeast. Each mutation was combined with a frequently occurring pore mutation, POLR3A G672E, which was also wild-type for growth and transcription. The double mutants showed a spectrum of phenotypes from wild-type to lethal, with only the least fit combinations showing an effect on Pol III transcription. In one slow-growing temperature-sensitive mutant the steady-state level of tRNAs was unaffected, however global tRNA synthesis was compromised, as was the synthesis of RPR1 and SNR52 RNAs. Affinity-purified mutant Pol III was broadly defective in both factor-independent and factor-dependent transcription in vitro across genes that represent the yeast Pol III transcriptome. Thus, the robustness of yeast Rpc160 to single Pol III leukodystrophy mutations in the pore domain can be overcome by a second mutation in the domain.
Collapse
Affiliation(s)
- Robyn D Moir
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Christian Lavados
- Graduate Program in Biomedical Science, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - JaeHoon Lee
- Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ian M Willis
- Departments of Biochemistry and Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
85
|
Kashiki H, Li H, Miyamoto S, Ueno H, Tsurusaki Y, Ikeda C, Kurata H, Okada T, Shimazu T, Imamura H, Enomoto Y, Takanashi JI, Kurosawa K, Saitsu H, Inoue K. POLR1C variants dysregulate splicing and cause hypomyelinating leukodystrophy. NEUROLOGY-GENETICS 2020; 6:e524. [PMID: 33134519 PMCID: PMC7577547 DOI: 10.1212/nxg.0000000000000524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/19/2020] [Indexed: 12/20/2022]
Abstract
Objective To further clarify the molecular pathogenesis of RNA polymerase III (Pol III)-related leukodystrophy caused by biallelic POLR1C variants at a cellular level and potential effects on its downstream genes. Methods Exome analysis and molecular functional studies using cell expression and long-read sequencing analyses were performed on 1 family with hypomyelinating leukodystrophy showing no clinical and MRI findings characteristic of Pol III–related leukodystrophy other than hypomyelination. Results Biallelic novel POLR1C alterations, c.167T>A, p.M56K and c.595A>T, p.I199F, were identified as causal variants. Functional analyses showed that these variants not only resulted in altered protein subcellular localization and decreased protein expression but also caused abnormal inclusion of introns in 85% of the POLR1C transcripts in patient cells. Unexpectedly, allelic segregation analysis in each carrier parent revealed that each heterozygous variant also caused the inclusion of introns on both mutant and wild-type alleles. These findings suggest that the abnormal splicing is not direct consequences of the variants, but rather reflect the downstream effect of the variants in dysregulating splicing of POLR1C, and potentially other target genes. Conclusions The lack of characteristic clinical findings in this family confirmed the broad clinical spectrum of Pol III–related leukodystrophy. Molecular studies suggested that dysregulation of splicing is the potential downstream pathomechanism for POLR1C variants.
Collapse
Affiliation(s)
- Hitoshi Kashiki
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Heng Li
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Sachiko Miyamoto
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hiroe Ueno
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yoshinori Tsurusaki
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Chizuru Ikeda
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hirofumi Kurata
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Takumi Okada
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Tomoyuki Shimazu
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hoseki Imamura
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yumi Enomoto
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Jun-Ichi Takanashi
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kenji Kurosawa
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hirotomo Saitsu
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| | - Ken Inoue
- Department of Pediatrics (H.K.), Minamata City General Hospital & Medical Center, Kumamoto; Department of Mental Retardation and Birth Defect Research (H.L., K.I.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo; Department of Biochemistry (S.M., H.S.), Hamamatsu University School of Medicine, Shizuoka; Department of Pediatrics (H.U.), Kumamoto Takumadai Rehabilitation Hospital; Kanagawa Children's Medical Center (Y.T., Y.E.), Clinical Research Institute, Yokohama, Kanagawa; Department of Pediatrics (C.I., H.K., T.O., T.S., H.I.), National Hospital Organization Kumamoto Saishun Medical Center, Koshi; Clinical Research Institute, Kanagawa Children's Medical Center, (Y.E.), Yokohama, Kanagawa; Department of Pediatric Neurology (J.T.), Tokyo Women's Medical University Yachiyo Medical Center, Chiba; and Division of Medical Genetics (K.K.), Kanagawa Children's Medical Center, Yokohama, Japan
| |
Collapse
|
86
|
Schnabel F, Kornak U, Wollnik B. Premature aging disorders: A clinical and genetic compendium. Clin Genet 2020; 99:3-28. [PMID: 32860237 DOI: 10.1111/cge.13837] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/22/2022]
Abstract
Progeroid disorders make up a heterogeneous group of very rare hereditary diseases characterized by clinical signs that often mimic physiological aging in a premature manner. Apart from Hutchinson-Gilford progeria syndrome, one of the best-investigated progeroid disorders, a wide spectrum of other premature aging phenotypes exist, which differ significantly in their clinical presentation and molecular pathogenesis. Next-generation sequencing (NGS)-based approaches have made it feasible to determine the molecular diagnosis in the early stages of a disease. Nevertheless, a broad clinical knowledge on these disorders and their associated symptoms is still fundamental for a comprehensive patient management and for the interpretation of variants of unknown significance from NGS data sets. This review provides a detailed overview on characteristic clinical features and underlying molecular genetics of well-known as well as only recently identified premature aging disorders and also highlights novel findings towards future therapeutic options.
