1
|
Al-Saady ML, Galabova H, Schoenmakers DH, Beerepoot S, Lindemans C, van Hasselt PM, van der Knaap MS, Wolf NI, Pouwels PJW. Longitudinal volumetric analysis of gray matter atrophy in metachromatic leukodystrophy. J Inherit Metab Dis 2024; 47:792-804. [PMID: 38430011 DOI: 10.1002/jimd.12725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
Metachromatic leukodystrophy (MLD) is an inherited lysosomal storage disorder characterized by arylsulfatase A (ASA) deficiency, leading to sulfatide accumulation and myelin degeneration in the central nervous system. While primarily considered a white matter (WM) disease, gray matter (GM) is also affected in MLD, and hematopoietic stem cell transplantation (HSCT) may have limited effect on GM atrophy. We cross-sectionally and longitudinally studied GM volumes using volumetric MRI in a cohort of 36 (late-infantile, juvenile and adult type) MLD patients containing untreated and HSCT treated subjects. Cerebrum, cortical GM, (total) CSF, cerebellum, deep gray matter (DGM) (excluding thalamus) and thalamus volumes were analyzed. Longitudinal correlations with measures of cognitive and motor functioning were assessed. Cross-sectionally, juvenile and adult type patients (infantiles excluded based on limited numbers) were compared with controls at earliest scan, before possible treatment. Patients had lower cerebrum, cortical GM, DGM and thalamus volumes. Differences were most pronounced for adult type patients. Longitudinal analyses showed substantial and progressive atrophy of all regions and increase of CSF in untreated patients. Similar, albeit less pronounced, effects were seen in treated patients for cerebrum, cortical GM, CSF and thalamus volumes. Deterioration in motor performance (all patients) was related to atrophy, and increase of CSF, in all regions. Cognitive functioning (data available for treated patients) was related to cerebral, cortical GM and thalamus atrophy; and to CSF increase. Our findings illustrate the importance of recognizing GM pathology as a potentially substantial, clinically relevant part of MLD, apparently less amenable to treatment.
Collapse
Affiliation(s)
- Murtadha L Al-Saady
- Department of Pediatric Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, The Netherlands
| | - Hristina Galabova
- Department of Pediatric Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, The Netherlands
| | - Daphne H Schoenmakers
- Department of Pediatric Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, The Netherlands
- Medicine for Society, Platform at Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Shanice Beerepoot
- Department of Pediatric Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Nierkens and Lindemans group, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Caroline Lindemans
- Pediatric blood and Bone marrow transplantation, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Peter M van Hasselt
- Department of Metabolic Diseases, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marjo S van der Knaap
- Department of Pediatric Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, The Netherlands
| | - Nicole I Wolf
- Department of Pediatric Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, The Netherlands
| | - Petra J W Pouwels
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Vrije Universiteit & Universiteit van Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Groeschel S, Beerepoot S, Amedick LB, Krӓgeloh-Mann I, Li J, Whiteman DAH, Wolf NI, Port JD. The effect of intrathecal recombinant arylsulfatase A therapy on structural brain magnetic resonance imaging in children with metachromatic leukodystrophy. J Inherit Metab Dis 2024; 47:778-791. [PMID: 38321717 DOI: 10.1002/jimd.12706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 02/08/2024]
Abstract
This study aimed to evaluate the effect of intrathecal (IT) recombinant human arylsulfatase A (rhASA) on magnetic resonance imaging (MRI)-assessed brain tissue changes in children with metachromatic leukodystrophy (MLD). In total, 510 MRI scans were collected from 12 intravenous (IV) rhASA-treated children with MLD, 24 IT rhASA-treated children with MLD, 32 children with untreated MLD, and 156 normally developing children. Linear mixed models were fitted to analyze the time courses of gray matter (GM) volume and fractional anisotropy (FA) in the posterior limb of the internal capsule. Time courses for demyelination load and FA in the centrum semiovale were visualized using locally estimated scatterplot smoothing regression curves. All assessed imaging parameters demonstrated structural evidence of neurological deterioration in children with MLD. GM volume was significantly lower at follow-up (median duration, 104 weeks) in IV rhASA-treated versus IT rhASA-treated children. GM volume decline over time was steeper in children receiving low-dose (10 or 30 mg) versus high-dose (100 mg) IT rhASA. Similar effects were observed for demyelination. FA in the posterior limb of the internal capsule showed a higher trend over time in IT rhASA-treated versus children with untreated MLD, but FA parameters were not different between children receiving the low doses versus those receiving the high dose. GM volume in IT rhASA-treated children showed a strong positive correlation with 88-item Gross Motor Function Measure score over time. In some children with MLD, IT administration of high-dose rhASA may delay neurological deterioration (assessed using MRI), offering potential therapeutic benefit.
Collapse
Affiliation(s)
- Samuel Groeschel
- Department of Pediatric Neurology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Shanice Beerepoot
- Amsterdam Leukodystrophy Center, Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience-Cellular and Molecular Mechanisms, Vrije Universiteit, Amsterdam, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Lucas Bastian Amedick
- Department of Pediatric Neurology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Ingeborg Krӓgeloh-Mann
- Department of Pediatric Neurology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Jing Li
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| | | | - Nicole I Wolf
- Amsterdam Leukodystrophy Center, Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience-Cellular and Molecular Mechanisms, Vrije Universiteit, Amsterdam, Netherlands
| | - John D Port
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
3
|
Chang SC, Eichinger CS, Field P. The natural history and burden of illness of metachromatic leukodystrophy: a systematic literature review. Eur J Med Res 2024; 29:181. [PMID: 38494502 PMCID: PMC10946116 DOI: 10.1186/s40001-024-01771-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Metachromatic leukodystrophy (MLD; OMIM 250100 and 249900) is a rare lysosomal storage disease caused by deficient arylsulfatase A activity, leading to accumulation of sulfatides in the nervous system. This systematic literature review aimed to explore the effect of MLD on the lives of patients. METHODS The Ovid platform was used to search Embase, MEDLINE, and the Cochrane Library for articles related to the natural history, clinical outcomes, and burden of illness of MLD; congress and hand searches were performed using 'metachromatic leukodystrophy' as a keyword. Of the 531 publications identified, 120 were included for data extraction following screening. A subset of findings from studies relating to MLD natural history and burden of illness (n = 108) are presented here. RESULTS The mean age at symptom onset was generally 16-18 months for late-infantile MLD and 6-10 years for juvenile MLD. Age at diagnosis and time to diagnosis varied widely. Typically, patients with late-infantile MLD presented predominantly with motor symptoms and developmental delay; patients with juvenile MLD presented with motor, cognitive, and behavioral symptoms; and patients with adult MLD presented with cognitive symptoms and psychiatric and mood disorders. Patients with late-infantile MLD had more rapid decline of motor function over time and lower survival than patients with juvenile MLD. Commonly reported comorbidities/complications included ataxia, epilepsy, gallbladder abnormalities, incontinence, neuropathy, and seizures. CONCLUSIONS Epidemiology of MLD by geographic regions, quantitative cognitive data, data on the differences between early- and late-juvenile MLD, and humanistic or economic outcomes were limited. Further studies on clinical, humanistic (i.e., quality of life), and economic outcomes are needed to help inform healthcare decisions for patients with MLD.
Collapse
Affiliation(s)
- Shun-Chiao Chang
- Takeda Development Center Americas, Inc., 125 Binney Street, Cambridge, MA, USA.
| | | | | |
Collapse
|
4
|
Laugwitz L, Schoenmakers DH, Adang LA, Beck-Woedl S, Bergner C, Bernard G, Bley A, Boyer A, Calbi V, Dekker H, Eichler F, Eklund E, Fumagalli F, Gavazzi F, Grønborg SW, van Hasselt P, Langeveld M, Lindemans C, Mochel F, Oberg A, Ram D, Saunier-Vivar E, Schöls L, Scholz M, Sevin C, Zerem A, Wolf NI, Groeschel S. Newborn screening in metachromatic leukodystrophy - European consensus-based recommendations on clinical management. Eur J Paediatr Neurol 2024; 49:141-154. [PMID: 38554683 DOI: 10.1016/j.ejpn.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 04/02/2024]
Abstract
INTRODUCTION Metachromatic leukodystrophy (MLD) is a rare autosomal recessive lysosomal storage disorder resulting from arylsulfatase A enzyme deficiency, leading to toxic sulfatide accumulation. As a result affected individuals exhibit progressive neurodegeneration. Treatments such as hematopoietic stem cell transplantation (HSCT) and gene therapy are effective when administered pre-symptomatically. Newborn screening (NBS) for MLD has recently been shown to be technically feasible and is indicated because of available treatment options. However, there is a lack of guidance on how to monitor and manage identified cases. This study aims to establish consensus among international experts in MLD and patient advocates on clinical management for NBS-identified MLD cases. METHODS A real-time Delphi procedure using eDELPHI software with 22 experts in MLD was performed. Questions, based on a literature review and workshops, were answered during a seven-week period. Three levels of consensus were defined: A) 100%, B) 75-99%, and C) 50-74% or >75% but >25% neutral votes. Recommendations were categorized by agreement level, from strongly recommended to suggested. Patient advocates participated in discussions and were involved in the final consensus. RESULTS The study presents 57 statements guiding clinical management of NBS-identified MLD patients. Key recommendations include timely communication by MLD experts with identified families, treating early-onset MLD with gene therapy and late-onset MLD with HSCT, as well as pre-treatment monitoring schemes. Specific knowledge gaps were identified, urging prioritized research for future evidence-based guidelines. DISCUSSION Consensus-based recommendations for NBS in MLD will enhance harmonized management and facilitate integration in national screening programs. Structured data collection and monitoring of screening programs are crucial for evidence generation and future guideline development. Involving patient representatives in the development of recommendations seems essential for NBS programs.
