1
|
Kaur N, Singh J. Generation and Characterization of Human iPSC-Derived Astrocytes with Potential for Modeling X-Linked Adrenoleukodystrophy Phenotypes. Int J Mol Sci 2025; 26:1576. [PMID: 40004040 PMCID: PMC11855073 DOI: 10.3390/ijms26041576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
X-adrenoleukodystrophy (X-ALD) is a peroxisomal metabolic disorder caused by mutations in the ABCD1 gene encoding the peroxisomal ABC transporter adrenoleukodystrophy protein (ALDP). Similar mutations in ABCD1 may result in a spectrum of phenotypes in males with slow progressing adrenomyeloneuropathy (AMN) and fatal cerebral adrenoleukodystrophy (cALD) dominating most cases. Mouse models of X-ALD do not capture the phenotype differences and an appropriate model to investigate the mechanism of disease onset and progress remains a critical need. Here, we generated induced pluripotent stem cell (iPSC) lines from skin fibroblasts of two each of apparently healthy control, AMN, and cALD patients with non-integrating mRNA-based reprogramming. iPSC lines expanded normally and expressed pluripotency markers Oct4, SOX2, NANOG, SSEA, and TRA-1-60. Expression of markers SOX17, Brachyury, Desmin, OXT2, and beta tubulin III demonstrated the ability of the iPSCs to differentiate into all three germ layers. iPSC-derived lines from CTL, AMN, and cALD male patients were differentiated into astrocytes. Differentiated AMN and cALD astrocytes lacked ABCD1 expression and accumulated saturated very long chain fatty acids (VLCFAs), a hallmark of X-ALD, and demonstrated differential mitochondrial bioenergetics, cytokine gene expression, and differences in STAT3 and AMPK signaling between AMN and cALD astrocytes. These patient astrocytes provide disease-relevant tools to investigate the mechanism of differential neuroinflammatory response in X-ALD and will be valuable cell models for testing new therapeutics.
Collapse
Affiliation(s)
- Navtej Kaur
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA;
| | - Jaspreet Singh
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA;
- Department of Physiology, Michigan State University, Lansing, MI 48824, USA
| |
Collapse
|
2
|
Kornbluh AB, Baldwin A, Fatemi A, Vanderver A, Adang LA, Van Haren K, Sampson J, Eichler FS, Sadjadi R, Engelen M, Orthmann-Murphy JL. Practical Approach to Longitudinal Neurologic Care of Adults With X-Linked Adrenoleukodystrophy and Adrenomyeloneuropathy. Neurol Genet 2024; 10:e200192. [PMID: 39372123 PMCID: PMC11450743 DOI: 10.1212/nxg.0000000000200192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/26/2024] [Indexed: 10/08/2024]
Abstract
Although X-linked adrenoleukodystrophy (ALD) has historically been considered a childhood disease managed by pediatric neurologists, it is one of the most common leukodystrophies diagnosed in adulthood. An increase in both male and female adults reaching diagnosis due to familial cases identified by state newborn screening panels and more widespread use of genetic testing results in a large cohort of presymptomatic or early symptomatic adults. This population is in urgent need of standardized assessments and follow-up care. Adults with ALD/adrenomyeloneuropathy (AMN) may be diagnosed in a variety of ways, including after another family member is identified via genetic testing or newborn screening, presenting for symptomatic evaluation, or following diagnosis with primary adrenal insufficiency. Significant provider, patient, and systems-based barriers prevent adult patients with ALD/AMN from receiving appropriate care, including lack of awareness of the importance of longitudinal neurologic management. Confirmation of and education about the diagnosis should be coordinated in conjunction with a genetic counselor. Routine surveillance for adrenal insufficiency and onset of cerebral ALD (CALD) in men should be performed systematically to avoid preventable morbidity and mortality. While women with ALD do not usually develop cerebral demyelination or adrenal insufficiency, they remain at risk for myeloneuropathy and are no longer considered "carriers." After diagnosis, patients should be connected to the robust support networks, foundations, and research organizations available for ALD/AMN. Core principles of neurologic symptom management parallel those for patients with other etiologies of progressive spastic paraplegia. Appropriate patient candidates for hematopoietic stem cell transplant (HSCT) and other investigational disease-modifying strategies require early identification to achieve optimal outcomes. All patients with ALD/AMN, regardless of sex, age, or symptom severity, benefit from a multidisciplinary approach to longitudinal care spearheaded by the neurologist. This review proposes key strategies for diagnostic confirmation, laboratory and imaging surveillance, approach to symptom management, and guidance for identification of appropriate candidates for HSCT and investigational treatments.
Collapse
Affiliation(s)
- Alexandra B Kornbluh
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| | - Aaron Baldwin
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| | - Ali Fatemi
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| | - Adeline Vanderver
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| | - Laura A Adang
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| | - Keith Van Haren
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| | - Jacinda Sampson
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| | - Florian S Eichler
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| | - Reza Sadjadi
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| | - Marc Engelen
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| | - Jennifer L Orthmann-Murphy
- From the Division of Neurology (A.B.K.), Children's National Hospital, George Washington University Medical School, Washington DC; Division of Neurology (A.B.), Neurogenetics Translational Center of Excellence, University of Pennsylvania, Philadelphia; Kennedy Krieger Institute and The Johns Hopkins University School of Medicine (A.F.), Baltimore, MD; Division of Neurology (A.V., L.A.A.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Department of Neurology and Pediatrics (K.V.H., J.S.), Lucile Packard Children's Hospital, Stanford University School of Medicine, Palo Alto, CA; Department of Neurology (F.S.E., R.S.), Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatric Neurology (M.E.), Amsterdam UMC location, University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, the Netherlands; and Department of Neurology (J.L.O.-M.), University of Pennsylvania, Philadelphia
| |
Collapse
|
3
|
Qi W, Cao D, Hao L, Guo X. A female adult-onset X-ALD patient with pure cerebellar symptoms:a case report. Heliyon 2024; 10:e35705. [PMID: 39170489 PMCID: PMC11336854 DOI: 10.1016/j.heliyon.2024.e35705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD) caused by ATP-binding cassette subfamily D member 1 (ABCD1) gene defects is the most common inherited peroxisomal disorder.The female cerebello-brainstem dominant type in which cerebellum and brainstem are mainly involved is very rare. We report a 40-year-old female who was diagnosed as the rare disorder with magnetic resonance imaging (MRI) and genetic analysis mainly. Her initial symptoms were progressive slurred speech and writing disturbance. Her brain MRI showed obvious atrophy of brainstem and cerebellum. She did not have adrenal insufficiency. Genetic analysis showed a heterozygous missense mutation in exon 4 of the coding region of ABCD1 (c.1252C > T, p.Arg418Trp).This is the first report of this particular mutation being associated with the cerebello-brainstem dominant phenotype of X-ALD, as well as the first description of this X-ALD variant in a (heterozygous) female patient.X-ALD should be considered in young and middle-aged patients with slow-progressing ataxia and dysarthria.
Collapse
Affiliation(s)
| | | | - Lei Hao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, China
| | - Xiuming Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, China
| |
Collapse
|
4
|
Chakraborty S, Gupta AK, Gupta N, Meena JP, Seth R, Kabra M. Hematopoietic Stem Cell Transplantation for Storage Disorders: Present Status. Indian J Pediatr 2024; 91:830-838. [PMID: 38639861 DOI: 10.1007/s12098-024-05110-4] [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: 12/25/2023] [Accepted: 03/15/2024] [Indexed: 04/20/2024]
Abstract
Storage disorders are a group of inborn errors of metabolism caused by the defective activity of lysosomal enzymes or transporters. All of these disorders have multisystem involvement with variable degrees of neurological features. Neurological manifestations are one of the most difficult aspects of treatment concerning these diseases. The available treatment modalities for some of these disorders include enzyme replacement therapy, substrate reduction therapy, hematopoietic stem cell transplantation (HSCT) and the upcoming gene therapies. As a one-time intervention, the economic feasibility of HSCT makes it an attractive option for treating these disorders, especially in lower and middle-income countries. Further, improvements in peri-transplantation medical care, better conditioning regimens and better supportive care have improved the outcomes of patients undergoing HSCT. In this review, we discuss the current evidence for HSCT in various storage disorders and its suitability as a mode of therapy for the developing world.