Collapse
Affiliation(s)
- Franziska Schnabel
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Uwe Kornak
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Bernd Wollnik
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable cells" (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
87
|
Tailland M, Gonzalez V, Prin P, Carra-Dallière C, Ayrignac X, Labauge P. POLR3-related disorder revealed by movement disorders. Rev Neurol (Paris) 2020; 177:328-330. [PMID: 33004219 DOI: 10.1016/j.neurol.2020.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/02/2020] [Accepted: 06/22/2020] [Indexed: 11/24/2022]
Affiliation(s)
- Marion Tailland
- MS Unit and Reference Center for adult-onset leukoencephalopathies (MT, PP, XA, CCD, PL) and department of Neurology (VG), Montpellier University Hospital, Gui de Chauliac, 34295 Montpellier cedex 5, France
| | - Victoria Gonzalez
- MS Unit and Reference Center for adult-onset leukoencephalopathies (MT, PP, XA, CCD, PL) and department of Neurology (VG), Montpellier University Hospital, Gui de Chauliac, 34295 Montpellier cedex 5, France
| | - Pauline Prin
- MS Unit and Reference Center for adult-onset leukoencephalopathies (MT, PP, XA, CCD, PL) and department of Neurology (VG), Montpellier University Hospital, Gui de Chauliac, 34295 Montpellier cedex 5, France
| | - Clarisse Carra-Dallière
- MS Unit and Reference Center for adult-onset leukoencephalopathies (MT, PP, XA, CCD, PL) and department of Neurology (VG), Montpellier University Hospital, Gui de Chauliac, 34295 Montpellier cedex 5, France
| | - Xavier Ayrignac
- MS Unit and Reference Center for adult-onset leukoencephalopathies (MT, PP, XA, CCD, PL) and department of Neurology (VG), Montpellier University Hospital, Gui de Chauliac, 34295 Montpellier cedex 5, France
| | - Pierre Labauge
- MS Unit and Reference Center for adult-onset leukoencephalopathies (MT, PP, XA, CCD, PL) and department of Neurology (VG), Montpellier University Hospital, Gui de Chauliac, 34295 Montpellier cedex 5, France.
| |
Collapse
|
88
|
Malik P, Muthusamy K, Mankad K, Shroff M, Sudhakar S. Solving the hypomyelination conundrum - Imaging perspectives. Eur J Paediatr Neurol 2020; 27:9-24. [PMID: 32418752 DOI: 10.1016/j.ejpn.2020.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 03/25/2020] [Accepted: 04/14/2020] [Indexed: 11/26/2022]
Abstract
Hypomyelinating Leukodystrophies (HLDs) are a genetically heterogeneous, clinically overlapping group of disorders with the unifying MR imaging appearance of myelin deficit in the brain. In fact, it is the MRI phenotype that typically raises the diagnostic suspicion in this single largest group of undiagnosed leukodystrophies and guides gene testing for confirmation. This article reviews the neurobiology of myelination, focussing on the complex interplay of molecular genetic pathways and presents a practical clinico-radiological diagnostic algorithm based on the neuroimaging patterns of the common hypomyelinating disorders. The authors also address the current controversies about the definition and use of the term 'hypomyelination'.