Collapse
Affiliation(s)
- Lucia Laugwitz
- Neuropediatrics, General Pediatrics, Diabetology, Endocrinology and Social Pediatrics, University of Tuebingen, University Hospital Tübingen, 72016, Tübingen, Germany; Institute for Medical Genetics and Applied Genomics, University of Tübingen, 72070, Tübingen, Germany.
| | - Daphne H Schoenmakers
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC Location Vrije Universiteit, Amsterdam, the Netherlands; Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, the Netherlands; Medicine for Society, Platform at Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
| | - Laura A Adang
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stefanie Beck-Woedl
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, 72070, Tübingen, Germany
| | - Caroline Bergner
- Leukodystrophy Center, Departement of Neurology, University Hospital Leipzig, Germany
| | - Geneviève Bernard
- Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University, Montreal, Canada; Department Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | | | | | - Valeria Calbi
- Pediatric Immuno-Hematology Unit, Ospedale San Raffaele Milan, Italy; San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy
| | - Hanka Dekker
- Dutch Association for Inherited Metabolic Diseases (VKS), the Netherlands
| | | | - Erik Eklund
- Pediatrics, Clinical Sciences, Lund University, Sweden
| | - Francesca Fumagalli
- Pediatric Immuno-Hematology Unit, Ospedale San Raffaele Milan, Italy; San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy; Unit of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Gavazzi
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sabine W Grønborg
- Center for Inherited Metabolic Diseases, Department of Pediatrics and Adolescent Medicine and Department of Clinical Genetics, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Peter van Hasselt
- Department of Metabolic Diseases, University Medical Center Utrecht, the Netherlands
| | - Mirjam Langeveld
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, University of Amsterdam, Amsterdam, the Netherlands
| | - Caroline Lindemans
- Department of Pediatric Hematopoietic Stem Cell Transplantation, UMC Utrecht and Princess Maxima Center, the Netherlands
| | - Fanny Mochel
- Reference Center for Adult Leukodystrophy, Department of Medical Genetics, Sorbonne University, Paris Brain Institute, La Pitié-Salpêtrière University Hospital, Paris, France
| | - Andreas Oberg
- Norwegian National Unit for Newborn Screening, Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Norway
| | - Dipak Ram
- Department of Paediatric Neurology, Royal Manchester Children's Hospital, Manchester, UK
| | | | - Ludger Schöls
- Department of Neurology and Hertie-Institute for Clinical Brain Research, German Center of Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | | | - Ayelet Zerem
- Pediatric Neurology Institute, Leukodystrophy Center, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nicole I Wolf
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC Location Vrije Universiteit, Amsterdam, the Netherlands; Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, the Netherlands
| | - Samuel Groeschel
- Neuropediatrics, General Pediatrics, Diabetology, Endocrinology and Social Pediatrics, University of Tuebingen, University Hospital Tübingen, 72016, Tübingen, Germany
| |
Collapse
|
5
|
Farah MH, Dali CÍ, Groeschel S, Moldovan M, Whiteman DAH, Malanga CJ, Krägeloh‐Mann I, Li J, Barton N, Krarup C. Effects of sulfatide on peripheral nerves in metachromatic leukodystrophy. Ann Clin Transl Neurol 2024; 11:328-341. [PMID: 38146590 PMCID: PMC10863914 DOI: 10.1002/acn3.51954] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 12/27/2023] Open
Abstract
OBJECTIVE To evaluate the longitudinal correlations between sulfatide/lysosulfatide levels and central and peripheral nervous system function in children with metachromatic leukodystrophy (MLD) and to explore the impact of intravenous recombinant human arylsulfatase A (rhASA) treatment on myelin turnover. METHODS A Phase 1/2 study of intravenous rhASA investigated cerebrospinal fluid (CSF) and sural nerve sulfatide levels, 88-item Gross Motor Function Measure (GMFM-88) total score, sensory and motor nerve conduction, brain N-acetylaspartate (NAA) levels, and sural nerve histology in 13 children with MLD. Myelinated and unmyelinated nerves from an untreated MLD mouse model were also analyzed. RESULTS CSF sulfatide levels correlated with neither Z-scores for GMFM-88 nor brain NAA levels; however, CSF sulfatide levels correlated negatively with Z-scores of nerve conduction parameters, number of large (≥7 μm) myelinated fibers, and myelin/fiber diameter slope, and positively with nerve g-ratios and cortical latencies of somatosensory-evoked potentials. Quantity of endoneural litter positively correlated with sural nerve sulfatide/lysosulfatide levels. CSF sulfatide levels decreased with continuous high-dose treatment; this change correlated with improved nerve conduction. At 26 weeks after treatment, nerve g-ratio decreased by 2%, and inclusion bodies per Schwann cell unit increased by 55%. In mice, abnormal sulfatide storage was observed in non-myelinating Schwann cells in Remak bundles of sciatic nerves but not in unmyelinated urethral nerves. INTERPRETATION Lower sulfatide levels in the CSF and peripheral nerves correlate with better peripheral nerve function in children with MLD; intravenous rhASA treatment may reduce CSF sulfatide levels and enhance sulfatide/lysosulfatide processing and remyelination in peripheral nerves.
Collapse
Affiliation(s)
- Mohamed H. Farah
- Department of NeurologyJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Christine í Dali
- Department of Clinical GeneticsRigshospitaletCopenhagenDenmark
- Present address:
Zevra Denmark A/S
| | - Samuel Groeschel
- Department of Pediatric NeurologyUniversity Children's Hospital TübingenTübingenGermany
| | - Mihai Moldovan
- Department of Clinical NeurophysiologyRigshospitaletCopenhagenDenmark
- Department of NeuroscienceUniversity of CopenhagenCopenhagenDenmark
| | | | - C. J. Malanga
- Takeda Development Center Americas, Inc.LexingtonMassachusettsUSA
| | | | - Jing Li
- Takeda Development Center Americas, Inc.LexingtonMassachusettsUSA
| | - Norman Barton
- Takeda Development Center Americas, Inc.LexingtonMassachusettsUSA
| | - Christian Krarup
- Department of Clinical NeurophysiologyRigshospitaletCopenhagenDenmark
- Department of NeuroscienceUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
6
|
Shim G, Romero-Morales AI, Sripathy SR, Maher BJ. Utilizing hiPSC-derived oligodendrocytes to study myelin pathophysiology in neuropsychiatric and neurodegenerative disorders. Front Cell Neurosci 2024; 17:1322813. [PMID: 38273973 PMCID: PMC10808804 DOI: 10.3389/fncel.2023.1322813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Oligodendrocytes play a crucial role in our central nervous system (CNS) by myelinating axons for faster action potential conduction, protecting axons from degeneration, structuring the position of ion channels, and providing nutrients to neurons. Oligodendrocyte dysfunction and/or dysmyelination can contribute to a range of neurodegenerative diseases and neuropsychiatric disorders such as Multiple Sclerosis (MS), Leukodystrophy (LD), Schizophrenia (SCZ), and Autism Spectrum Disorder (ASD). Common characteristics identified across these disorders were either an inability of oligodendrocytes to remyelinate after degeneration or defects in oligodendrocyte development and maturation. Unfortunately, the causal mechanisms of oligodendrocyte dysfunction are still uncertain, and therapeutic targets remain elusive. Many studies rely on the use of animal models to identify the molecular and cellular mechanisms behind these disorders, however, such studies face species-specific challenges and therefore lack translatability. The use of human induced pluripotent stem cells (hiPSCs) to model neurological diseases is becoming a powerful new tool, improving our understanding of pathophysiology and capacity to explore therapeutic targets. Here, we focus on the application of hiPSC-derived oligodendrocyte model systems to model disorders caused by oligodendrocyte dysregulation.
Collapse
Affiliation(s)
- Gina Shim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, United States
| | - Alejandra I. Romero-Morales
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, United States
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Srinidhi R. Sripathy
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, United States
| | - Brady J. Maher
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, United States
- The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
7
|
Al‐Saady M, Beerepoot S, Plug BC, Breur M, Galabova H, Pouwels PJW, Boelens J, Lindemans C, van Hasselt PM, Matzner U, Vanderver A, Bugiani M, van der Knaap MS, Wolf NI. Neurodegenerative disease after hematopoietic stem cell transplantation in metachromatic leukodystrophy. Ann Clin Transl Neurol 2023; 10:1146-1159. [PMID: 37212343 PMCID: PMC10351661 DOI: 10.1002/acn3.51796] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/23/2023] Open
Abstract
OBJECTIVE Metachromatic leukodystrophy is a lysosomal storage disease caused by deficient arylsulfatase A. It is characterized by progressive demyelination and thus mainly affects the white matter. Hematopoietic stem cell transplantation may stabilize and improve white matter damage, yet some patients deteriorate despite successfully treated leukodystrophy. We hypothesized that post-treatment decline in metachromatic leukodystrophy might be caused by gray matter pathology. METHODS Three metachromatic leukodystrophy patients treated with hematopoietic stem cell transplantation with a progressive clinical course despite stable white matter pathology were clinically and radiologically analyzed. Longitudinal volumetric MRI was used to quantify atrophy. We also examined histopathology in three other patients deceased after treatment and compared them with six untreated patients. RESULTS The three clinically progressive patients developed cognitive and motor deterioration after transplantation, despite stable mild white matter abnormalities on MRI. Volumetric MRI identified cerebral and thalamus atrophy in these patients, and cerebellar atrophy in two. Histopathology showed that in brain tissue of transplanted patients, arylsulfatase A expressing macrophages were clearly present in the white matter, but absent in the cortex. Arylsulfatase A expression within patient thalamic neurons was lower than in controls, the same was found in transplanted patients. INTERPRETATION Neurological deterioration may occur after hematopoietic stem cell transplantation in metachromatic leukodystrophy despite successfully treated leukodystrophy. MRI shows gray matter atrophy, and histological data demonstrate absence of donor cells in gray matter structures. These findings point to a clinically relevant gray matter component of metachromatic leukodystrophy, which does not seem sufficiently affected by transplantation.