Collapse
Affiliation(s)
- Soumalya Chakraborty
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Aditya Kumar Gupta
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Neerja Gupta
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Jagdish Prasad Meena
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rachna Seth
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Madhulika Kabra
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
5
|
Kaur N, Singh J. Generating human AMN and cALD iPSC-derived astrocytes with potential for modeling X-linked adrenoleukodystrophy phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596696. [PMID: 38854155 PMCID: PMC11160757 DOI: 10.1101/2024.05.31.596696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
X-adrenoleukodystrophy (X-ALD) is a peroxisomal metabolic disorder caused by mutations in the ABCD1 gene encoding the peroxisomal ABC transporter adrenoleukodystrophy protein (ALDP). Similar mutations in ABCD1 may result in a spectrum of phenotypes in males with slow progressing adrenomyeloneuropathy (AMN) and fatal cerebral adrenoleukodystrophy (cALD) dominating the majority of cases. Mouse model of X-ALD does not capture the phenotype differences and an appropriate model to investigate mechanism of disease onset and progress remains a critical need. Induced pluripotent stem cell (iPSC)-derived and cell models derived from them have provided useful tools for investigating cell-type specific disease mechanisms. Here, we generated induced pluripotent stem cell (iPSC) lines from skin fibroblasts of two each of apparently healthy control, AMN and cALD patients with non-integrating mRNA-based reprogramming. iPSC lines expanded normally and expressed pluripotency markers Oct4, SOX2, Nanog, SSEA and TRA-1-60. Expression of markers SOX17, brachyury, Desmin, Oxt2 and beta tubulin III demonstrated the ability of the iPSCs to differentiate into all three germ layers. iPSC-derived lines from CTL, AMN and cALD male patients were differentiated into astrocytes. Differentiated AMN and cALD astrocytes lacked ABCD1 expression and accumulated VLCFA, a hallmark of X-ALD. These patient astrocytes provide disease-relevant tools to investigate mechanism of differential neuroinflammatory response and metabolic reprogramming in X-ALD. Further these patient-derived human astrocyte cell models will be valuable for testing new therapeutics.
Collapse
|
6
|
Chen Y, Xu LP, Zhang XH, Chen H, Liu KY, Qing J, Yang YL, Huang XJ. Haploidentical hematopoietic stem cell transplantation with busulfan, cyclophosphamide, and fludarabine conditioning for X-linked adrenal cerebral leukodystrophy. Pediatr Transplant 2024; 28:e14735. [PMID: 38602169 DOI: 10.1111/petr.14735] [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: 07/04/2023] [Revised: 11/20/2023] [Accepted: 02/20/2024] [Indexed: 04/12/2024]
Abstract
OBJECTIVE We investigated the safety and efficacy of haploidentical stem cell transplantation (SCT) in pediatric patients with X-linked adrenoleukodystrophy (ALD). METHODS A retrospective analysis of transplantation data from 29 cases of ALD, treated between December 2014 and April 2022, was conducted. Neurologic function scores (NFS) were assessed. The conditioning regimen was busulfan 9.6 mg/kg, cyclophosphamide 200 mg/kg, and fludarabine 90 mg/m2 (BFC). Graft-versus-host disease prophylaxis consisted of anti-human thymocyte globulin, cyclosporine A, mycophenolate mofetil, and short course of methotrexate. RESULTS Among the 29 cases, 14 cases (NFS = 0) were asymptomatic, and 15 (NFS ≥ 1) were symptomatic. The median age at SCT was 8 years (range: 4-16 years); the median follow-up time was 1058 days (range: 398-3092 days); 28 cases were father donors and 1 case was a grandfather donor. Hematopoietic reconstitution was successful in all patients, and all of them achieved complete donor chimerism at the time of engraftment. The leading cause of death was still primary disease progression (n = 4). Survival free of major functional disabilities was 100% in asymptomatic patients versus 66.67% in the symptomatic group (p = .018). CONCLUSION BFC regimen used in haploidentical SCT was administered safely without major transplant-related complications even in symptomatic patients, and neurological symptoms were stabilized after SCT.
Collapse
Affiliation(s)
- Yao Chen
- Peking University People's Hospital, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Huan Chen
- Peking University People's Hospital, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Kai-Yan Liu
- Peking University People's Hospital, Beijing, China
- Peking University Institute of Hematology, Beijing, China
| | - Jiong Qing
- Peking University People's Hospital, Beijing, China
| | | | - Xiao-Jun Huang
- Peking University People's Hospital, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| |
Collapse
|
7
|
Weldrick CL, Boers P, Kiely P, O'Halloran L. X-linked cerebral adrenoleukodystrophy. BMJ Case Rep 2023; 16:e237905. [PMID: 37907311 PMCID: PMC10619069 DOI: 10.1136/bcr-2020-237905] [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] [Indexed: 11/02/2023] Open
Abstract
A man in his 30s presented with a 6-month history of progressive left face, arm and leg weakness. Medical history included epilepsy and vitamin B12 deficiency. Three maternal second degree relatives died before the age of 7 from various neurological disorders. Examination revealed a mild left facial droop and weakness of the left shoulder, hip and ankle. Reflexes were symmetrical and tone was normal. Differential diagnosis included glioma, subacute infarction, lymphoma and demyelination. MRI brain showed an extensive right sided subcortical white matter lesion, with extension into the brainstem. The patient's weakness progressed over 3 months. Brain biopsy showed evidence of demyelination and gliosis. A pathological diagnosis of tumefactive multiple sclerosis was made, but also rare metabolic disorders such as X-linked adrenoleukodystrophy (X-ALD) were proposed. Serum very long-chain fatty acids were significantly elevated. Genetic testing showed a mutation in the ABCD1 gene, confirming a diagnosis of X-ALD.
Collapse
Affiliation(s)
| | - Peter Boers
- Neurology, University Hospital Limerick, Limerick, Ireland
| | - Patrick Kiely
- Radiology, University Hospital Limerick, Dooradoyle, Ireland
| | - Liam O'Halloran
- Radiology, University Hospital Limerick, Dooradoyle, Ireland
| |
Collapse
|
8
|
Shim YJ, Shin MK, Jung J, Koo B, Jang W. An in-silico approach to studying a very rare neurodegenerative disease using a disease with higher prevalence with shared pathways and genes: Cerebral adrenoleukodystrophy and Alzheimer’s disease. Front Mol Neurosci 2022; 15:996698. [PMID: 36245924 PMCID: PMC9553843 DOI: 10.3389/fnmol.2022.996698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022] Open
Abstract
Cerebral adrenoleukodystrophy (cALD) is a rare neurodegenerative disease characterized by inflammatory demyelination in the central nervous system. Another neurodegenerative disease with a high prevalence, Alzheimer’s disease (AD), shares many common features with cALD such as cognitive impairment and the alleviation of symptoms by erucic acid. We investigated cALD and AD in parallel to study the shared pathological pathways between a rare disease and a more common disease. The approach may expand the biological understandings and reveal novel therapeutic targets. Gene set enrichment analysis (GSEA) and weighted gene correlation network analysis (WGCNA) were conducted to identify both the resemblance in gene expression patterns and genes that are pathologically relevant in the two diseases. Within differentially expressed genes (DEGs), GSEA identified 266 common genes with similar up- or down-regulation patterns in cALD and AD. Among the interconnected genes in AD data, two gene sets containing 1,486 genes preserved in cALD data were selected by WGCNA that may significantly affect the development and progression of cALD. WGCNA results filtered by functional correlation via protein–protein interaction analysis overlapping with GSEA revealed four genes (annexin A5, beta-2-microglobulin, CD44 molecule, and fibroblast growth factor 2) that showed robust associations with the pathogeneses of cALD and AD, where they were highly involved in inflammation, apoptosis, and the mitogen-activated protein kinase pathway. This study provided an integrated strategy to provide new insights into a rare disease with scant publicly available data (cALD) using a more prevalent disorder with some pathological association (AD), which suggests novel druggable targets and drug candidates.