Collapse
|
89
|
Campopiano R, Ferese R, Zampatti S, Giardina E, Biagioni F, Colonnese C, Centonze D, Storto M, Buttari F, Fraviga E, Broccoli V, Fanelli M, Fornai F, Gambardella S. A novel POLR3A genotype leads to leukodystrophy type-7 in two siblings with unusually late age of onset. BMC Neurol 2020; 20:258. [PMID: 32600288 PMCID: PMC7322863 DOI: 10.1186/s12883-020-01835-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/19/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Leukodystrophies are familial heterogeneous disorders primarily affecting the white matter, which are defined as hypomyelinating or demyelinating based on disease severity as assessed at MRI. Recently, a group of clinically overlapping hypomyelinating leukodystrophies (HL) has been associated with mutations in RNA polymerase III enzymes (Pol III) subunits. CASE PRESENTATION In this manuscript, we describe two Italian siblings carrying a novel POLR3A genotype. MRI imaging, genetic analysis, and clinical data led to diagnosing HL type 7. The female sibling, at the age of 34, is tetra-paretic and suffers from severe cognitive regression. She had a disease onset at the age of 19, characterized by slow and progressive cognitive impairment associated with gait disturbances and amenorrhea. The male sibling was diagnosed during an MRI carried out for cephalalgia at the age of 41. After 5 years, he developed mild cognitive impairment, dystonia with 4-limb hypotonia, and moderate dysmetria with balance and gait impairment. CONCLUSIONS The present study provides the first evidence of unusually late age of onset in HL, describing two siblings with a novel POLR3A genotype which showed the first symptoms at the age of 41 and 19, respectively. This provides a powerful insight into clinical heterogeneity and genotype-phenotype correlation in POLR3A related HL.
Collapse
Affiliation(s)
- Rosa Campopiano
- I.R.C.C.S. I.N.M. Neuromed, via Atinense 18, 86077, Pozzilli, Italy
| | - Rosangela Ferese
- I.R.C.C.S. I.N.M. Neuromed, via Atinense 18, 86077, Pozzilli, Italy
| | - Stefania Zampatti
- Molecular Genetics Laboratory UILDM, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Emiliano Giardina
- Molecular Genetics Laboratory UILDM, Santa Lucia Foundation IRCCS, Rome, Italy.,Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | | | | | - Diego Centonze
- I.R.C.C.S. I.N.M. Neuromed, via Atinense 18, 86077, Pozzilli, Italy.,Dipartimento di Medicina dei Sistemi, Università di Roma Tor Vergata, Rome, Italy
| | - Marianna Storto
- I.R.C.C.S. I.N.M. Neuromed, via Atinense 18, 86077, Pozzilli, Italy
| | - Fabio Buttari
- I.R.C.C.S. I.N.M. Neuromed, via Atinense 18, 86077, Pozzilli, Italy
| | - Edoardo Fraviga
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy.,CNR Institute of Neuroscience, Milan, Italy
| | - Mirco Fanelli
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Francesco Fornai
- I.R.C.C.S. I.N.M. Neuromed, via Atinense 18, 86077, Pozzilli, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Stefano Gambardella
- I.R.C.C.S. I.N.M. Neuromed, via Atinense 18, 86077, Pozzilli, Italy. .,Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy.
| |
Collapse
|
90
|
Perrier S, Gauquelin L, Fallet-Bianco C, Dishop MK, Michell-Robinson MA, Tran LT, Guerrero K, Darbelli L, Srour M, Petrecca K, Renaud DL, Saito M, Cohen S, Leiz S, Alhaddad B, Haack TB, Tejera-Martin I, Monton FI, Rodriguez-Espinosa N, Pohl D, Nageswaran S, Grefe A, Glamuzina E, Bernard G. Expanding the phenotypic and molecular spectrum of RNA polymerase III-related leukodystrophy. NEUROLOGY-GENETICS 2020; 6:e425. [PMID: 32582862 PMCID: PMC7238899 DOI: 10.1212/nxg.0000000000000425] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/25/2020] [Indexed: 12/16/2022]
Abstract
Objective To expand the phenotypic spectrum of severity of POLR3-related leukodystrophy and identify genotype-phenotype correlations through study of patients with extremely severe phenotypes. Methods We performed an international cross-sectional study on patients with genetically proven POLR3-related leukodystrophy and atypical phenotypes to identify 6 children, 3 males and 3 females, with an extremely severe phenotype compared with that typically reported. Clinical, radiologic, and molecular features were evaluated for all patients, and functional and neuropathologic studies were performed on 1 patient. Results Each patient presented between 1 and 3 months of age with failure to thrive, severe dysphagia, and developmental delay. Four of the 6 children died before age 3 years. MRI of all patients revealed a novel pattern with atypical characteristics, including progressive basal ganglia and thalami abnormalities. Neuropathologic studies revealed patchy areas of decreased myelin in the cerebral hemispheres, cerebellum, brainstem, and spinal cord, with astrocytic gliosis in the white matter and microglial activation. Cellular vacuolization was observed in the thalamus and basal ganglia, and neuronal loss was evident in the putamen and caudate. Genotypic similarities were also present between all 6 patients, with one allele containing a POLR3A variant causing a premature stop codon and the other containing a specific intronic splicing variant (c.1771-7C>G), which produces 2 aberrant transcripts along with some wild-type transcript. Conclusions We describe genotype-phenotype correlations at the extreme end of severity of the POLR3-related leukodystrophy spectrum and shed light on the complex disease pathophysiology.