Collapse
Affiliation(s)
- Murtadha Al‐Saady
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
| | - Shanice Beerepoot
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
- Center for Translational ImmunologyUniversity Medical Center UtrechtUtrechtthe Netherlands
- Nierkens and Lindemans GroupPrincess Máxima Center for Pediatric OncologyUtrechtthe Netherlands
| | - Bonnie C. Plug
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
- Department of Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical CentersVU University and Neuroscience Campus AmsterdamAmsterdamthe Netherlands
| | - Marjolein Breur
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
- Department of Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical CentersVU University and Neuroscience Campus AmsterdamAmsterdamthe Netherlands
| | - Hristina Galabova
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, Amsterdam University Medical CentersVU universityAmsterdamthe Netherlands
| | - Petra J. W. Pouwels
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, Amsterdam University Medical CentersVU universityAmsterdamthe Netherlands
| | - Jaap‐Jan Boelens
- Stem Cell Transplantation and Cellular Therapies Program, Department of PediatricsMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Caroline Lindemans
- Stem Cell Transplantation and Cellular Therapies Program, Department of PediatricsMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
- Pediatric Blood and Bone Marrow Transplantation, Princess Máxima Center for Pediatric OncologyUtrechtthe Netherlands
| | - Peter M. van Hasselt
- Stem Cell Transplantation and Cellular Therapies Program, Department of PediatricsMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Ulrich Matzner
- Institute of Biochemistry and Molecular Biology, Medical FacultyRheinische Friedrich‐Wilhelm UniversityBonnGermany
| | - Adeline Vanderver
- Division of Neurology, Department of Pediatrics, Children's Hospital of PhiladelphiaUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Marianna Bugiani
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
- Department of Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical CentersVU University and Neuroscience Campus AmsterdamAmsterdamthe Netherlands
| | - Marjo S. van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
- Department of Integrative NeurophysiologyVU UniversityAmsterdamthe Netherlands
| | - Nicole I. Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular MechanismsVrije UniversiteitAmsterdamthe Netherlands
| |
Collapse
|
8
|
Feldmann J, Martin P, Bender B, Laugwitz L, Zizmare L, Trautwein C, Krägeloh-Mann I, Klose U, Groeschel S. MR-spectroscopy in metachromatic leukodystrophy: A model free approach and clinical correlation. Neuroimage Clin 2023; 37:103296. [PMID: 36563646 PMCID: PMC9800432 DOI: 10.1016/j.nicl.2022.103296] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/23/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Metachromatic leukodystrophy (MLD) is a lysosomal enzyme deficiency disorder leading to demyelination and subsequently to a progressive decline in cognitive and motor function. It affects mainly white matter where changes during the course of the disease can be visualized on T2-weighted MRI as hyperintense areas. Associated changes in brain metabolism can be quantified by MR spectroscopy (MRS) and may give complementary information as biomarkers for disease characterisation and progression. Our study aimed to further investigate the correlation of MRS with clinical parameters for motor and cognitive function by using a model free MRS analysis approach that would be precise and straightforward to implement. MATERIALS AND METHODS 53 MRS datasets derived from 29 patients (10 late-infantile, 19 juvenile) and 12 controls were acquired using a semi-LASER CSI sequence covering a slice through the centrum semiovale above the corpus callosum. We defined four regions of interest in the white matter (frontal white matter [FWM] and the cortico-spinal tract [CST] area, each left and right) and one in cortical grey matter. Spectra were analysed using a model and fitting free approach by calculating the definite integral of 10 intervals which were distributed along the whole spectrum. These 10 intervals were orientated towards the main peaks of the metabolites N-acetylaspartate (NAA), creatine, myo-inositol, choline, glutamine/glutamate and aspartate to approximately attribute changes in the intervals to corresponding metabolites. Their ratios to the main creatine peak integral were correlated with clinical parameters assessing motor and cognitive abilities. Furthermore, in a post-hoc analysis, NAA levels of a subset of 21 MR datasets were correlated to NAA levels in urine measured by 1H (proton) nuclear magnetic resonance (NMR) spectroscopy. The applied interval integration method was validated in the control cohort against the standard approach, using spectral profile templates of known metabolites (LCModel). Both methods showed good agreement, with coefficients of variance being slightly lower for our approach compared to the related LCModel results. Moreover, the new approach was able to extract information out of the frequency range around the main peaks of aspartate and glutamine where LCModel showed only few usable values for the respective metabolites. RESULTS MLD spectra clearly differed from controls. The most pronounced differences were found in white matter (much less in grey matter), with larger values corresponding to main peaks of myo-inositol, choline and aspartate, and smaller values associated with NAA and glutamine. Late-infantile patients had more severe changes compared to later-onset patients, especially in intervals corresponding to NAA, aspartate, myo-inositol, choline and glutamine. There was a high correlation of several intervals in the corticospinal tract region with motor function (with the most relevant interval corresponding to NAA peak with a correlation coefficient of -0.75; p < 0.001), while cognitive function, by means of IQ, was found to be most correlating in frontal white matter corresponding to the NAA peak (r = 0.84, p < 0.001). The post-hoc analysis showed that the main NAA peak interval correlated negatively with the NAA in urine (r = -0.584, p < 0.001). CONCLUSION The applied model and fitting free interval integration approach to analyse MRS data of a semi-LASER sequence at 3T suits well to detect and quantify pathological changes in MLD patients through the different courses of the disease and correlates well with clinical symptoms while showing smaller dimensions of variation compared to the more sophisticated single metabolite analysis using LCModel. NAA seems the most clinically meaningful biomarker to use in this context. Its correlation with urine measurements further underlines its potential as a clinically and biologically useful parameter of disease progression in MLD.
Collapse
Affiliation(s)
- Joana Feldmann
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, University of Tübingen, 72076 Tübingen, Germany
| | - Pascal Martin
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany.
| | - Benjamin Bender
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Lucia Laugwitz
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, University of Tübingen, 72076 Tübingen, Germany
| | - Laimdota Zizmare
- Werner Siemens Imaging Center, University of Tübingen, 72076 Tübingen, Germany
| | - Christoph Trautwein
- Werner Siemens Imaging Center, University of Tübingen, 72076 Tübingen, Germany
| | - Ingeborg Krägeloh-Mann
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, University of Tübingen, 72076 Tübingen, Germany
| | - Uwe Klose
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Samuel Groeschel
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
9
|
Schoenmakers DH, Beerepoot S, Krägeloh‐Mann I, Elgün S, Bender B, van der Knaap MS, Wolf NI, Groeschel S. Recognizing early MRI signs (or their absence) is crucial in diagnosing metachromatic leukodystrophy. Ann Clin Transl Neurol 2022; 9:1999-2009. [PMID: 36334091 PMCID: PMC9735365 DOI: 10.1002/acn3.51692] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES Metachromatic leukodystrophy (MLD) has characteristic white matter (WM) changes on brain MRI, which often trigger biochemical and genetic confirmation of the diagnosis. In early or pre-symptomatic disease stages, these typical MRI changes might be absent, hampering early diagnosis. This study aims to describe the characteristics of MRI WM abnormalities at diagnosis, related to clinical presentation. METHODS We retrospectively reviewed brain MRIs of MLD patients followed in 2 centers at the time of diagnosis regarding MLD MRI score and presence of tigroid pattern. In addition, MLD subtype, symptom status, CNS/PNS phenotype, motor/cognitive/mixed phenotype, and the presence of CNS symptoms were evaluated. RESULTS We included 104 brain MRIs from patients with late-infantile (n = 43), early-juvenile (n = 24), late-juvenile (n = 20) and adult (n = 17) onset. Involvement of the corpus callosum was a characteristic early MRI sign and was present in 71% of the symptomatic late-infantile patients, 94% of the symptomatic early-juvenile patients and 100% of the symptomatic late-juvenile and adult patients. Symptomatic early-juvenile, late-juvenile and adult patients generally had WM abnormalities on MRI suggestive of MLD. By contrast, 47% of the early-symptomatic late-infantile patients had no or only mild WM abnormalities on MRI, even in the presence of CNS symptoms including pyramidal signs. INTERPRETATION Patients with late-infantile MLD may have no or only mild, nonspecific abnormalities at brain MRI, partly suggestive of 'delayed myelination', even with clear clinical symptoms. This may lead to significant diagnostic delay. Knowledge of these early MRI signs (or their absence) is important for fast diagnosis.