Collapse
Affiliation(s)
- Yu Jeong Shim
- Department of Life Science, Dongguk University, Goyang-si, South Korea
| | - Min Kyoung Shin
- Department of Life Science, Dongguk University, Goyang-si, South Korea
| | - Junghyun Jung
- Department of Life Science, Dongguk University, Goyang-si, South Korea
| | | | - Wonhee Jang
- Department of Life Science, Dongguk University, Goyang-si, South Korea
- *Correspondence: Wonhee Jang,
| |
Collapse
|
9
|
Gupta AO, Raymond G, Pierpont RI, Kemp S, McIvor RS, Rayannavar A, Miller B, Lund TC, Orchard PJ. Treatment of cerebral adrenoleukodystrophy: allogeneic transplantation and lentiviral gene therapy. Expert Opin Biol Ther 2022; 22:1151-1162. [DOI: 10.1080/14712598.2022.2124857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Ashish O Gupta
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapies, University of Minnesota
| | - Gerald Raymond
- Division of Neurogenetics and The Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Rene I Pierpont
- Division of Clinical Behavioral Neuroscience, Department of Pediatrics, University of Minnesota
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC - University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Neuroscience, 1105 AZ Amsterdam, The Netherlands
| | - R Scott McIvor
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota
| | | | - Bradley Miller
- Division of Pediatric Endocrinology, University of Minnesota
| | - Troy C Lund
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapies, University of Minnesota
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplant and Cellular Therapies, University of Minnesota
| |
Collapse
|
10
|
de Vasconcelos P, Lacerda JF. Hematopoietic Stem Cell Transplantation for Neurological Disorders: A Focus on Inborn Errors of Metabolism. Front Cell Neurosci 2022; 16:895511. [PMID: 35693884 PMCID: PMC9178264 DOI: 10.3389/fncel.2022.895511] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022] Open
Abstract
Hematopoietic stem cells have been investigated and applied for the treatment of certain neurological disorders for a long time. Currently, their therapeutic potential is harnessed in autologous and allogeneic hematopoietic stem cell transplantation (HSCT). Autologous HSCT is helpful in immune-mediated neurological diseases such as Multiple Sclerosis. However, clinical benefits derive more from the immunosuppressive conditioning regimen than the interaction between stem cells and the nervous system. Mainly used for hematologic malignancies, allogeneic HSCT explores the therapeutic potential of donor-derived hematopoietic stem cells. In the neurological setting, it has proven to be most valuable in Inborn Errors of Metabolism, a large spectrum of multisystem disorders characterized by congenital deficiencies in enzymes involved in metabolic pathways. Inborn Errors of Metabolism such as X-linked Adrenoleukodystrophy present with brain accumulation of enzymatic substrates that result in progressive inflammatory demyelination. Allogeneic HSCT can halt ongoing inflammatory neural destruction by replacing hematopoietic-originated microglia with donor-derived myeloid precursors. Microglia, the only neural cells successfully transplanted thus far, are the most valuable source of central nervous system metabolic correction and play a significant role in the crosstalk between the brain and hematopoietic stem cells. After transplantation, engrafted donor-derived myeloid cells modulate the neural microenvironment by recapitulating microglial functions and enhancing repair mechanisms such as remyelination. In some disorders, additional benefits result from the donor hematopoietic stem cell secretome that cross-corrects neighboring neural cells via mannose-6-phosphatase paracrine pathways. The limitations of allogeneic HSCT in this setting relate to the slow turnover of microglia and complications such as graft-vs.-host disease. These restraints have accelerated the development of hematopoietic stem cell gene therapy, where autologous hematopoietic stem cells are collected, manipulated ex vivo to overexpress the missing enzyme, and infused back into the patient. With this cellular drug vehicle strategy, the brain is populated by improved cells and exposed to supraphysiological levels of the flawed protein, resulting in metabolic correction. This review focuses on the mechanisms of brain repair resulting from HSCT and gene therapy in Inborn Errors of Metabolism. A brief mention will also be made on immune-mediated nervous system diseases that are treated with this approach.
Collapse
Affiliation(s)
- Pedro de Vasconcelos
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - João F. Lacerda
- Serviço de Hematologia e Transplantação de Medula, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- JLacerda Lab, Hematology and Transplantation Immunology, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
11
|
Nisticò D, Bossini B, Benvenuto S, Pellegrin MC, Tornese G. Pediatric Adrenal Insufficiency: Challenges and Solutions. Ther Clin Risk Manag 2022; 18:47-60. [PMID: 35046659 PMCID: PMC8761033 DOI: 10.2147/tcrm.s294065] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/28/2021] [Indexed: 01/19/2023] Open
Abstract
Adrenal insufficiency is an insidious diagnosis that can be initially misdiagnosed as other life-threatening endocrine conditions, as well as sepsis, metabolic disorders, or cardiovascular disease. In newborns, cortisol deficiency causes delayed bile acid synthesis and transport maturation, determining prolonged cholestatic jaundice. Subclinical adrenal insufficiency is a particular challenge for a pediatric endocrinologist, representing the preclinical stage of acute adrenal insufficiency. Although often included in the extensive work-up of an unwell child, a single cortisol value is usually difficult to interpret; therefore, in most cases, a dynamic test is required for diagnosis to assess the hypothalamic-pituitary-adrenal axis. Stimulation tests using corticotropin analogs are recommended as first-line for diagnosis. All patients with adrenal insufficiency need long-term glucocorticoid replacement therapy, and oral hydrocortisone is the first-choice replacement treatment in pediatric. However, children that experience low cortisol concentrations and symptoms of cortisol insufficiency can take advantage using a modified release hydrocortisone formulation. The acute adrenal crisis is a life-threatening condition in all ages, treatment is effective if administered promptly, and it must not be delayed for any reason.
Collapse
Affiliation(s)
| | | | | | | | - Gianluca Tornese
- Department of Pediatrics, Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
12
|
Structure and Function of the ABCD1 Variant Database: 20 Years, 940 Pathogenic Variants, and 3400 Cases of Adrenoleukodystrophy. Cells 2022; 11:cells11020283. [PMID: 35053399 PMCID: PMC8773697 DOI: 10.3390/cells11020283] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/16/2022] Open
Abstract
The progressive neurometabolic disorder X-linked adrenoleukodystrophy (ALD) is caused by pathogenic variants in the ABCD1 gene, which encodes the peroxisomal ATP-binding transporter for very-long-chain fatty acids. The clinical spectrum of ALD includes adrenal insufficiency, myelopathy, and/or leukodystrophy. A complicating factor in disease management is the absence of a genotype–phenotype correlation in ALD. Since 1999, most ABCD1 (likely) pathogenic and benign variants have been reported in the ABCD1 Variant Database. In 2017, following the expansion of ALD newborn screening, the database was rebuilt. To add an additional level of confidence with respect to pathogenicity, for each variant, it now also reports the number of cases identified and, where available, experimental data supporting the pathogenicity of the variant. The website also provides information on a number of ALD-related topics in several languages. Here, we provide an updated analysis of the known variants in ABCD1. The order of pathogenic variant frequency, overall clustering of disease-causing variants in exons 1–2 (transmembrane domain spanning region) and 6–9 (ATP-binding domain), and the most commonly reported pathogenic variant p.Gln472Argfs*83 in exon 5 are consistent with the initial reports of the mutation database. Novel insights include nonrandom clustering of high-density missense variant hotspots within exons 1, 2, 6, 8, and 9. Perhaps more importantly, we illustrate the importance of collaboration and utility of the database as a scientific, clinical, and ALD-community-wide resource.
Collapse
|
13
|
Boyd MJ, Collier PN, Clark MP, Deng H, Kesavan S, Ronkin SM, Waal N, Wang J, Cao J, Li P, Come J, Davies I, Duffy JP, Cochran JE, Court JJ, Chandupatla K, Jackson KL, Maltais F, O'Dowd H, Boucher C, Considine T, Taylor WP, Gao H, Chakilam A, Engtrakul J, Crawford D, Doyle E, Phillips J, Kemper R, Swett R, Empfield J, Bunnage ME, Charifson PS, Magavi SS. Discovery of Novel, Orally Bioavailable Pyrimidine Ether-Based Inhibitors of ELOVL1. J Med Chem 2021; 64:17777-17794. [PMID: 34871500 DOI: 10.1021/acs.jmedchem.1c00948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In our efforts to identify novel small molecule inhibitors for the treatment of adrenoleukodystrophy (ALD), we conducted a high-throughput radiometric screen for inhibitors of elongation of very long chain fatty acid 1 (ELOVL1) enzyme. We developed a series of highly potent, central nervous system (CNS)-penetrant pyrimidine ether-based compounds with favorable pharmacokinetics culminating in compound 22. Compound 22 is a selective inhibitor of ELOVL1, reducing C26:0 VLCFA synthesis in ALD patient fibroblasts and lymphocytes in vitro. Compound 22 reduced C26:0 lysophosphatidyl choline (LPC), a subtype of VLCFA, in the blood of ATP binding cassette transporter D1 (ABCD1) KO mice, a murine model of ALD to near wild-type levels. Compound 22 is a low-molecular-weight, potent ELOVL1 inhibitor that may serve as a useful tool for exploring therapeutic approaches to the treatment of ALD.