Collapse
Affiliation(s)
- Stefanie Perrier
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Laurence Gauquelin
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Catherine Fallet-Bianco
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Megan K Dishop
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Mackenzie A Michell-Robinson
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Luan T Tran
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Kether Guerrero
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Lama Darbelli
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Myriam Srour
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Kevin Petrecca
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Deborah L Renaud
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Michael Saito
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Seth Cohen
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Steffen Leiz
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Bader Alhaddad
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Tobias B Haack
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Ingrid Tejera-Martin
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Fernando I Monton
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Norberto Rodriguez-Espinosa
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Daniela Pohl
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Savithri Nageswaran
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Annette Grefe
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Emma Glamuzina
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| | - Geneviève Bernard
- Department of Neurology and Neurosurgery (S.P., L.G., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, K.P., G.B.), McGill University; Child Health and Human Development Program (S.P., M.A.M.-R., L.T.T., K.G., L.D., M. Srour, G.B.), Research Institute of the McGill University Health Centre; Department of Pediatrics (L.G., L.T.T., K.G., L.D., M. Srour, G.B.), McGill University, Montreal, Quebec, Canada; Division of Clinical and Metabolic Genetics (L.G.), Division of Neurology, the Hospital for Sick Children, University of Toronto, Ontario, Canada; Department of Pathology (C.F.-B.), CHU Sainte-Justine, Université de Montreal, Quebec, Canada; Division of Pathology and Laboratory Medicine (M.K.D.), Phoenix Children's Hospital, AZ; Department of Human Genetics (L.T.T., K.G., L.D., G.B.), McGill University, Montreal, Quebec, Canada; McGill University (K.P.), Brain Tumour Research Center Montreal Neurological Institute and Hospital, Quebec, Canada; Department of Neurology (D.L.R.), Department of Clinical Genomics, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Pediatrics (M. Saito), University of California Riverside School of Medicine, Riverside Medical Clinic, CA; Department of Pediatrics (S.C.), Beaver Medical Group, Redlands, CA; Division of Pediatric Neurology (S.L.), Department of Pediatrics, Klinikum Dritter Orden, Munich, Germany; Institute of Human Genetics (B.A., T.B.H.), Technische Universität München, Munich, Germany; Institute of Medical Genetics and Applied Genomics (T.B.H.), University of Tübingen, Germany; Department of Neurology (I.T.-M., F.I.M., N.R.-E.), Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canary Islands, Spain; Department of Neurology (D.P.), Children's Hospital of Eastern Ontario, University of Ottawa, Ontario, Canada; Department of Pediatrics (S.N.) and Department of Neurology (A.G.), Wake Forest School of Medicine, Winston-Salem, NC; Adult and Paediatric National Metabolic Service (E.G.), Starship Children's Hospital, Auckland, New Zealand; and Division of Medical Genetics (G.B.), Department of Specialized Medicine, Montreal Children's Hospital and McGill University Health Centre, Quebec, Canada
| |
Collapse
|
91
|
Verberne EA, Dalen Meurs L, Wolf NI, van Haelst MM. 4H leukodystrophy caused by a homozygous POLR3B mutation: Further delineation of the phenotype. Am J Med Genet A 2020; 182:1776-1779. [PMID: 32319736 PMCID: PMC7318643 DOI: 10.1002/ajmg.a.61600] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/13/2020] [Accepted: 04/01/2020] [Indexed: 11/11/2022]
Abstract
4H leukodystrophy, also known as Pol III-related leukodystrophy, is a rare autosomal recessive neurodegenerative disorder characterized by hypomyelination, hypodontia, and hypogonadotropic hypogonadism. It is caused by biallelic mutations in POLR3A, POL3RB, or POLR1C. So far, only two patients have been described with homozygosity for the common c.1568T>A (p.Val523Glu) POLR3B mutation, both of them showing a remarkably mild clinical course. Here, we report another patient with homozygosity for the same mutation, but with a more severe phenotype including ataxia, developmental delay, and intellectual disability. This information is of importance for clinicians to provide comprehensive counseling to patients with 4H leukodystrophy and their families.