Collapse
Affiliation(s)
- Daphne H. Schoenmakers
- Department of Child Neurology, Amsterdam Leukodystrophy CenterAmsterdam UMC location Vrije Universiteit Amsterdam, Emma's Children's HospitalBoelelaan 1117AmsterdamThe Netherlands,Amsterdam Neuroscience, Cellular & Molecular MechanismsAmsterdamThe Netherlands,Department of Endocrinology and MetabolismAmsterdam UMC location University of AmsterdamMeibergdreef 9AmsterdamThe Netherlands
| | - Shanice Beerepoot
- Department of Child Neurology, Amsterdam Leukodystrophy CenterAmsterdam UMC location Vrije Universiteit Amsterdam, Emma's Children's HospitalBoelelaan 1117AmsterdamThe Netherlands,Amsterdam Neuroscience, Cellular & Molecular MechanismsAmsterdamThe Netherlands,Center for Translational ImmunologyUniversity Medical Center UtrechtUtrechtThe Netherlands,Pediatric Transplant CenterPrincess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Ingeborg Krägeloh‐Mann
- Department of Child Neurology and Developmental MedicineUniversity Children's Hospital TübingenHoppe‐Seyler‐Straße 172076TübingenGermany
| | - Saskia Elgün
- Department of Child Neurology and Developmental MedicineUniversity Children's Hospital TübingenHoppe‐Seyler‐Straße 172076TübingenGermany
| | - Benjamin Bender
- Diagnostic and Interventional Neuroradiology, Department of RadiologyUniversity Hospital TübingenHoppe‐Seyler‐Straße 372076TübingenGermany
| | - Marjo S. van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy CenterAmsterdam UMC location Vrije Universiteit Amsterdam, Emma's Children's HospitalBoelelaan 1117AmsterdamThe Netherlands,Amsterdam Neuroscience, Cellular & Molecular MechanismsAmsterdamThe Netherlands,Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive ResearchVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Nicole I. Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy CenterAmsterdam UMC location Vrije Universiteit Amsterdam, Emma's Children's HospitalBoelelaan 1117AmsterdamThe Netherlands,Amsterdam Neuroscience, Cellular & Molecular MechanismsAmsterdamThe Netherlands
| | - Samuel Groeschel
- Department of Child Neurology and Developmental MedicineUniversity Children's Hospital TübingenHoppe‐Seyler‐Straße 172076TübingenGermany
| |
Collapse
|
10
|
Hossain MA, Hasegawa-Ogawa M, Manome Y, Igarashi M, Wu C, Suzuki K, Igarashi J, Iwamoto T, Okano HJ, Eto Y. Generation and characterization of motor neuron progenitors and motor neurons using metachromatic leukodystrophy-induced pluripotent stem cells. Mol Genet Metab Rep 2022; 31:100852. [PMID: 35782608 PMCID: PMC9248224 DOI: 10.1016/j.ymgmr.2022.100852] [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: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 10/29/2022] Open
|
11
|
Bae I, Chae JH, Han Y. A brain extraction algorithm for infant T2 weighted magnetic resonance images based on fuzzy c-means thresholding. Sci Rep 2021; 11:23347. [PMID: 34857824 PMCID: PMC8640033 DOI: 10.1038/s41598-021-02722-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 11/22/2021] [Indexed: 11/28/2022] Open
Abstract
It is challenging to extract the brain region from T2-weighted magnetic resonance infant brain images because conventional brain segmentation algorithms are generally optimized for adult brain images, which have different spatial resolution, dynamic changes of imaging intensity, brain size and shape from infant brain images. In this study, we propose a brain extraction algorithm for infant T2-weighted images. The proposed method utilizes histogram partitioning to separate brain regions from the background image. Then, fuzzy c-means thresholding is performed to obtain a rough brain mask for each image slice, followed by refinement steps. For slices that contain eye regions, an additional eye removal algorithm is proposed to eliminate eyes from the brain mask. By using the proposed method, accurate masks for infant T2-weighted brain images can be generated. For validation, we applied the proposed algorithm and conventional methods to T2 infant images (0–24 months of age) acquired with 2D and 3D sequences at 3T MRI. The Dice coefficients and Precision scores, which were calculated as quantitative measures, showed the highest values for the proposed method as follows: For images acquired with a 2D imaging sequence, the average Dice coefficients were 0.9650 ± 0.006 for the proposed method, 0.9262 ± 0.006 for iBEAT, and 0.9490 ± 0.006 for BET. For the data acquired with a 3D imaging sequence, the average Dice coefficient was 0.9746 ± 0.008 for the proposed method, 0.9448 ± 0.004 for iBEAT, and 0.9622 ± 0.01 for BET. The average Precision was 0.9638 ± 0.009 and 0.9565 ± 0.016 for the proposed method, 0.8981 ± 0.01 and 0.8968 ± 0.008 for iBEAT, and 0.9346 ± 0.014 and 0.9282 ± 0.019 for BET for images acquired with 2D and 3D imaging sequences, respectively, demonstrating that the proposed method could be efficiently used for brain extraction in T2-weighted infant images.
Collapse
Affiliation(s)
- Inyoung Bae
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, Republic of Korea
| | - Jong-Hee Chae
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yeji Han
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, Republic of Korea. .,Department of Biomedical Engineering, College of Health Sciences, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
12
|
Fumagalli F, Zambon AA, Rancoita PMV, Baldoli C, Canale S, Spiga I, Medaglini S, Penati R, Facchini M, Ciotti F, Sarzana M, Lorioli L, Cesani M, Natali Sora MG, Del Carro U, Cugnata F, Antonioli G, Recupero S, Calbi V, Di Serio C, Aiuti A, Biffi A, Sessa M. Metachromatic leukodystrophy: A single-center longitudinal study of 45 patients. J Inherit Metab Dis 2021; 44:1151-1164. [PMID: 33855715 DOI: 10.1002/jimd.12388] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/24/2021] [Accepted: 04/12/2021] [Indexed: 11/10/2022]
Abstract
In this study, we characterize the natural course of metachromatic leukodystrophy (MLD), explore intra/inter group differences, and identify biomarkers to monitor disease progression. This is a longitudinal observational study. Genotype and characteristics at disease onset were recorded. Time-to-event analyses were performed to assess time to major disease-related milestones in different subgroups. Longitudinal trajectories of nerve conduction velocities (NCV), brain MRI score, and brainstem auditory evoked responses (BAERs) were described. We recruited 22 late-infantile, 14 early-juvenile, 5 late-juvenile, and 4 adult MLD patients. Thirty-four were prospectively evaluated (median FU time 43 months). In late-infantile patients, the attainment of independent walking was associated with a later age at dysphagia. In early-juvenile, the presence of isolated cognitive impairment at onset was not a favorable prognostic factor. Late-infantile and early-juvenile subjects showed similar rapid loss of ambulation and onset of seizures, but late-infantile displayed earlier loss of trunk control, dysphagia, and death. We found significant differences in all major disease-related milestones (except death) between early-juvenile and late-juvenile patients. Late-juvenile and adult patients both presented with a predominant cognitive impairment, mild/no peripheral neuropathy, lower brain MRI score at plateau compared to LI/EJ, and later cerebellar involvement. NCV and BAER were consistently severely abnormal in late-infantile but not in older subjects, in whom both NCV and BAER were variably affected, with no deterioration over time in some cases. This study clarifies intra/inter group differences between MLD subtypes and provides additional indications regarding reliable clinical and instrumental tools to monitor disease progression and to serve as areference to evaluate the efficacy of future therapeutic interventions inthe different MLD variants.
Collapse
Affiliation(s)
- Francesca Fumagalli
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto A Zambon
- Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Paola M V Rancoita
- University Centre of Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan, Italy
| | - Cristina Baldoli
- Neuroradiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sabrina Canale
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Specialistic Neurological Rehabilitation, IRCCS Multimedica, Sesto San Giovanni, Italy
| | - Ivana Spiga
- Clinical Molecular Biology Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefania Medaglini
- Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rachele Penati
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Marcella Facchini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Ciotti
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marina Sarzana
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Lorioli
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Cesani
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- AGC Biologics S.p.a, Bresso (MI), Italy
| | | | - Ubaldo Del Carro
- Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Cugnata
- University Centre of Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan, Italy
| | - Gigliola Antonioli
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Salvatore Recupero
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Valeria Calbi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Clelia Di Serio
- University Centre of Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandra Biffi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Department of Women and Child Health, University of Padova, Padova, Italy
| | - Maria Sessa
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Neurology, ASST Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
13
|
Beck‐Wödl S, Kehrer C, Harzer K, Haack TB, Bürger F, Haas D, Rieß A, Groeschel S, Krägeloh‐Mann I, Böhringer J. Long-term disease course of two patients with multiple sulfatase deficiency differs from metachromatic leukodystrophy in a broad cohort. JIMD Rep 2021; 58:80-88. [PMID: 33728250 PMCID: PMC7932862 DOI: 10.1002/jmd2.12189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/27/2020] [Accepted: 11/12/2020] [Indexed: 12/16/2022] Open
Abstract
Multiple sulfatase deficiency (MSD) is a lysosomal storage disease caused by a deficiency of formylglycine-generating enzyme due to SUMF1 defects. MSD may be misdiagnosed as metachromatic leukodystrophy (MLD), as neurological and neuroimaging findings are similar, and arylsulfatase A (ARSA) deficiency and enhanced urinary sulfatide excretion may also occur. While ARSA deficiency seems a cause for neurological symptoms and later neurodegenerative disease course, deficiency of other sulfatases results in clinical features such as dysmorphism, dysostosis, or ichthyosis. We report on a girl and a boy of the same origin presenting with severe ARSA deficiency and neurological and neuroimaging features compatible with MLD. However, exome sequencing revealed not yet described homozygosity of the missense variant c.529G > C, p.Ala177Pro in SUMF1. We asked whether dynamics of disease course differs between MSD and MLD. Comparison to a cohort of 59 MLD patients revealed different disease course concerning onset and disease progression in both MSD patients. The MSD patients showed first gross motor symptoms earlier than most patients with juvenile MLD (<10th percentile of Gross-Motor-Function in MLD [GMFC-MLD] 1). However, subsequent motor decline was more protracted (75th and 90th percentile of GMFC-MLD 2 (loss of independent walking) and 75th percentile of GMFC-MLD 5 (loss of any locomotion)). Language decline started clearly after 50th percentile of juvenile MLD and progressed rapidly. Thus, dynamics of disease course may be a further clue for the characterization of MSD. These data may contribute to knowledge of natural course of ultra-rare MSD and be relevant for counseling and therapy.