Collapse
Affiliation(s)
- Michael J Boyd
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Philip N Collier
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Michael P Clark
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Hongbo Deng
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Sarathy Kesavan
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Steven M Ronkin
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Nathan Waal
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Jian Wang
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Jingrong Cao
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Pan Li
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Jon Come
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Ioana Davies
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - John P Duffy
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - John E Cochran
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - John J Court
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Kishan Chandupatla
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Katrina L Jackson
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Francois Maltais
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Hardwin O'Dowd
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Christina Boucher
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Tony Considine
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - William P Taylor
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Hong Gao
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Ananthisrinivas Chakilam
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Juntyma Engtrakul
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Dan Crawford
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Elizabeth Doyle
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Jonathan Phillips
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Raymond Kemper
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Rebecca Swett
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - James Empfield
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Mark E Bunnage
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Paul S Charifson
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Sanjay Shivayogi Magavi
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| |
Collapse
|
14
|
Come JH, Senter TJ, Clark MP, Court JJ, Gale-Day Z, Gu W, Krueger E, Liang J, Morris M, Nanthakumar S, O'Dowd H, Maltais F, Iyer G, Andreassi J, Boucher C, Considine T, Moody CS, Taylor W, Mohanty AK, Huang Y, Zuccola H, Coll J, Bonanno KC, Gagnon KJ, Gan L, Lu F, Gao H, Chakilam A, Engtrakul J, Song B, Crawford D, Doyle E, Kramer T, Vought B, Phillips J, Kemper R, Sanders M, Swett R, Furey B, Winquist R, Bunnage ME, Jackson KL, Charifson PS, Magavi SS. Discovery and Optimization of Pyrazole Amides as Inhibitors of ELOVL1. J Med Chem 2021; 64:17753-17776. [PMID: 34748351 DOI: 10.1021/acs.jmedchem.1c00944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Accumulation of very long chain fatty acids (VLCFAs) due to defects in ATP binding cassette protein D1 (ABCD1) is thought to underlie the pathologies observed in adrenoleukodystrophy (ALD). Pursuing a substrate reduction approach based on the inhibition of elongation of very long chain fatty acid 1 enzyme (ELOVL1), we explored a series of thiazole amides that evolved into compound 27─a highly potent, central nervous system (CNS)-penetrant compound with favorable in vivo pharmacokinetics. Compound 27 selectively inhibits ELOVL1, reducing C26:0 VLCFA synthesis in ALD patient fibroblasts, lymphocytes, and microglia. In mouse models of ALD, compound 27 treatment reduced C26:0 VLCFA concentrations to near-wild-type levels in blood and up to 65% in the brain, a disease-relevant tissue. Preclinical safety findings in the skin, eye, and CNS precluded progression; the origin and relevance of these findings require further study. ELOVL1 inhibition is an effective approach for normalizing VLCFAs in models of ALD.
Collapse
Affiliation(s)
- Jon H Come
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Timothy J Senter
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Michael P Clark
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - John J Court
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Zachary Gale-Day
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Wenxin Gu
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Elaine Krueger
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Jianglin Liang
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Mark Morris
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Suganthini Nanthakumar
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Hardwin O'Dowd
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Francois Maltais
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Ganesh Iyer
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - John Andreassi
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Christina Boucher
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Tony Considine
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Cameron S Moody
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - William Taylor
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Arun K Mohanty
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Yulin Huang
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Harmon Zuccola
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Joyce Coll
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Kenneth C Bonanno
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Kevin J Gagnon
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Lu Gan
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Fan Lu
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Hong Gao
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Ananthisrinivas Chakilam
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Juntyma Engtrakul
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Bin Song
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Dan Crawford
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Elisabeth Doyle
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Tal Kramer
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Bryan Vought
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Jonathan Phillips
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Raymond Kemper
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Martin Sanders
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Rebecca Swett
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Brinley Furey
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Ray Winquist
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Mark E Bunnage
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Katrina L Jackson
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Paul S Charifson
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| | - Sanjay S Magavi
- Vertex Pharmaceuticals Incorporated, 50 Northern Ave, Boston, Massachusetts 02210, United States
| |
Collapse
|
15
|
Mallack EJ, Turk BR, Yan H, Price C, Demetres M, Moser AB, Becker C, Hollandsworth K, Adang L, Vanderver A, Van Haren K, Ruzhnikov M, Kurtzberg J, Maegawa G, Orchard PJ, Lund TC, Raymond GV, Regelmann M, Orsini JJ, Seeger E, Kemp S, Eichler F, Fatemi A. MRI surveillance of boys with X-linked adrenoleukodystrophy identified by newborn screening: Meta-analysis and consensus guidelines. J Inherit Metab Dis 2021; 44:728-739. [PMID: 33373467 PMCID: PMC8113077 DOI: 10.1002/jimd.12356] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/11/2020] [Accepted: 12/28/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Among boys with X-Linked adrenoleukodystrophy, a subset will develop childhood cerebral adrenoleukodystrophy (CCALD). CCALD is typically lethal without hematopoietic stem cell transplant before or soon after symptom onset. We sought to establish evidence-based guidelines detailing the neuroimaging surveillance of boys with neurologically asymptomatic adrenoleukodystrophy. METHODS To establish the most frequent age and diagnostic neuroimaging modality for CCALD, we completed a meta-analysis of relevant studies published between January 1, 1970 and September 10, 2019. We used the consensus development conference method to incorporate the resulting data into guidelines to inform the timing and techniques for neuroimaging surveillance. Final guideline agreement was defined as >80% consensus. RESULTS One hundred twenty-three studies met inclusion criteria yielding 1285 patients. The overall mean age of CCALD diagnosis is 7.91 years old. The median age of CCALD diagnosis calculated from individual patient data is 7.0 years old (IQR: 6.0-9.5, n = 349). Ninety percent of patients were diagnosed between 3 and 12. Conventional MRI was most frequently reported, comprised most often of T2-weighted and contrast-enhanced T1-weighted MRI. The expert panel achieved 95.7% consensus on the following surveillance parameters: (a) Obtain an MRI between 12 and 18 months old. (b) Obtain a second MRI 1 year after baseline. (c) Between 3 and 12 years old, obtain a contrast-enhanced MRI every 6 months. (d) After 12 years, obtain an annual MRI. CONCLUSION Boys with adrenoleukodystrophy identified early in life should be monitored with serial brain MRIs during the period of highest risk for conversion to CCALD.