Collapse
Affiliation(s)
- Eline A Verberne
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Lotje Dalen Meurs
- Department of Pediatrics, Fundashon Mariadal, Kralendijk, Bonaire, Netherlands.,Department of Pediatrics, Meander Medical Centre, Amersfoort, Netherlands
| | - Nicole I Wolf
- Department of Child Neurology, Emma Children's Hospital, Amsterdam UMC, and Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, Netherlands
| | - Mieke M van Haelst
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
92
|
de Assis Pereira Matos PCA, Gama MTD, Bezerra MLE, da Rocha AJ, Barsottini OGP, Pedroso JL. POLR3A-Related Disorder Presenting with Late-Onset Dystonia and Spastic Paraplegia. Mov Disord Clin Pract 2020; 7:467-469. [PMID: 32373668 DOI: 10.1002/mdc3.12945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/06/2020] [Accepted: 03/16/2020] [Indexed: 11/09/2022] Open
Affiliation(s)
| | | | | | - Antônio José da Rocha
- Department of Radiology, Faculdade de Ciências Médicas da Santa Casa de São Paulo São Paulo Brazil
| | | | - José Luiz Pedroso
- Department of Neurology, Ataxia Unit Universidade Federal de São Paulo São Paulo Brazil
| |
Collapse
|
93
|
Hiraide T, Kubota K, Kono Y, Watanabe S, Matsubayashi T, Nakashima M, Kaname T, Fukao T, Shimozawa N, Ogata T, Saitsu H. POLR3A variants in striatal involvement without diffuse hypomyelination. Brain Dev 2020; 42:363-368. [PMID: 31932101 DOI: 10.1016/j.braindev.2019.12.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/26/2019] [Accepted: 12/27/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Biallelic variants in POLR3A encoding the largest subunit of RNA polymerase III cause POLR3-related (or 4H) leukodystrophy characterized by neurologic dysfunction, abnormal dentition, endocrine abnormalities and ocular abnormality. Recently, whole-exome sequencing enabled the discovery of POLR3A variants in cases lacking diffuse hypomyelination, the principal MRI phenotype of POLR3-related leukodystrophy. Homozygous c.1771-6C > G variants in POLR3A were recently suggested to cause striatal and red nucleus involvement without white matter involvement. CASE REPORT Here, we report three cases in two families with biallelic POLR3A variants. We identified two sets of compound heterozygous variants in POLR3A, c.1771-6C > G and c.791C > T, p.(Pro264Leu) for family 1 and c.1771-6C > G and c.2671C > T, p.(Arg891*) for family 2. Both families had the c.1771-6C > G variant, which led to aberrant mRNA splicing. Neuropsychiatric regression and severe intellectual disability were identified in three patients. Two cases showed dystonia and oligodontia. Notably, characteristic bilateral symmetric atrophy and abnormal signal of the striatum without diffuse white matter signal change were observed in brain MRI of all three individuals. CONCLUSIONS Striatum abnormalities may be another distinctive MRI finding associated with POLR3A variants, especially in cases including c.1771-6C > G variants and our cases can expand the phenotypic spectrum of POLR3A-related disorders.