Collapse
Affiliation(s)
- Stefanie Beck‐Wödl
- Institute of Medical Genetics and Applied GenomicsUniversity of TübingenTübingenGermany
| | - Christiane Kehrer
- Department of NeuropediatricsUniversity Children's HospitalTübingenGermany
| | - Klaus Harzer
- Department of NeuropediatricsUniversity Children's HospitalTübingenGermany
| | - Tobias B. Haack
- Institute of Medical Genetics and Applied GenomicsUniversity of TübingenTübingenGermany
| | | | - Dorothea Haas
- Metabolic CentreUniversity Children's HospitalHeidelbergGermany
| | - Angelika Rieß
- Institute of Medical Genetics and Applied GenomicsUniversity of TübingenTübingenGermany
| | - Samuel Groeschel
- Department of NeuropediatricsUniversity Children's HospitalTübingenGermany
| | | | - Judith Böhringer
- Department of NeuropediatricsUniversity Children's HospitalTübingenGermany
| |
Collapse
|
14
|
Í Dali C, Groeschel S, Moldovan M, Farah MH, Krägeloh-Mann I, Wasilewski M, Li J, Barton N, Krarup C. Intravenous arylsulfatase A in metachromatic leukodystrophy: a phase 1/2 study. Ann Clin Transl Neurol 2020; 8:66-80. [PMID: 33332761 PMCID: PMC7818087 DOI: 10.1002/acn3.51254] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/12/2020] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Metachromatic leukodystrophy (MLD) is an autosomal recessive lysosomal storage disease caused by deficient activity of arylsulfatase A (ASA), resulting in severe motor and cognitive dysfunction. This phase 1/2 study evaluated the safety and efficacy of intravenous (IV) recombinant human ASA (rhASA; HGT-1111, previously known as Metazym) in children with MLD. METHODS Thirteen children with MLD (symptom onset < 4 years of age) were enrolled in an open-label, nonrandomized, dose-escalation trial and received IV rhASA at 50, 100, or 200 U/kg body weight every 14 (± 4) days for 52 weeks (NCT00418561; NCT00633139). Eleven children continued to receive rhASA at 100 or 200 U/kg during a 24-month extension period (NCT00681811). Outcome measures included safety observations, changes in motor and cognitive function, and changes in nerve conduction and morphometry. RESULTS There were no serious adverse events considered related to IV rhASA. Motor function and developmental testing scores declined during the study in all dose groups; no significant differences were observed between groups. Nerve conduction studies and morphometric analysis indicated that peripheral nerve pathology did not worsen during the study in any dose group. INTERPRETATION IV rhASA was generally well tolerated. There was no evidence of efficacy in preventing motor and cognitive deterioration, suggesting that IV rhASA may not cross the blood-brain barrier in therapeutic quantities. The relative stability of peripheral nerve function during the study indicates that rhASA may be beneficial if delivered to the appropriate target site and supports the development of rhASA for intrathecal administration in MLD.
Collapse
Affiliation(s)
- Christine Í Dali
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Samuel Groeschel
- Department of Neuropediatrics, University Children's Hospital Tübingen, Tübingen, Germany
| | - Mihai Moldovan
- Department of Clinical Neurophysiology, Rigshospitalet, Copenhagen, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Mohamed H Farah
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ingeborg Krägeloh-Mann
- Department of Neuropediatrics, University Children's Hospital Tübingen, Tübingen, Germany
| | - Margaret Wasilewski
- Shire (a member of the Takeda group of companies), Lexington, Massachusetts, USA
| | - Jing Li
- Shire (a member of the Takeda group of companies), Lexington, Massachusetts, USA
| | - Norman Barton
- Shire (a member of the Takeda group of companies), Lexington, Massachusetts, USA
| | - Christian Krarup
- Department of Clinical Neurophysiology, Rigshospitalet, Copenhagen, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Gowrishankar S, Cologna SM, Givogri MI, Bongarzone ER. Deregulation of signalling in genetic conditions affecting the lysosomal metabolism of cholesterol and galactosyl-sphingolipids. Neurobiol Dis 2020; 146:105142. [PMID: 33080336 PMCID: PMC8862610 DOI: 10.1016/j.nbd.2020.105142] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 09/04/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022] Open
Abstract
The role of lipids in neuroglial function is gaining momentum in part due to a better understanding of how many lipid species contribute to key cellular signalling pathways at the membrane level. The description of lipid rafts as membrane domains composed by defined classes of lipids such as cholesterol and sphingolipids has greatly helped in our understanding of how cellular signalling can be regulated and compartmentalized in neurons and glial cells. Genetic conditions affecting the metabolism of these lipids greatly impact on how some of these signalling pathways work, providing a context to understand the biological function of the lipid. Expectedly, abnormal metabolism of several lipids such as cholesterol and galactosyl-sphingolipids observed in several metabolic conditions involving lysosomal dysfunction are often accompanied by neuronal and myelin dysfunction. This review will discuss the role of lysosomal biology in the context of deficiencies in the metabolism of cholesterol and galactosyl-sphingolipids and their impact on neural function in three genetic disorders: Niemann-Pick type C, Metachromatic leukodystrophy and Krabbe's disease.
Collapse
Affiliation(s)
- S Gowrishankar
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL, USA.
| | - S M Cologna
- Department of Chemistry, University of Illinois, Chicago, IL, USA.
| | - M I Givogri
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL, USA.
| | - E R Bongarzone
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL, USA.
| |
Collapse
|
16
|
Í Dali C, Sevin C, Krägeloh-Mann I, Giugliani R, Sakai N, Wu J, Wasilewski M. Safety of intrathecal delivery of recombinant human arylsulfatase A in children with metachromatic leukodystrophy: Results from a phase 1/2 clinical trial. Mol Genet Metab 2020; 131:235-244. [PMID: 32792226 DOI: 10.1016/j.ymgme.2020.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Metachromatic leukodystrophy (MLD) is an autosomal recessive disorder caused by deficient arylsulfatase A (ASA) activity and characterized by neurological involvement that results in severe disability and premature death. We examined the safety and tolerability of intrathecally delivered recombinant human ASA (rhASA; SHP611, now TAK-611) in children with MLD (NCT01510028). Secondary endpoints included change in cerebrospinal fluid (CSF) sulfatide and lysosulfatide levels, and motor function (assessed by Gross Motor Function Measure-88 total score). METHODS Twenty-four children with MLD who experienced symptom onset aged ≤ 30 months were enrolled. Patients received rhASA every other week (EOW) for 38 weeks at 10, 30, or 100 mg (cohorts 1-3; n = 6 per cohort), or 100 mg manufactured using a revised process (cohort 4; n = 6). RESULTS No rhASA-related serious adverse events (SAEs) were observed; 25% of patients experienced an SAE related to the intrathecal device or drug delivery method. Mean CSF sulfatide and lysosulfatide levels fell to within normal ranges in both 100 mg cohorts following treatment. Although there was a general decline in motor function over time, there was a tendency towards a less pronounced decline in patients receiving 100 mg. CONCLUSION Intrathecal rhASA was generally well tolerated at doses up to 100 mg EOW. These preliminary data support further development of rhASA as a therapy for patients with MLD.
Collapse
Affiliation(s)
- Christine Í Dali
- Department of Clinical Genetics, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.
| | | | - Ingeborg Krägeloh-Mann
- Department of Neuropediatrics, University Children's Hospital Tübingen, Tübingen, Germany.
| | - Roberto Giugliani
- Medical Genetics Service, HCPA, Department of Genetics, UFRGS, and INAGEMP, Porto Alegre, Brazil.
| | | | - James Wu
- Shire, a member of the Takeda group of companies, Lexington, MA, USA.
| | | |
Collapse
|
17
|
Troy S, Wasilewski M, Beusmans J, Godfrey CJ. Pharmacokinetic Modeling of Intrathecally Administered Recombinant Human Arylsulfatase A (TAK-611) in Children With Metachromatic Leukodystrophy. Clin Pharmacol Ther 2020; 107:1394-1404. [PMID: 31868225 PMCID: PMC7325319 DOI: 10.1002/cpt.1752] [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: 07/19/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022]
Abstract
Metachromatic leukodystrophy (MLD) is a lysosomal storage disease caused by deficient arylsulfatase A (ASA) activity, which leads to neuronal sulfatide accumulation and motor and cognitive deterioration. Intrathecal delivery of a recombinant human ASA (TAK-611, formerly SHP611) is under development as a potential therapy for MLD. We used serum and cerebrospinal fluid (CSF) TAK-611 concentrations measured during the phase I/II trial of intrathecal TAK-611 to develop a pharmacokinetic (PK) model describing drug disposition. CSF data were well characterized by a two-compartment model in the central nervous system (CNS); a single central compartment described the serum data. Estimated parameters suggested rapid distribution of TAK-611 from CSF into the putative brain tissue compartment, with persistence in the brain between doses (median distributive and terminal half-lives in the CNS: 1.02 and 477 hours, respectively). This model provides a valuable basis for understanding the PK distribution of TAK-611 and for PK/pharmacodynamic analyses of functional outcomes.