Collapse
Affiliation(s)
- Eric J. Mallack
- Department of Pediatrics, Division of Child Neurology, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York, New York
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Bela R. Turk
- Division of Neurogenetics and The Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland
| | - Helena Yan
- Department of Pediatrics, Division of Child Neurology, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York, New York
| | - Carrie Price
- Division of Neurogenetics and The Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland
| | - Michelle Demetres
- Department of Pediatrics, Division of Child Neurology, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York, New York
| | - Ann B. Moser
- Division of Neurogenetics and The Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland
| | - Catherine Becker
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Kim Hollandsworth
- Division of Neurogenetics and The Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland
| | - Laura Adang
- Division of Neurology, Perelman School of Medicine at the University of Pennsylvania, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Adeline Vanderver
- Division of Neurology, Perelman School of Medicine at the University of Pennsylvania, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Keith Van Haren
- Department of Neurology, Stanford University School of Medicine, Lucile Packard Children’s Hospital, Stanford, California
| | - Maura Ruzhnikov
- Department of Neurology, Stanford University School of Medicine, Lucile Packard Children’s Hospital, Stanford, California
| | - Joanne Kurtzberg
- Department of Pediatrics, Duke University School of Medicine, Duke Children’s Hospital and Health Center, Durham, North Carolina
| | - Gustavo Maegawa
- Department of Pediatrics, Division of Genetics and Metabolism, University of Florida College of Medicine, University of Florida Health Shands Children’s Hospital, Gainesville, Florida
| | - Paul J. Orchard
- Department of Pediatrics, Division of Bone Marrow Transplantation, University of Minnesota Children’s Hospital, Minneapolis, Minnesota
| | - Troy C. Lund
- Department of Pediatrics, Division of Bone Marrow Transplantation, University of Minnesota Children’s Hospital, Minneapolis, Minnesota
| | - Gerald V. Raymond
- Division of Neurogenetics and The Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland
| | - Molly Regelmann
- Department of Pediatrics, Division of Endocrinology & Diabetes, Children’s Hospital at Montefiore, Bronx, New York
| | - Joseph J. Orsini
- Newborn Screening Program, NY State Department of Health, New York, New York
| | - Elisa Seeger
- Aidan Jack Seeger Foundation, Brooklyn, New York
| | - Stephan Kemp
- Department of Pediatric Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Florian Eichler
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Ali Fatemi
- Division of Neurogenetics and The Moser Center for Leukodystrophies, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
16
|
Palakuzhiyil SV, Christopher R, Chandra SR. Deciphering the modifiers for phenotypic variability of X-linked adrenoleukodystrophy. World J Biol Chem 2020; 11:99-111. [PMID: 33274015 PMCID: PMC7672940 DOI: 10.4331/wjbc.v11.i3.99] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/10/2020] [Accepted: 09/18/2020] [Indexed: 02/05/2023] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD), an inborn error of peroxisomal β-oxidation, is caused by defects in the ATP Binding Cassette Subfamily D Member 1 (ABCD1) gene. X-ALD patients may be asymptomatic or present with several clinical phenotypes varying from severe to mild, severe cerebral adrenoleuko-dystrophy to mild adrenomyeloneuropathy (AMN). Although most female heterozygotes present with AMN-like symptoms after 60 years of age, occasional cases of females with the cerebral form have been reported. Phenotypic variability has been described within the same kindreds and even among monozygotic twins. There is no association between the nature of ABCD1 mutation and the clinical phenotypes, and the molecular basis of phenotypic variability in X-ALD is yet to be resolved. Various genetic, epigenetic, and environmental influences are speculated to modify the disease onset and severity. In this review, we summarize the observations made in various studies investigating the potential modifying factors regulating the clinical manifestation of X-ALD, which could help understand the pathogenesis of the disease and develop suitable therapeutic strategies.
Collapse
Affiliation(s)
- Shruti V Palakuzhiyil
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | - Rita Christopher
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | | |
Collapse
|
17
|
Titus HE, Chen Y, Podojil JR, Robinson AP, Balabanov R, Popko B, Miller SD. Pre-clinical and Clinical Implications of "Inside-Out" vs. "Outside-In" Paradigms in Multiple Sclerosis Etiopathogenesis. Front Cell Neurosci 2020; 14:599717. [PMID: 33192332 PMCID: PMC7654287 DOI: 10.3389/fncel.2020.599717] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple Sclerosis (MS) is an immune-mediated neurological disorder, characterized by central nervous system (CNS) inflammation, oligodendrocyte loss, demyelination, and axonal degeneration. Although autoimmunity, inflammatory demyelination and neurodegeneration underlie MS, the initiating event has yet to be clarified. Effective disease modifying therapies need to both regulate the immune system and promote restoration of neuronal function, including remyelination. The challenge in developing an effective long-lived therapy for MS requires that three disease-associated targets be addressed: (1) self-tolerance must be re-established to specifically inhibit the underlying myelin-directed autoimmune pathogenic mechanisms; (2) neurons must be protected from inflammatory injury and degeneration; (3) myelin repair must be engendered by stimulating oligodendrocyte progenitors to remyelinate CNS neuronal axons. The combined use of chronic and relapsing remitting experimental autoimmune encephalomyelitis (C-EAE, R-EAE) (“outside-in”) as well as progressive diphtheria toxin A chain (DTA) and cuprizone autoimmune encephalitis (CAE) (“inside-out”) mouse models allow for the investigation and specific targeting of all three of these MS-associated disease parameters. The “outside-in” EAE models initiated by myelin-specific autoreactive CD4+ T cells allow for the evaluation of both myelin-specific tolerance in the absence or presence of neuroprotective and/or remyelinating agents. The “inside-out” mouse models of secondary inflammatory demyelination are triggered by toxin-induced oligodendrocyte loss or subtle myelin damage, which allows evaluation of novel therapeutics that could promote remyelination and neuroprotection in the CNS. Overall, utilizing these complementary pre-clinical MS models will open new avenues for developing therapeutic interventions, tackling MS from the “outside-in” and/or “inside-out”.
Collapse
Affiliation(s)
- Haley E Titus
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Yanan Chen
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Joseph R Podojil
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Cour Pharmaceutical Development Company, Inc., Northbrook, IL, United States
| | - Andrew P Robinson
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Roumen Balabanov
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Brian Popko
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Stephen D Miller
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States.,Cour Pharmaceutical Development Company, Inc., Northbrook, IL, United States.,Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
18
|
Tang H, Matteson J, Rinaldo P, Tortorelli S, Currier R, Sciortino S. The Clinical Impact of CLIR Tools toward Rapid Resolution of Post-Newborn Screening Confirmatory Testing for X-Linked Adrenoleukodystrophy in California. Int J Neonatal Screen 2020; 6:62. [PMID: 33123639 PMCID: PMC7570356 DOI: 10.3390/ijns6030062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
Since the start of X-linked adrenoleukodystrophy (ALD) newborn screening in California, more than half of the diagnosed cases were found to have an ATP binding cassette subfamily D member 1 (ABCD1) gene variant of uncertain significance (VUS). To determine retrospectively the likelihood that these were true positive cases, we used a web-based post-analytical tool in Collaborative Laboratory Integrated Reports (CLIR). Confirmatory plasma very long-chain fatty-acids (VLCFA) profiles for ALD screen positive infant boys were run through the CLIR ALD tool. We compared the distribution by ABCD1 variant classification (pathogenic, likely pathogenic, VUS, and no variant) with the CLIR tool score interpretation (non-informative, possibly ALD, likely ALD, and very likely ALD) and the current case diagnosis. The study showed that CLIR tool positive interpretations were consistent with 100% of the pathogenic and likely pathogenic variants on the ABCD1 gene if a more conservative guideline was used. The tool interpretations were also consistent with screened cases that were determined to not have disease (our no-disorder group). The CLIR tool identified 19 diagnosed ALD cases with VUS to be potential false positives, representing a 40% reduction among all diagnosed ALD cases with VUS. The reduction could be extended to 65% if a more aggressive threshold was used. Identifying such preventable false positives could alleviate the follow-up burden for patients, their families, and California Special Care Centers.
Collapse
Affiliation(s)
- Hao Tang
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA 94804, USA; (J.M.); (S.S.)
| | - Jamie Matteson
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA 94804, USA; (J.M.); (S.S.)
| | - Piero Rinaldo
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (P.R.); (S.T.)
| | - Silvia Tortorelli
- Biochemical Genetics Laboratory, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (P.R.); (S.T.)
| | - Robert Currier
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA;
| | - Stanley Sciortino
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA 94804, USA; (J.M.); (S.S.)
| |
Collapse
|
19
|
Mallack EJ, van de Stadt S, Caruso PA, Musolino PL, Sadjadi R, Engelen M, Eichler FS. Clinical and radiographic course of arrested cerebral adrenoleukodystrophy. Neurology 2020; 94:e2499-e2507. [PMID: 32482842 PMCID: PMC7455338 DOI: 10.1212/wnl.0000000000009626] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/10/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To gain insight into the natural history of arrested cerebral adrenoleukodystrophy (CALD) by quantifying the change in Neurologic Function Score (NFS) and Loes Score (LS) over time in patients whose cerebral lesions spontaneously stopped progressing. METHODS We retrospectively reviewed a series of 22 patients with arrested CALD followed longitudinally over a median time of 2.4 years (0.7-17.0 years). Primary outcomes were change in radiographic disease burden (measured by LS) and clinical symptoms (measured by NFS) between patients who never developed a contrast-enhancing lesion (gadolinium enhancement (GdE)- subgroup) and those who did (GdE+ subgroup). Secondary analyses comparing patterns of neuroanatomic involvement and lesion number, and prevalence estimates, were performed. RESULTS Cerebral lesions were first detected at a median age of 23.3 years (8.0-67.6 years) with an initial LS of 4 (0.5-9). NFS was 0.5 (0-6). Overall change in NFS or LS per year did not differ between subgroups. No patients who remained GdE- converted to a progressive CALD phenotype. The presence of contrast enhancement was associated with disease progression (r s = 0.559, p < 0.001). Four patients (18.2%) underwent step-wise progression, followed by spontaneous resolution of contrast enhancement and rearrest of disease. Three patients (13.6%) converted to progressive CALD. Nineteen patients (86.4%) had arrested CALD at the most recent follow-up. The prevalence of arrested CALD is 12.4%. CONCLUSION Arrested CALD lesions can begin in childhood, and patients are often asymptomatic early in disease. The majority of patients remain stable. However, clinical and MRI surveillance is recommended because a minority of patients undergo step-wise progression or conversion to progressive CALD.