Collapse
Affiliation(s)
- Takuya Hiraide
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan; Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuo Kubota
- Department of Pediatrics, Gifu University Graduate School of Medicine, Gifu, Japan; Division of Clinical Genetics, Gifu University Hospital, Japan
| | - Yu Kono
- Department of Neurology, Fuji City General Hospital, Shizuoka, Japan
| | - Seiji Watanabe
- Department of Pediatric Neurology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Tomoko Matsubayashi
- Department of Pediatric Neurology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Mitsuko Nakashima
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Toshiyuki Fukao
- Department of Pediatrics, Gifu University Graduate School of Medicine, Gifu, Japan; Division of Clinical Genetics, Gifu University Hospital, Japan
| | - Nobuyuki Shimozawa
- Division of Clinical Genetics, Gifu University Hospital, Japan; Division of Genomics Research, Life Science Research Center, Gifu University, Gifu, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| |
Collapse
|
94
|
DeGasperis SM, Bernard G, Wolf NI, Miller E, Pohl D. 4H leukodystrophy: Mild clinical phenotype and comorbidity with multiple sclerosis. NEUROLOGY-GENETICS 2020; 6:e409. [PMID: 32337336 PMCID: PMC7164972 DOI: 10.1212/nxg.0000000000000409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/18/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Stephanie M DeGasperis
- Faculty of Medicine (S.M.D., D.P.), University of Ottawa, ON, Canada; Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics (G.B.), McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center; MyeliNeuroGene Laboratory (G.B.), Research Institute of the McGill University Health Center, Montreal, Quebec, Canada; Department of Pediatric Neurology (N.I.W.), Emma Children's Hospital, Amsterdam, Netherlands; Amsterdam Neuroscience (N.I.W.), Vrije Universiteit, Netherlands; and Department of Medical Imaging (E.M.) and Division of Neurology (D.P.), CHEO, University of Ottawa, ON, Canada
| | - Geneviève Bernard
- Faculty of Medicine (S.M.D., D.P.), University of Ottawa, ON, Canada; Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics (G.B.), McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center; MyeliNeuroGene Laboratory (G.B.), Research Institute of the McGill University Health Center, Montreal, Quebec, Canada; Department of Pediatric Neurology (N.I.W.), Emma Children's Hospital, Amsterdam, Netherlands; Amsterdam Neuroscience (N.I.W.), Vrije Universiteit, Netherlands; and Department of Medical Imaging (E.M.) and Division of Neurology (D.P.), CHEO, University of Ottawa, ON, Canada
| | - Nicole I Wolf
- Faculty of Medicine (S.M.D., D.P.), University of Ottawa, ON, Canada; Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics (G.B.), McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center; MyeliNeuroGene Laboratory (G.B.), Research Institute of the McGill University Health Center, Montreal, Quebec, Canada; Department of Pediatric Neurology (N.I.W.), Emma Children's Hospital, Amsterdam, Netherlands; Amsterdam Neuroscience (N.I.W.), Vrije Universiteit, Netherlands; and Department of Medical Imaging (E.M.) and Division of Neurology (D.P.), CHEO, University of Ottawa, ON, Canada
| | - Elka Miller
- Faculty of Medicine (S.M.D., D.P.), University of Ottawa, ON, Canada; Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics (G.B.), McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center; MyeliNeuroGene Laboratory (G.B.), Research Institute of the McGill University Health Center, Montreal, Quebec, Canada; Department of Pediatric Neurology (N.I.W.), Emma Children's Hospital, Amsterdam, Netherlands; Amsterdam Neuroscience (N.I.W.), Vrije Universiteit, Netherlands; and Department of Medical Imaging (E.M.) and Division of Neurology (D.P.), CHEO, University of Ottawa, ON, Canada
| | - Daniela Pohl
- Faculty of Medicine (S.M.D., D.P.), University of Ottawa, ON, Canada; Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics (G.B.), McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center; MyeliNeuroGene Laboratory (G.B.), Research Institute of the McGill University Health Center, Montreal, Quebec, Canada; Department of Pediatric Neurology (N.I.W.), Emma Children's Hospital, Amsterdam, Netherlands; Amsterdam Neuroscience (N.I.W.), Vrije Universiteit, Netherlands; and Department of Medical Imaging (E.M.) and Division of Neurology (D.P.), CHEO, University of Ottawa, ON, Canada
| |
Collapse
|
95
|
Phenotypic and genotypic characterization of families with complex intellectual disability identified pathogenic genetic variations in known and novel disease genes. Sci Rep 2020; 10:968. [PMID: 31969655 PMCID: PMC6976666 DOI: 10.1038/s41598-020-57929-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/03/2020] [Indexed: 12/04/2022] Open
Abstract
Intellectual disability (ID), which presents itself during childhood, belongs to a group of neurodevelopmental disorders (NDDs) that are clinically widely heterogeneous and highly heritable, often being caused by single gene defects. Indeed, NDDs can be attributed to mutations at over 1000 loci, and all type of mutations, ranging from single nucleotide variations (SNVs) to large, complex copy number variations (CNVs), have been reported in patients with ID and other related NDDs. In this study, we recruited seven different recessive NDD families with comorbidities to perform a detailed clinical characterization and a complete genomic analysis that consisted of a combination of high throughput SNP-based genotyping and whole-genome sequencing (WGS). Different disease-associated loci and pathogenic gene mutations were identified in each family, including known (n = 4) and novel (n = 2) mutations in known genes (NAGLU, SLC5A2, POLR3B, VPS13A, SYN1, SPG11), and the identification of a novel disease gene (n = 1; NSL1). Functional analyses were additionally performed in a gene associated with autism-like symptoms and epileptic seizures for further proof of pathogenicity. Lastly, detailed genotype-phenotype correlations were carried out to assist with the diagnosis of prospective families and to determine genomic variation with clinical relevance. We concluded that the combination of linkage analyses and WGS to search for disease genes still remains a fruitful strategy for complex diseases with a variety of mutated genes and heterogeneous phenotypic manifestations, allowing for the identification of novel mutations, genes, and phenotypes, and leading to improvements in both diagnostic strategies and functional characterization of disease mechanisms.