Collapse
Affiliation(s)
- Steven Troy
- Shire (a member of the Takeda group of companies), Lexington, Massachusetts, USA.,Radius Health, Waltham, Massachusetts, USA
| | - Margaret Wasilewski
- Shire (a member of the Takeda group of companies), Lexington, Massachusetts, USA.,ID Remedies LLC, Medford, Massachusetts, USA
| | - Jack Beusmans
- Metrum Research Group, Tariffville, Connecticut, USA
| | - C J Godfrey
- Metrum Research Group, Tariffville, Connecticut, USA
| |
Collapse
|
18
|
Page KM, Stenger EO, Connelly JA, Shyr D, West T, Wood S, Case L, Kester M, Shim S, Hammond L, Hammond M, Webb C, Biffi A, Bambach B, Fatemi A, Kurtzberg J. Hematopoietic Stem Cell Transplantation to Treat Leukodystrophies: Clinical Practice Guidelines from the Hunter's Hope Leukodystrophy Care Network. Biol Blood Marrow Transplant 2019; 25:e363-e374. [PMID: 31499213 DOI: 10.1016/j.bbmt.2019.09.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 08/09/2019] [Accepted: 09/02/2019] [Indexed: 12/22/2022]
Abstract
The leukodystrophies are a heterogeneous group of inherited diseases characterized by progressive demyelination of the central nervous system leading to devastating neurologic symptoms and premature death. Hematopoietic stem cell transplantation (HSCT) has been successfully used to treat certain leukodystrophies, including adrenoleukodystrophy, globoid leukodystrophy (Krabbe disease), and metachromatic leukodystrophy, over the past 30 years. To date, these complex patients have primarily been transplanted at a limited number of pediatric centers. As the number of cases identified through pregnancy and newborn screening is increasing, additional centers will be required to treat these children. Hunter's Hope created the Leukodystrophy Care Network in part to create and standardize high-quality clinical practice guidelines to guide the care of affected patients. In this report the clinical guidelines for the care of pediatric patients with leukodystrophies undergoing treatment with HSCT are presented. The initial transplant evaluation, determination of patient eligibility, donor selection, conditioning, supportive care, and post-transplant follow-up are discussed. Throughout these guidelines the need for early detection and treatment and the role of the partnership between families and multidisciplinary providers are emphasized.
Collapse
Affiliation(s)
- Kristin M Page
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina.
| | - Elizabeth O Stenger
- Aflac Cancer & Blood Disorders Center, Children's Hospital of Atlanta/Emory University
| | - James A Connelly
- Monroe Carell Jr. Children's Hospital at Vanderbilt University, Nashville, Tennessee
| | - David Shyr
- Division of Pediatric Hematology/Oncology, University of Utah School of Medicine
| | - Tara West
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina
| | - Susan Wood
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina
| | - Laura Case
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina
| | - Maureen Kester
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina
| | - Soo Shim
- Ann & Robert H. Lurie Children's Hospital, Chichago, Illinois
| | - Lauren Hammond
- Leukodystrophy Care Network Steering Committee, Orchard Park, New York
| | - Matthew Hammond
- Leukodystrophy Care Network Steering Committee, Orchard Park, New York
| | - Christin Webb
- Leukodystrophy Care Network Steering Committee, Orchard Park, New York
| | - Alessandra Biffi
- Dana Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | | | - Ali Fatemi
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland
| | - Joanne Kurtzberg
- Pediatric Transplant and Cellular Therapy, Duke University, Durham, North Carolina
| |
Collapse
|
19
|
Frati G, Luciani M, Meneghini V, De Cicco S, Ståhlman M, Blomqvist M, Grossi S, Filocamo M, Morena F, Menegon A, Martino S, Gritti A. Human iPSC-based models highlight defective glial and neuronal differentiation from neural progenitor cells in metachromatic leukodystrophy. Cell Death Dis 2018; 9:698. [PMID: 29899471 PMCID: PMC5997994 DOI: 10.1038/s41419-018-0737-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/22/2022]
Abstract
The pathological cascade leading from primary storage to neural cell dysfunction and death in metachromatic leukodystrophy (MLD) has been poorly elucidated in human-derived neural cell systems. In the present study, we have modeled the progression of pathological events during the differentiation of patient-specific iPSCs to neuroepithelial progenitor cells (iPSC-NPCs) and mature neurons, astrocytes, and oligodendrocytes at the morphological, molecular, and biochemical level. We showed significant sulfatide accumulation and altered sulfatide composition during the differentiation of MLD iPSC-NPCs into neuronal and glial cells. Changes in sulfatide levels and composition were accompanied by the expansion of the lysosomal compartment, oxidative stress, and apoptosis. The neuronal and glial differentiation capacity of MLD iPSC-NPCs was significantly impaired. We showed delayed appearance and/or reduced levels of oligodendroglial and astroglial markers as well as reduced number of neurons and disorganized neuronal network. Restoration of a functional Arylsulfatase A (ARSA) enzyme in MLD cells using lentiviral-mediated gene transfer normalized sulfatide levels and composition, globally rescuing the pathological phenotype. Our study points to MLD iPSC-derived neural progeny as a useful in vitro model to assess the impact of ARSA deficiency along NPC differentiation into neurons and glial cells. In addition, iPSC-derived neural cultures allowed testing the impact of ARSA reconstitution/overexpression on disease correction and, importantly, on the biology and functional features of human NPCs, with important therapeutic implications.
Collapse
Affiliation(s)
- Giacomo Frati
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milano, Italy
| | - Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milano, Italy
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milano, Italy.,Institute Imagine, 24 Boulevard du Montparnasse, 75015, Paris, France
| | - Silvia De Cicco
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milano, Italy.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Otfried-Müller Str.23, 72076, Tübingen, Germany
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital, SE-41101, Gothenburg, Sweden
| | - Maria Blomqvist
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-41101, Gothenburg, Sweden
| | - Serena Grossi
- UOSD Centro di diagnostica genetica e biochimica delle malattie metaboliche, IRCCS G. Gaslini Institute, Via G. Gaslini, 16147, Genova, Italy
| | - Mirella Filocamo
- UOSD Centro di diagnostica genetica e biochimica delle malattie metaboliche, IRCCS G. Gaslini Institute, Via G. Gaslini, 16147, Genova, Italy
| | - Francesco Morena
- Department of Chemistry, Biology, and Biotechnologies, University of Perugia, Via del Giochetto, 06126, Perugia, Italy
| | - Andrea Menegon
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sabata Martino
- Department of Chemistry, Biology, and Biotechnologies, University of Perugia, Via del Giochetto, 06126, Perugia, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milano, Italy.
| |
Collapse
|
20
|
Strölin M, Krägeloh-Mann I, Kehrer C, Wilke M, Groeschel S. Demyelination load as predictor for disease progression in juvenile metachromatic leukodystrophy. Ann Clin Transl Neurol 2017; 4:403-410. [PMID: 28589167 PMCID: PMC5454396 DOI: 10.1002/acn3.420] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 11/15/2022] Open
Abstract
Objective The aim of this study was to investigate whether the extent and topography of cerebral demyelination correlates with and predicts disease progression in patients with juvenile metachromatic leukodystrophy (MLD). Methods A total of 137 MRIs of 46 patients with juvenile MLD were analyzed. Demyelination load and brain volume were quantified using the previously developed Software “clusterize.” Clinical data were collected within the German Leukodystrophy Network and included full scale intelligence quotient (FSIQ) and gross motor function data. Voxel‐based lesion‐symptom mapping (VLSM) across the whole brain was performed to investigate the spatial relationship of cerebral demyelination with motor or cognitive function. The prognostic value of the demyelination load at disease onset was assessed to determine the severity of disease progression. Results The demyelination load (corrected by the individual brain volume) correlated significantly with gross motor function (r = +0.55) and FSIQ (r = −0.55). Demyelination load at disease onset was associated with the severity of disease progression later on (P < 0.01). VLSM results associated frontal lobe demyelination with loss in FSIQ and more central region demyelination with decline of motor function. Especially progression of demyelination within the motor area was associated with severe disease progression. Interpretation We were able to show for the first time in a large cohort of patients with juvenile MLD that the demyelination load correlates with motor and cognitive symptoms. Moreover, demyelination load at disease onset, especially the involvement of the central region, predicts severity of disease progression. Thus, demyelination load seems a functionally relevant MRI parameter.
Collapse
Affiliation(s)
- Manuel Strölin
- Department of Pediatric Neurology & Developmental Medicine University Children's Hospital Tübingen Germany
| | - Ingeborg Krägeloh-Mann
- Department of Pediatric Neurology & Developmental Medicine University Children's Hospital Tübingen Germany
| | - Christiane Kehrer
- Department of Pediatric Neurology & Developmental Medicine University Children's Hospital Tübingen Germany
| | - Marko Wilke
- Department of Pediatric Neurology & Developmental Medicine University Children's Hospital Tübingen Germany.,Experimental Pediatric Neuroimaging University Children's Hospital Tübingen Germany
| | - Samuel Groeschel
- Department of Pediatric Neurology & Developmental Medicine University Children's Hospital Tübingen Germany.,Experimental Pediatric Neuroimaging University Children's Hospital Tübingen Germany
| |
Collapse
|
21
|
In Vivo NMR Studies of the Brain with Hereditary or Acquired Metabolic Disorders. Neurochem Res 2015; 40:2647-85. [PMID: 26610379 DOI: 10.1007/s11064-015-1772-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 11/10/2015] [Accepted: 11/12/2015] [Indexed: 01/09/2023]
Abstract
Metabolic disorders, whether hereditary or acquired, affect the brain, and abnormalities of the brain are related to cellular integrity; particularly in regard to neurons and astrocytes as well as interactions between them. Metabolic disturbances lead to alterations in cellular function as well as microscopic and macroscopic structural changes in the brain with diabetes, the most typical example of metabolic disorders, and a number of hereditary metabolic disorders. Alternatively, cellular dysfunction and degeneration of the brain lead to metabolic disturbances in hereditary neurological disorders with neurodegeneration. Nuclear magnetic resonance (NMR) techniques allow us to assess a range of pathophysiological changes of the brain in vivo. For example, magnetic resonance spectroscopy detects alterations in brain metabolism and energetics. Physiological magnetic resonance imaging (MRI) detects accompanying changes in cerebral blood flow related to neurovascular coupling. Diffusion and T1/T2-weighted MRI detect microscopic and macroscopic changes of the brain structure. This review summarizes current NMR findings of functional, physiological and biochemical alterations within a number of hereditary and acquired metabolic disorders in both animal models and humans. The global view of the impact of these metabolic disorders on the brain may be useful in identifying the unique and/or general patterns of abnormalities in the living brain related to the pathophysiology of the diseases, and identifying future fields of inquiry.