Collapse
Affiliation(s)
- Eric J Mallack
- From the Department of Neurology (E.J.M., P.L.M., R.S., F.S.E.) and Department of Radiology (P.A.C.), Division of Neuroradiology, Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatrics (E.J.M.), Division of Child Neurology, Weill Cornell Medical College, New York-Presbyterian Hospital, NY; and Department of Pediatric Neurology (S.v.d.S., M.E.), Emma Children's Hospital, Amsterdam University Medical Center, the Netherlands
| | - Stephanie van de Stadt
- From the Department of Neurology (E.J.M., P.L.M., R.S., F.S.E.) and Department of Radiology (P.A.C.), Division of Neuroradiology, Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatrics (E.J.M.), Division of Child Neurology, Weill Cornell Medical College, New York-Presbyterian Hospital, NY; and Department of Pediatric Neurology (S.v.d.S., M.E.), Emma Children's Hospital, Amsterdam University Medical Center, the Netherlands
| | - Paul A Caruso
- From the Department of Neurology (E.J.M., P.L.M., R.S., F.S.E.) and Department of Radiology (P.A.C.), Division of Neuroradiology, Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatrics (E.J.M.), Division of Child Neurology, Weill Cornell Medical College, New York-Presbyterian Hospital, NY; and Department of Pediatric Neurology (S.v.d.S., M.E.), Emma Children's Hospital, Amsterdam University Medical Center, the Netherlands
| | - Patricia L Musolino
- From the Department of Neurology (E.J.M., P.L.M., R.S., F.S.E.) and Department of Radiology (P.A.C.), Division of Neuroradiology, Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatrics (E.J.M.), Division of Child Neurology, Weill Cornell Medical College, New York-Presbyterian Hospital, NY; and Department of Pediatric Neurology (S.v.d.S., M.E.), Emma Children's Hospital, Amsterdam University Medical Center, the Netherlands
| | - Reza Sadjadi
- From the Department of Neurology (E.J.M., P.L.M., R.S., F.S.E.) and Department of Radiology (P.A.C.), Division of Neuroradiology, Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatrics (E.J.M.), Division of Child Neurology, Weill Cornell Medical College, New York-Presbyterian Hospital, NY; and Department of Pediatric Neurology (S.v.d.S., M.E.), Emma Children's Hospital, Amsterdam University Medical Center, the Netherlands
| | - Marc Engelen
- From the Department of Neurology (E.J.M., P.L.M., R.S., F.S.E.) and Department of Radiology (P.A.C.), Division of Neuroradiology, Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatrics (E.J.M.), Division of Child Neurology, Weill Cornell Medical College, New York-Presbyterian Hospital, NY; and Department of Pediatric Neurology (S.v.d.S., M.E.), Emma Children's Hospital, Amsterdam University Medical Center, the Netherlands
| | - Florian S Eichler
- From the Department of Neurology (E.J.M., P.L.M., R.S., F.S.E.) and Department of Radiology (P.A.C.), Division of Neuroradiology, Harvard Medical School, Massachusetts General Hospital, Boston; Department of Pediatrics (E.J.M.), Division of Child Neurology, Weill Cornell Medical College, New York-Presbyterian Hospital, NY; and Department of Pediatric Neurology (S.v.d.S., M.E.), Emma Children's Hospital, Amsterdam University Medical Center, the Netherlands.
| |
Collapse
|
20
|
Choi HW, Raymond GV, Miller W. Rare Spontaneous Attenuation of Childhood Inflammatory Cerebral Adrenoleukodystrophy. JOURNAL OF PEDIATRIC NEUROLOGY 2020; 18:106-109. [DOI: 10.1055/s-0039-1677805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
AbstractX-linked adrenoleukodystrophy (ALD) is a neurodegenerative peroxisomal disorder with variable clinical phenotypes. Childhood cerebral ALD (CCALD) is at the most severe end of the disease spectrum. In CCALD, the clinical manifestations include increasing deficits in behavior, vision, hearing, coordination, and motor function, as well as seizures. Without treatment, CCALD often results in apparent vegetative state within 1 to 2 years of appearance of initial signs and symptoms. We present the case of a boy with classic inflammatory CCALD who exhibited spontaneous attenuation in disease progression. While extremely rare, spontaneous arrest of disease progression may occur in boys with inflammatory CCALD.
Collapse
Affiliation(s)
- Hyoung Won Choi
- Division of Pediatric Neurology, Department of Neurology, University of Minnesota, Minneapolis, Minnesota, United States
| | - Gerald Vincent Raymond
- Division of Pediatric Neurology, Department of Pediatrics and Neurology, Penn State University, Hershey, Pennsylvania, United States
| | - Weston Miller
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
21
|
The Landscape of Hematopoietic Stem Cell Transplant and Gene Therapy for X-Linked Adrenoleukodystrophy. Curr Treat Options Neurol 2019; 21:61. [PMID: 31768791 DOI: 10.1007/s11940-019-0605-y] [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] [Indexed: 10/25/2022]
Abstract
PURPOSE OF REVIEW To present an updated appraisal of hematopoietic stem cell transplant (HSCT) and gene therapy for X-linked adrenoleukodystrophy (ALD) in the setting of a novel, presymptomatic approach to disease. RECENT FINDINGS Outcomes in HSCT for ALD have been optimized over time due to early patient detection, improved myeloablative conditioning regimens, and adjunctive treatment for patients with advanced cerebral disease. Gene therapy has arrested disease progression in a cohort of boys with childhood cerebral ALD. New therapeutic strategies have provided the clinical basis for the implementation of Newborn Screening (NBS). With the help of advocacy groups, NBS has been implemented, allowing for MRI screening for the onset of cerebral ALD from birth. Gene therapy and optimized hematopoietic stem cell transplant for childhood CALD have changed the natural history of this previously devastating neurological disease.
Collapse
|
22
|
Liberato AP, Mallack EJ, Aziz-Bose R, Hayden D, Lauer A, Caruso PA, Musolino PL, Eichler FS. MRI brain lesions in asymptomatic boys with X-linked adrenoleukodystrophy. Neurology 2019; 92:e1698-e1708. [PMID: 30902905 PMCID: PMC6511088 DOI: 10.1212/wnl.0000000000007294] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/30/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To describe the brain MRI findings in asymptomatic patients with childhood cerebral adrenoleukodystrophy (CCALD). METHODS We retrospectively reviewed a series of biochemically or genetically confirmed cases of adrenoleukodystrophy followed at our institution between 2001 and 2015. We identified and analyzed 219 brain MRIs from 47 asymptomatic boys (median age 6.0 years). Patient age, MRI scan, and brain lesion characteristics (e.g., contrast enhancement, volume, and Loes score) were recorded. The rate of lesion growth was estimated using a linear mixed effect model. RESULTS Sixty percent of patients (28/47) showed brain lesions (median Loes score of 3.0 points; range 0.5-11). Seventy-nine percent of patients with CCALD (22/28) had contrast enhancement on first lesional or subsequent MRI. Lesion progression (Loes increase of ≥0.5 point) was seen in 50% of patients (14/28). The rate of lesion growth (mL/mo) was faster in younger patients (r = -0.745; p < 0.0001). Older patients (median age 14.4 y/o) tended to undergo spontaneous arrest of disease. Early lesions grew 46× faster when still limited to the splenium, genu of the corpus callosum, or the brainstem (p = 0.001). CONCLUSION We provide a description of CCALD lesion development in a cohort of asymptomatic boys. Understanding the early stages of CCALD is crucial to optimize treatments for children diagnosed by newborn screening.