Collapse
|
96
|
POLR3A variants with striatal involvement and extrapyramidal movement disorder. Neurogenetics 2020; 21:121-133. [PMID: 31940116 PMCID: PMC7064625 DOI: 10.1007/s10048-019-00602-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/27/2019] [Indexed: 01/10/2023]
Abstract
Biallelic variants in POLR3A cause 4H leukodystrophy, characterized by hypomyelination in combination with cerebellar and pyramidal signs and variable non-neurological manifestations. Basal ganglia are spared in 4H leukodystrophy, and dystonia is not prominent. Three patients with variants in POLR3A, an atypical presentation with dystonia, and MR involvement of putamen and caudate nucleus (striatum) and red nucleus have previously been reported. Genetic, clinical findings and 18 MRI scans from nine patients with homozygous or compound heterozygous POLR3A variants and predominant striatal changes were retrospectively reviewed in order to characterize the striatal variant of POLR3A-associated disease. Prominent extrapyramidal involvement was the predominant clinical sign in all patients. The three youngest children were severely affected with muscle hypotonia, impaired head control, and choreic movements. Presentation of the six older patients was milder. Two brothers diagnosed with juvenile parkinsonism were homozygous for the c.1771-6C > G variant in POLR3A; the other seven either carried c.1771-6C > G (n = 1) or c.1771-7C > G (n = 7) together with another variant (missense, synonymous, or intronic). Striatal T2-hyperintensity and atrophy together with involvement of the superior cerebellar peduncles were characteristic. Additional MRI findings were involvement of dentate nuclei, hila, or peridentate white matter (3, 6, and 4/9), inferior cerebellar peduncles (6/9), red nuclei (2/9), and abnormal myelination of pyramidal and visual tracts (6/9) but no frank hypomyelination. Clinical and MRI findings in patients with a striatal variant of POLR3A-related disease are distinct from 4H leukodystrophy and associated with one of two intronic variants, c.1771-6C > G or c.1771-7C > G, in combination with another POLR3A variant.
Collapse
|
97
|
Abstract
Supplemental Digital Content is available in the text
Collapse
|
98
|
Han JY, Kim SY, Cheon JE, Choi M, Lee JS, Chae JH. A Familial Case of Childhood Ataxia with Leukodystrophy Due to Novel POLR1C Mutations. J Clin Neurol 2020; 16:338-340. [PMID: 32319256 PMCID: PMC7174127 DOI: 10.3988/jcn.2020.16.2.338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 11/17/2022] Open
Affiliation(s)
- Ji Yeon Han
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Yeon Kim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Eun Cheon
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Jin Sook Lee
- Department of Pediatrics, Gachon Institute of Genome Medicine and Science, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea.