Collapse
|
22
|
Liaw HR, Lee HF, Chi CS, Tsai CR. Late infantile metachromatic leukodystrophy: Clinical manifestations of five Taiwanese patients and Genetic features in Asia. Orphanet J Rare Dis 2015; 10:144. [PMID: 26553228 PMCID: PMC4638099 DOI: 10.1186/s13023-015-0363-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/30/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study was conducted to describe the clinical and genetic features of patients with late infantile metachromatic leukodystrophy. METHODS Clinical and genetic manifestations of five Taiwanese patients with late infantile metachromatic leukodystrophy from January 2003 to April 2014 were reviewed. The genetic features of such patients reported in Asian countries during a period of 20 years were also analyzed. RESULTS The median age at disease onset was 1 year and 3 months with the first clinical symptom being gait disturbance. All five patients became bed-ridden at a median age of 2 years and 5 months. Nerve conduction velocity revealed demyelinating polyneuropathy and brain MRI disclosed tigroid and leopard skin pattern of dysmyelination in all 5 patients. All patients had decreased ARSA activities in leukocytes accounting for 15.88% to 30.75% of controls. Five novel mutations, p.A316D, p.G303R, p.Q176X, p.R293X, and c.749 insGCGGGCCA, were identified in our case series. Eighteen patients, including our 5 patients, were reported in Asian countries. A total of 22 different disease-causing alleles were found, in which p.W320X was identified in Taiwan and China, and p.G101V was found in Taiwan and Korea. CONCLUSIONS Patients with late infantile metachromatic leukodystrophy exhibited a rapid and devastating clinical course. The pattern of dysmyelination on brain MRI together with peripheral demyelination polyneuropathy indicates that evaluation of ARSA activity in leukocytes is warranted. A wide diversity of ARSA gene mutations was noted in Asia.
Collapse
Affiliation(s)
- Hsiang-Ru Liaw
- Department of Pediatrics, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung, 40705, Taiwan
| | - Hsiu-Fen Lee
- Department of Pediatrics, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung, 40705, Taiwan.,School of Medicine, Chung Shan Medical University, 110, Sec. 1, Jianguo N. Rd, Taichung, 40201, Taiwan
| | - Ching-Shiang Chi
- Department of Pediatrics, Tungs' Taichung Metroharbor Hospital, 699, Taiwan Boulevard Sec. 8, Wuchi, Taichung, 435, Taiwan. .,School of Medicine, Chung Shan Medical University, 110, Sec. 1, Jianguo N. Rd, Taichung, 40201, Taiwan.
| | - Chi-Ren Tsai
- Department of Pediatrics, Taichung Veterans General Hospital, 1650, Taiwan Boulevard Sec. 4, Taichung, 40705, Taiwan.,Institute of Molecular Biology, National Chung Hsing University, 250, Kuo Kuang Rd, Taichung, 402, Taiwan
| |
Collapse
|
23
|
Patay Z, Blaser SI, Poretti A, Huisman TAGM. Neurometabolic diseases of childhood. Pediatr Radiol 2015; 45 Suppl 3:S473-84. [PMID: 26346153 DOI: 10.1007/s00247-015-3279-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/12/2014] [Accepted: 01/06/2015] [Indexed: 01/09/2023]
Abstract
Metabolic diseases affecting the pediatric brain are complex conditions, the underlying mechanisms leading to structural damage are diverse and the diagnostic imaging manifestations are often non-specific; hence early, sensitive and specific diagnosis can be challenging for the radiologist. However, misdiagnosis or a delayed diagnosis can result in a devastating, irreversible injury to the developing brain. Based upon the inborn error, neurometabolic diseases can be subdivided in various groups depending on the predominantly involved tissue (e.g., white matter in leukodystrophies or leukoencephalopathies), the involved metabolic processes (e.g., organic acidurias and aminoacidopathies) and primary age of the child at presentation (e.g., neurometabolic disorders of the newborn). This manuscript summarizes these topics.
Collapse
Affiliation(s)
- Zoltan Patay
- Section of Neuroradiology, Division of Radiology, Department of Radiological Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | | | | |
Collapse
|
24
|
Tillema JM, Derks MG, Pouwels PJW, de Graaf P, van Rappard DF, Barkhof F, Steenweg ME, van der Knaap MS, Wolf NI. Volumetric MRI data correlate to disease severity in metachromatic leukodystrophy. Ann Clin Transl Neurol 2015; 2:932-40. [PMID: 26401514 PMCID: PMC4574810 DOI: 10.1002/acn3.232] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/13/2015] [Accepted: 07/20/2015] [Indexed: 11/13/2022] Open
Abstract
Objective Metachromatic leukodystrophy (MLD) is an inherited lysosomal disorder due to a deficiency in arylsulfatase A with progressive demyelination and neurological decline. This retrospective MRI study investigated the extent of cortical involvement at time of diagnosis, and clinical correlates to both conventional and regional volumetric measures of brain involvement. Methods 3D-T1-weighted MRI scans were used to determine cortical thickness and surface-based cerebral cortical gray matter (GM) and cerebral white matter (WM) volume (GMV and WMV), WM lesions, thalamus, and cerebellum. MRI-MLD severity scores were obtained from FLAIR images. Associations between clinical and imaging data were examined using correlation coefficients. Results Twenty patients with MLD (mean age 13.7 years, range 2–35) and 20 controls (mean age 13.9 years, range 2–40) were included. Compared with control subjects, late-infantile, and juvenile patients (n = 14) had significantly diminished cerebral cortical GMV and thalamus volume (P < 0.05), but did not differ in WMV and cortical thickness. Adult patients (n = 6) showed significantly reduced GMV, WMV and cortical thickness (all P < 0.05). Regional analysis showed statistically significant cortical thinning in the cingulate gyrus and most pronounced thinning with age in the frontal lobe of MLD patients. Intelligence quotient (IQ) correlated with MRI-MLD scores (r = −0.87, P < 0.001). Interpretation Significant cerebral cortical GMV loss is already present in early stages of MLD. IQ correlates with WM severity scores and lesion volume, but not with volumetric measures. In adult presentations, there is more pronounced global atrophy with GMV and WMV loss and accelerated cortical thinning, most prominently in the cingulate gyrus and frontal lobes.
Collapse
Affiliation(s)
- Jan-Mendelt Tillema
- Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, The Netherlands ; Department of Neurology, Mayo Clinic 200 First St. SW, Rochester, Minnesota, 55905
| | - Marloes Gm Derks
- Department of Child Neurology, VU University Medical Center Amsterdam, The Netherlands
| | - Petra J W Pouwels
- Department of Physics and Medical Technology, VU University Medical Center Amsterdam, The Netherlands ; Neuroscience Campus Amsterdam Amsterdam, The Netherlands
| | - Pim de Graaf
- Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, The Netherlands ; Neuroscience Campus Amsterdam Amsterdam, The Netherlands
| | - Diane F van Rappard
- Department of Child Neurology, VU University Medical Center Amsterdam, The Netherlands ; Neuroscience Campus Amsterdam Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, The Netherlands ; Neuroscience Campus Amsterdam Amsterdam, The Netherlands
| | - Marjan E Steenweg
- Department of Child Neurology, VU University Medical Center Amsterdam, The Netherlands ; Neuroscience Campus Amsterdam Amsterdam, The Netherlands
| | - Marjo S van der Knaap
- Department of Child Neurology, VU University Medical Center Amsterdam, The Netherlands ; Neuroscience Campus Amsterdam Amsterdam, The Netherlands ; Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, The Netherlands
| | - Nicole I Wolf
- Department of Child Neurology, VU University Medical Center Amsterdam, The Netherlands ; Neuroscience Campus Amsterdam Amsterdam, The Netherlands
| |
Collapse
|
25
|
Boucher AA, Miller W, Shanley R, Ziegler R, Lund T, Raymond G, Orchard PJ. Long-term outcomes after allogeneic hematopoietic stem cell transplantation for metachromatic leukodystrophy: the largest single-institution cohort report. Orphanet J Rare Dis 2015; 10:94. [PMID: 26245762 PMCID: PMC4545855 DOI: 10.1186/s13023-015-0313-y] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/29/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Metachromatic Leukodystrophy (MLD) is a rare, fatal demyelinating disorder with limited treatment options. Published outcomes after hematopoietic stem cell transplantation (HSCT) are scant and mixed. We report survival and function following HSCT for a large, single-center MLD cohort. METHODS Transplant-related data, survival and serial measures (brain MRI, nerve conduction velocity (NCV), neurologic and neuropsychology evaluations) were reviewed. When possible, parental interviews informed current neurologic status, quality-of-life, and adaptive functioning. Gross motor and expressive functions for late-infantile (LI-MLD) and juvenile (J-MLD) patients were described using previously reported, MLD-specific scales. RESULTS Forty patients with confirmed MLD have undergone HSCT at our center. Twenty-one (53 %) survive at a median 12 years post-HSCT. Most deaths (n = 17) were treatment-related; two died from disease progression. Survival did not depend upon MLD subtype or symptom status at transplant. LI-MLD patients survive beyond reported life expectancy in untreated disease. Abnormal brain MRI and peripheral nerve conduction velocities (NCV) were common before HSCT. Following transplant, fewer patients experienced MRI progression compared to NCV deterioration. Sixteen LI-MLD and J-MLD survivors were evaluable for long-term gross motor and/or expressive language functioning using existing MLD clinical scoring systems. While most J-MLD patients regressed, the aggregate cohort demonstrated superior retention of function compared to published natural history. Seventeen LI-MLD, J-MLD and adult subtype (A-MLD) survivors were evaluable for long-term adaptive functioning, activities of daily living, and/or cognition. Relative cognitive sparing was observed despite overall global decline. Five sibling pairs (one LI-MLD and four J-MLD), in which at least one underwent transplant in our cohort, were evaluable. Within each familial dyad, survival or function was superior for the treated sibling, or if both siblings were transplanted, for the pre-symptomatic sibling. CONCLUSIONS HSCT is a viable treatment option for MLD, but has significant limitations. Later-onset phenotypes may benefit most from early, pre-symptomatic transplant. Until superior, novel treatment strategies are demonstrated, MLD patients should be carefully considered for HSCT.