Collapse
Affiliation(s)
- Afonso P Liberato
- From the Department of Radiology, Division of Neuroradiology (A.P.L., P.A.C.), Department of Neurology (E.J.M., R.A.-B., A.L., P.L.M., F.S.E.), and Department of Biostatistics (D.H.), Harvard Medical School, Massachusetts General Hospital, Boston; and Department of Pediatrics, Division of Child Neurology (E.J.M.), Weill Cornell Medical College, New York-Presbyterian Hospital, New York
| | - Eric J Mallack
- From the Department of Radiology, Division of Neuroradiology (A.P.L., P.A.C.), Department of Neurology (E.J.M., R.A.-B., A.L., P.L.M., F.S.E.), and Department of Biostatistics (D.H.), Harvard Medical School, Massachusetts General Hospital, Boston; and Department of Pediatrics, Division of Child Neurology (E.J.M.), Weill Cornell Medical College, New York-Presbyterian Hospital, New York
| | - Razina Aziz-Bose
- From the Department of Radiology, Division of Neuroradiology (A.P.L., P.A.C.), Department of Neurology (E.J.M., R.A.-B., A.L., P.L.M., F.S.E.), and Department of Biostatistics (D.H.), Harvard Medical School, Massachusetts General Hospital, Boston; and Department of Pediatrics, Division of Child Neurology (E.J.M.), Weill Cornell Medical College, New York-Presbyterian Hospital, New York
| | - Doug Hayden
- From the Department of Radiology, Division of Neuroradiology (A.P.L., P.A.C.), Department of Neurology (E.J.M., R.A.-B., A.L., P.L.M., F.S.E.), and Department of Biostatistics (D.H.), Harvard Medical School, Massachusetts General Hospital, Boston; and Department of Pediatrics, Division of Child Neurology (E.J.M.), Weill Cornell Medical College, New York-Presbyterian Hospital, New York
| | - Arne Lauer
- From the Department of Radiology, Division of Neuroradiology (A.P.L., P.A.C.), Department of Neurology (E.J.M., R.A.-B., A.L., P.L.M., F.S.E.), and Department of Biostatistics (D.H.), Harvard Medical School, Massachusetts General Hospital, Boston; and Department of Pediatrics, Division of Child Neurology (E.J.M.), Weill Cornell Medical College, New York-Presbyterian Hospital, New York
| | - Paul A Caruso
- From the Department of Radiology, Division of Neuroradiology (A.P.L., P.A.C.), Department of Neurology (E.J.M., R.A.-B., A.L., P.L.M., F.S.E.), and Department of Biostatistics (D.H.), Harvard Medical School, Massachusetts General Hospital, Boston; and Department of Pediatrics, Division of Child Neurology (E.J.M.), Weill Cornell Medical College, New York-Presbyterian Hospital, New York
| | - Patricia L Musolino
- From the Department of Radiology, Division of Neuroradiology (A.P.L., P.A.C.), Department of Neurology (E.J.M., R.A.-B., A.L., P.L.M., F.S.E.), and Department of Biostatistics (D.H.), Harvard Medical School, Massachusetts General Hospital, Boston; and Department of Pediatrics, Division of Child Neurology (E.J.M.), Weill Cornell Medical College, New York-Presbyterian Hospital, New York
| | - Florian S Eichler
- From the Department of Radiology, Division of Neuroradiology (A.P.L., P.A.C.), Department of Neurology (E.J.M., R.A.-B., A.L., P.L.M., F.S.E.), and Department of Biostatistics (D.H.), Harvard Medical School, Massachusetts General Hospital, Boston; and Department of Pediatrics, Division of Child Neurology (E.J.M.), Weill Cornell Medical College, New York-Presbyterian Hospital, New York.
| |
Collapse
|
23
|
Tan EY, Boelens JJ, Jones SA, Wynn RF. Hematopoietic Stem Cell Transplantation in Inborn Errors of Metabolism. Front Pediatr 2019; 7:433. [PMID: 31709204 PMCID: PMC6824291 DOI: 10.3389/fped.2019.00433] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/07/2019] [Indexed: 01/26/2023] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) has been established as an effective therapy for selected inborn errors of metabolism. The success of HSCT in metabolic disease is best exemplified through the treatment of Hurler's syndrome, a lysosomal storage disease. Through the collaborative effort of several international centers, factors that predict successful patient and transplant outcomes have been identified. In this review, we discuss the principles that underlie the use of HSCT in metabolic diseases. We consider the clinical indications, conditioning regimens, and disease-specific follow-up for HSCT in different metabolic diseases. We highlight persisting challenges in HSCT to delay progression of certain organ systems that remain refractory to HSCT and the relatively high rates of aplastic graft failure. Finally, we evaluate the variable applicability of these principles to other inherited metabolic disorders including peroxisomal, mitochondrial, and other lysosomal storage diseases.
Collapse
Affiliation(s)
- Emily Y Tan
- Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jaap Jan Boelens
- Stem Cell Transplant and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Simon A Jones
- Metabolic and Blood and Marrow Transplant Units, Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Robert F Wynn
- Metabolic and Blood and Marrow Transplant Units, Royal Manchester Children's Hospital, Manchester, United Kingdom
| |
Collapse
|
24
|
Wang J, Zhu Q, Liu H. A novel mutation in the ABCD1 gene of a Chinese patient with X-linked adrenoleukodystrophy: Case report. Medicine (Baltimore) 2018; 97:e10837. [PMID: 29794777 PMCID: PMC6392892 DOI: 10.1097/md.0000000000010837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
RATIONALE X-linked adrenoleukodystrophy (X-ALD) is the most common peroxisomal disorder, which is inherited as an X-linked recessive trait. ATP binding cassette subfamily D member 1 (ABCD1) localized to Xq28 is the only gene associated with ALD. PATIENT CONCERNS We report a case of Chinese boy with childhood cerebral ALD, who began experiencing symptoms at the age of 5 years and 2 months. Very long chain fatty acids analysis revealed high levels of C24/C22 ratio and C26/C22 ratio in the plasma. Magnetic resonance imaging (MRI) showed abnormal bilateral white matter lesions in brainstem, temporal, occipital, and parietal lobes. DIAGNOSES Direct sequencing of the ABCD1 gene identified a novel c.1502del mutation on exon 6, which causes a substitution of the 501st amino acid from methionine to serine and finally the 557th codon is changed to stop codon. INTERVENTIONS Special education and rehabilitation therapy. OUTCOMES The disease progressed rapidly and resulted in death at the age of 8 years. LESSONS Early detection of mutations in the ABCD1 gene may facilitate diagnosis, genetic counseling and potentially aid prenatal diagnosis of the disease.
Collapse
Affiliation(s)
- Jing Wang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, China
| | - Qian Zhu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, China
| | - Hongqian Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, China
| |
Collapse
|
25
|
Kemp S, Huffnagel IC, Linthorst GE, Wanders RJ, Engelen M. Adrenoleukodystrophy - neuroendocrine pathogenesis and redefinition of natural history. Nat Rev Endocrinol 2016; 12:606-15. [PMID: 27312864 DOI: 10.1038/nrendo.2016.90] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
X-Linked adrenoleukodystrophy (ALD) is a peroxisomal metabolic disorder with a highly complex clinical presentation. ALD is caused by mutations in the ABCD1 gene, which leads to the accumulation of very long-chain fatty acids in plasma and tissues. Virtually all men with ALD develop adrenal insufficiency and myelopathy. Approximately 60% of men develop progressive cerebral white matter lesions (known as cerebral ALD). However, one cannot identify these individuals until the early changes are seen using brain imaging. Women with ALD also develop myelopathy, but generally at a later age than men and adrenal insufficiency or cerebral ALD are very rare. Owing to the multisystem symptomatology of the disease, patients can be assessed by the paediatrician, general practitioner, endocrinologist or a neurologist. This Review describes current knowledge on the clinical presentation, diagnosis and treatment of ALD, and highlights gaps in our knowledge of the natural history of the disease owing to an absence of large-scale prospective cohort studies. Such studies are necessary for the identification of new prognostic biomarkers to improve care for patients with ALD, which is particularly relevant now that newborn screening for ALD is being introduced.