| | - Jong Hee Chae
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
99
|
Neocleous V, Fanis P, Toumba M, Tanteles GA, Schiza M, Cinarli F, Nicolaides NC, Oulas A, Spyrou GM, Mantzoros CS, Vlachakis D, Skordis N, Phylactou LA. GnRH Deficient Patients With Congenital Hypogonadotropic Hypogonadism: Novel Genetic Findings in ANOS1, RNF216, WDR11, FGFR1, CHD7, and POLR3A Genes in a Case Series and Review of the Literature. Front Endocrinol (Lausanne) 2020; 11:626. [PMID: 32982993 PMCID: PMC7485345 DOI: 10.3389/fendo.2020.00626] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disease caused by Gonadotropin-Releasing Hormone (GnRH) deficiency. So far a limited number of variants in several genes have been associated with the pathogenesis of the disease. In this original research and review manuscript the retrospective analysis of known variants in ANOS1 (KAL1), RNF216, WDR11, FGFR1, CHD7, and POLR3A genes is described, along with novel variants identified in patients with CHH by the present study. Methods: Seven GnRH deficient unrelated Cypriot patients underwent whole exome sequencing (WES) by Next Generation Sequencing (NGS). The identified novel variants were initially examined by in silico computational algorithms and structural analysis of their predicted pathogenicity at the protein level was confirmed. Results: In four non-related GnRH males, a novel X-linked pathogenic variant in ANOS1 gene, two novel autosomal dominant (AD) probably pathogenic variants in WDR11 and FGFR1 genes and one rare AD probably pathogenic variant in CHD7 gene were identified. A rare autosomal recessive (AR) variant in the SRA1 gene was identified in homozygosity in a female patient, whilst two other male patients were also, respectively, found to carry novel or previously reported rare pathogenic variants in more than one genes; FGFR1/POLR3A and SRA1/RNF216. Conclusion: This report embraces the description of novel and previously reported rare pathogenic variants in a series of genes known to be implicated in the biological development of CHH. Notably, patients with CHH can harbor pathogenic rare variants in more than one gene which raises the hypothesis of locus-locus interactions providing evidence for digenic inheritance. The identification of such aberrations by NGS can be very informative for the management and future planning of these patients.
Collapse
Affiliation(s)
- Vassos Neocleous
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Pavlos Fanis
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Meropi Toumba
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Pediatric Endocrine Clinic, IASIS Hospital, Paphos, Cyprus
| | - George A. Tanteles
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Clinical Genetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Melpo Schiza
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Feride Cinarli
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Nicolas C. Nicolaides
- Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, “Aghia Sophia” Childrens Hospital, Athens, Greece
- Division of Endocrinology and Metabolism, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Anastasis Oulas
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Bioinformatics ERA Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - George M. Spyrou
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Bioinformatics ERA Chair, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Christos S. Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA, United States
| | - Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Food, Biotechnology and Development, Agricultural University of Athens, Athens, Greece
- Lab of Molecular Endocrinology, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - Nicos Skordis
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Division of Pediatric Endocrinology, Paedi Center for Specialized Pediatrics, Nicosia, Cyprus
- St George's, University of London Medical School at the University of Nicosia, Nicosia, Cyprus
- *Correspondence: Nicos Skordis
| | - Leonidas A. Phylactou
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Leonidas A. Phylactou
| |
Collapse
|
100
|
Whole exome sequencing in a child with acute disseminated encephalomyelitis, optic neuritis, and periodic fever syndrome: a case report. J Med Case Rep 2019; 13:368. [PMID: 31836009 PMCID: PMC6911267 DOI: 10.1186/s13256-019-2305-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 10/28/2019] [Indexed: 01/17/2023] Open
Abstract
Background Acute disseminated encephalomyelitis is generally preceded by an infection, and it is usually self-limiting and non-recurrent. However, when there are multiple attacks of acute disseminated encephalomyelitis followed by optic neuritis, it is defined as acute disseminated encephalomyelitis-optic neuritis. To the best of our knowledge, there are no previous reports of acute disseminated encephalomyelitis and optic neuritis preceded by autoinflammation, triggered by periodic fever syndrome. Case summary We report on a case of acute disseminated encephalomyelitis with optic neuritis and periodic fever syndrome in a 12-year-old Ecuadorian Hispanic boy with several relapses over the past 10 years, always preceded by autoinflammatory manifestations and without evidence of infectious processes. Whole exome sequencing was performed, and although the results were not conclusive, we found variants in genes associated with both autoinflammatory (NLRP12) and neurological (POLR3A) phenotypes that could be related to the disease pathogenesis having a polygenic rather than monogenic trait. Conclusion We propose that an autoinflammatory basis should be pursued in patients diagnosed as having acute disseminated encephalomyelitis and no record of infections. Also, we show that our patient had a good response after 1 year of treatment with low doses of intravenous immunoglobulin and colchicine.
Collapse
|