Collapse
Affiliation(s)
- Alexander A Boucher
- Department of Internal Medicine and Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Weston Miller
- Division of Pediatric Blood and Marrow Transplantation, 420 Delaware Street SE, MMC 484, Minneapolis, MN, 55455, USA.
| | - Ryan Shanley
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Richard Ziegler
- Division of Pediatric Clinical Neurosciences, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Troy Lund
- Division of Pediatric Blood and Marrow Transplantation, 420 Delaware Street SE, MMC 484, Minneapolis, MN, 55455, USA.
| | - Gerald Raymond
- Division of Pediatric Clinical Neurosciences, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplantation, 420 Delaware Street SE, MMC 484, Minneapolis, MN, 55455, USA.
| |
Collapse
|
26
|
van Rappard DF, Boelens JJ, Wolf NI. Metachromatic leukodystrophy: Disease spectrum and approaches for treatment. Best Pract Res Clin Endocrinol Metab 2015; 29:261-73. [PMID: 25987178 DOI: 10.1016/j.beem.2014.10.001] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Metachromatic leukodystrophy is an inherited lysosomal disorder caused by recessive mutations in ARSA encoding arylsulfatase A. Low activity of arylsulfatase A results in the accumulation of sulfatides in the central and peripheral nervous system leading to demyelination. The disease is classified in a late-infantile, juvenile and adult onset type based on the age of onset, all characterized by a variety of neurological symptoms, which eventually lead to death if untreated. There is no curative treatment for all types and stages. This review discusses diagnostic process and efficacy of current and possible future therapies such as hematopoietic stem cell transplantation, enzyme replacement therapy and gene therapy. A systematic evaluation regarding the efficacy of hematopoietic stem cell transplantation and a longer follow up period for gene therapy are needed to come to a general conclusion and improve treatment options for metachromatic leukodystrophy.
Collapse
Affiliation(s)
- Diane F van Rappard
- Department of Child Neurology, Center for Children with White Matter Disorders, VU Medical Centre and Neuroscience Campus, Postbox 7057, 1007 MB Amsterdam, The Netherlands.
| | - Jaap Jan Boelens
- Department of Pediatrics, Blood and Marrow Transplantation Program, University Medical Center Utrecht, PO Box 85090, 3503 AB Utrecht, The Netherlands.
| | - Nicole I Wolf
- Department of Child Neurology, Center for Children with White Matter Disorders, VU Medical Centre and Neuroscience Campus, Postbox 7057, 1007 MB Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Mahmood A, Chacham S, Reddy UN, Rao JN, Rao SP. A 5-year-old male child with late infantile metachromatic leukodystrophy: a case report. J Child Neurol 2015; 30:483-5. [PMID: 25117420 DOI: 10.1177/0883073814542948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 06/06/2014] [Indexed: 11/17/2022]
Abstract
Metachromatic leukodystrophy is a rare disorder of myelin metabolism. This degenerative disorder results from the accumulation of cerebroside sulfatide within the myelin sheath of central and peripheral nervous system, due to deficiency of aryl sulfatase A enzyme. We report a 5-year-old male child, who presented with regression of milestones, recurrent seizures and spasticity from second year of life. Initially neurodegenerative disorder was considered and the case was investigated with neuroimaging and enzyme levels. Computed tomography (CT) of the brain showed hypodensities in the corpus callosum and bilateral periventricular and deep cerebral white matter suggestive of neurodegenerative disorder. Subsequently, magnetic resonance imaging (MRI) of the brain was done, which showed symmetrical hyperintensities in the periventricular white matter with classical sparing of subcortical "U" fibers. The β-galactosidase enzyme activity was normal; however, the activity of aryl sulfatase A enzyme was undetectable, confirming the diagnosis of late infantile variant of metachromatic leukodystrophy.
Collapse
Affiliation(s)
- Afreen Mahmood
- Department of Paediatrics, Deccan College of Medical Sciences, Princess Esra Hospital, Hyderabad, Andhra Pradesh, India
| | - Swathi Chacham
- Department of Paediatrics, Deccan College of Medical Sciences, Princess Esra Hospital, Hyderabad, Andhra Pradesh, India
| | - Uppin Narayan Reddy
- Department of Paediatrics, Deccan College of Medical Sciences, Princess Esra Hospital, Hyderabad, Andhra Pradesh, India
| | - Jillalla Narsing Rao
- Department of Paediatrics, Deccan College of Medical Sciences, Princess Esra Hospital, Hyderabad, Andhra Pradesh, India
| | - S Pratap Rao
- Department of Paediatrics, Deccan College of Medical Sciences, Princess Esra Hospital, Hyderabad, Andhra Pradesh, India
| |
Collapse
|
28
|
The clinical features and diagnosis of Metachromatic leukodystrophy: A case series of Iranian Pediatric Patients. IRANIAN JOURNAL OF CHILD NEUROLOGY 2015; 9:57-61. [PMID: 26401154 PMCID: PMC4577699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 07/15/2015] [Accepted: 07/19/2015] [Indexed: 11/02/2022]
Abstract
OBJECTIVE Metachromatic leukodystrophy disorder (MLD) is one of the rare neurometabolic diseases caused due to lack of saposin B and arylsulfatase A enzyme deficiency. MATERIALS & METHODS Eighteen patients diagnosed as metachromatic leukodystrophy in the Neurology Department of Mofid Children's Hospital in Tehran, Iran between 2010 and 2014 were included in our study. The disorder was confirmed by clinical, EMG-NCV, arylsulfatase A enzyme checking and neuroimaging findings along with neurometabolic and genetic assessment from reference laboratory in Iran. We assessed age, gender, past medical history, developmental status, clinical manifestations, and neuroimaging findings of 18 patients with metachromatic leukodystrophy. RESULTS From 18 patients, 80% were offspring from consanguineous marriages. A family history of metachromatic leukodystrophy disease was positive for four patients. Twelve patients had late infantile form of this disorder and six patients had juvenile form. A history of tonic type seizure was positive in 20% of the patients and tonic spasm was confirmed with clinical information. Electromyographgraphy (EMG) in 96% of patients was abnormal with demyelinating sensorimotor neuropathy pattern. MRI in all patients showed the leukodystrophic pattern as arcuate fibers sparing and subcortical rim in white matter and periventricular involvement. Our diagnosis was confirmed by EMG-NCV findings with sensorimotor neuropathy pattern and the assessment of arylsulfatase A enzyme function. CONCLUSION MLD is an inheritance metabolic disorder, which was confirmed by the assessment of arylsulfatase A enzyme function, peripheral blood leukocyte that assessed in a referral laboratory in Iran.
Collapse
|
29
|
Alroy J, Garganta C, Wiederschain G. Secondary biochemical and morphological consequences in lysosomal storage diseases. BIOCHEMISTRY (MOSCOW) 2014; 79:619-36. [DOI: 10.1134/s0006297914070049] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
30
|
Kehrer C, Groeschel S, Kustermann-Kuhn B, Bürger F, Köhler W, Kohlschütter A, Bley A, Steinfeld R, Gieselmann V, Krägeloh-Mann I. Language and cognition in children with metachromatic leukodystrophy: onset and natural course in a nationwide cohort. Orphanet J Rare Dis 2014; 9:18. [PMID: 24499656 PMCID: PMC3922034 DOI: 10.1186/1750-1172-9-18] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/10/2014] [Indexed: 12/02/2022] Open
Abstract
Background Metachromatic leukodystrophy (MLD) is a rare, genetic neurodegenerative disease. It leads to progressive demyelination resulting in regression of development and early death. With regard to experimental therapies, knowledge of the natural course of the disease is highly important. We aimed to analyse onset and character of first symptoms in MLD and to provide detailed natural course data concerning language and cognition. Methods Patients with MLD were recruited nationwide within the scope of the German research network LEUKONET. 59 patients’ questionnaires (23 late-infantile, 36 juvenile) were analysed. Results Time from first symptoms (at a median age of 1.5 years in late-infantile and 6 years in juvenile MLD) to diagnosis took one year in late-infantile and two years in juvenile patients on average. Gait disturbances and abnormal movement patterns were first signs in all patients with late-infantile and in most with juvenile MLD. Onset in the latter was additionally characterized by problems in concentration, behaviour and fine motor function (p = 0.0011, p < 0.0001, and p = 0.0012). Half of late-infantile patients did not learn to speak in complete sentences after an initially normal language acquisition. They showed a rapid language decline with first language difficulties at a median age of 2.5 years and complete loss of expressive language within several months (median age 32, range 22–47 months). This was followed by total loss of communication at a median age of around four years. In juvenile patients, language decline was more protracted, and problems in concentration and behaviour were followed by decline in skills for reading, writing and calculating around four years after disease onset. Conclusions Our data reflect the natural course of decline in language and cognition in late-infantile and juvenile MLD in a large cohort over a long observation period. This is especially relevant to juvenile patients where the disease course is protracted and prospective studies are hardly feasible. Knowledge of first symptoms may lead to earlier diagnosis and subsequently to a better outcome following therapeutic intervention. Our data may serve as a reference for individual treatment decisions and for evaluation of clinical outcome after treatment intervention.
Collapse
Affiliation(s)
- Christiane Kehrer
- Department of Paediatric Neurology and Developmental Medicine, University Children's Hospital, Hoppe-Seyler-Strasse 1, 72076 Tübingen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|