Collapse
Affiliation(s)
- Stephan Kemp
- Department of Pediatrics, Academisch Medisch Centrum, University of Amsterdam Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Genetic Metabolic Diseases, Academisch Medisch Centrum, University of Amsterdam Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Irene C Huffnagel
- Department of Pediatrics, Academisch Medisch Centrum, University of Amsterdam Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Pediatric Neurology, Academisch Medisch Centrum, University of Amsterdam Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Gabor E Linthorst
- Endocrinology and Metabolism, Academisch Medisch Centrum, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Ronald J Wanders
- Department of Pediatrics, Academisch Medisch Centrum, University of Amsterdam Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Genetic Metabolic Diseases, Academisch Medisch Centrum, University of Amsterdam Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Marc Engelen
- Department of Pediatrics, Academisch Medisch Centrum, University of Amsterdam Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Pediatric Neurology, Academisch Medisch Centrum, University of Amsterdam Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
26
|
van Engen CE, Ofman R, Dijkstra IME, van Goethem TJ, Verheij E, Varin J, Vidaud M, Wanders RJA, Aubourg P, Kemp S, Barbier M. CYP4F2 affects phenotypic outcome in adrenoleukodystrophy by modulating the clearance of very long-chain fatty acids. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1861-70. [PMID: 27425035 DOI: 10.1016/j.bbadis.2016.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/04/2016] [Accepted: 07/13/2016] [Indexed: 01/08/2023]
Abstract
X-linked adrenoleukodystrophy (ALD) is a severe neurodegenerative disorder caused by the accumulation of very long-chain fatty acids (VLCFA) due to mutations in the ABCD1 gene. The phenotypic spectrum ranges from a fatal cerebral demyelinating disease in childhood (cerebral ALD) to a progressive myelopathy without cerebral involvement in adulthood (adrenomyeloneuropathy). Because ABCD1 mutations have no predictive value with respect to clinical outcome a role for modifier genes was postulated. We report that the CYP4F2 polymorphism rs2108622 increases the risk of developing cerebral ALD in Caucasian patients. The rs2108622 polymorphism (c.1297G>A) results in an amino acid substitution valine for methionine at position 433 (p.V433M). Using cellular models of VLCFA accumulation, we show that p.V433M decreases the conversion of VLCFA into very long-chain dicarboxylic acids by ω-oxidation, a potential escape route for the deficient peroxisomal β-oxidation of VLCFA in ALD. Although p.V433M does not affect the catalytic activity of CYP4F2 it reduces CYP4F2 protein levels markedly. These findings open perspectives for therapeutic interventions in a disease with currently limited treatment options.
Collapse
Affiliation(s)
- Catherine E van Engen
- Laboratory Genetic Metabolic Diseases, Departments of Pediatrics and Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Rob Ofman
- Laboratory Genetic Metabolic Diseases, Departments of Pediatrics and Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Inge M E Dijkstra
- Laboratory Genetic Metabolic Diseases, Departments of Pediatrics and Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Tessa Jacobs van Goethem
- Laboratory Genetic Metabolic Diseases, Departments of Pediatrics and Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Eveline Verheij
- Laboratory Genetic Metabolic Diseases, Departments of Pediatrics and Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jennifer Varin
- INSERM U745, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris-Descartes, Sorbonne Paris Cité, Paris, France
| | - Michel Vidaud
- INSERM U745, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris-Descartes, Sorbonne Paris Cité, Paris, France
| | - Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Departments of Pediatrics and Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Patrick Aubourg
- INSERM U986, Le Kremlin-Bicêtre, Paris, France; Faculté de Médecine, Université Paris-Sud, Assistance Publique des Hôpitaux de Paris, Le Kremlin-Bicêtre, Paris, France
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Departments of Pediatrics and Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|
27
|
Kemper AR, Brosco J, Comeau AM, Green NS, Grosse SD, Jones E, Kwon JM, Lam WKK, Ojodu J, Prosser LA, Tanksley S. Newborn screening for X-linked adrenoleukodystrophy: evidence summary and advisory committee recommendation. Genet Med 2016; 19:121-126. [PMID: 27337030 DOI: 10.1038/gim.2016.68] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/15/2016] [Indexed: 11/09/2022] Open
Abstract
The secretary of the US Department of Health and Human Services in February 2016 recommended that X-linked adrenoleukodystrophy (X-ALD) be added to the recommended uniform screening panel for state newborn screening programs. This decision was informed by data presented on the accuracy of screening from New York, the only state that currently offers X-ALD newborn screening, and published and unpublished data showing health benefits of earlier treatment (hematopoietic stem cell transplantation and adrenal hormone replacement therapy) for the childhood cerebral form of X-ALD. X-ALD newborn screening also identifies individuals with later-onset disease, but poor genotype-phenotype correlation makes predicting health outcomes difficult and might increase the risk of unnecessary treatment. Few data are available regarding the harms of screening and presymptomatic identification. Significant challenges exist for implementing comprehensive X-ALD newborn screening, including incorporation of the test, coordinating follow-up diagnostic and treatment care, and coordination of extended family testing after case identification.Genet Med 19 1, 121-126.
Collapse
Affiliation(s)
- Alex R Kemper
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Jeffrey Brosco
- Department of Pediatrics, University of Miami, Coral Gables, Florida, USA
| | - Anne Marie Comeau
- Department of Pediatrics, University of Massachusetts Medical School, Jamaica Plain, Massachusetts, USA
| | - Nancy S Green
- Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Scott D Grosse
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Elizabeth Jones
- Association of Public Health Laboratories, Silver Spring, Maryland, USA
| | - Jennifer M Kwon
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Wendy K K Lam
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Jelili Ojodu
- Association of Public Health Laboratories, Silver Spring, Maryland, USA
| | - Lisa A Prosser
- Child Health Evaluation and Research Unit, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Susan Tanksley
- Texas Department of State Health Services, Austin, Texas, USA
| |
Collapse
|
28
|
Chu SS, Ye J, Zhang HW, Han LS, Qiu WJ, Gao XL, Gu XF. Eight novel mutations in the ABCD1 gene and clinical characteristics of 25 Chinese patients with X-linked adrenoleukodystrophy. World J Pediatr 2015; 11:366-73. [PMID: 26454440 DOI: 10.1007/s12519-015-0044-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/19/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND X-linked adrenoleukodystrophy (X-ALD) is a fatal neurodegenerative disease caused by mutations in the adenosine triphosphate-binding cassette D1 (ABCD1) gene. This study aimed to retrospectively investigate the clinical characteristics of 25 patients with X-ALD including members of large pedigrees, to analyze ABCD1 gene mutations, the effect of gene novel variants on ALD protein (ALDP) structure and function, and to expand gene mutation spectrum of Chinese patients. METHODS Twenty-five male patients diagnosed with X-ALD were enrolled in this study. The clinical characteristics of the patients were retrospectively summarized by reviewing medical records or telephone consultation. ABCD1 gene mutations were analyzed. The pathogenicity of novel missense variants was analyzed using cobalt constraint-based multiple protein alignment tool, polymorphism phenotyping, sorting intolerant from tolerant, Align-Grantham variation and Grantham deviation, and Swiss-Program Database Viewer 4.04 software, respectively. RESULTS Childhood cerebral form ALD (CCALD) is the most common phenotype (64%) in the 25 patients with X-ALD. The progressive deterioration of neurological and cognitive functions is the main clinical feature. The demyelination of the brain white matter and elevated plasma very long chain fatty acids (VLCFAs) were found in all patients. Different phenotypes were also presented within family members of the patients. Twenty-two different mutations including 8 novel mutations in the ABCD1 gene were identified in the 25 patients. Of the mutations, 63.6% were missense mutations and 34.8% located in exon 1. The amino acid residues of three novel missense mutations in eight species were highly conserved, and were predicted to be "probably" damaging to ALDP function. The other five novel mutations were splice, nonsense, deletion or duplication mutations. CONCLUSIONS CCALD is the most common phenotype (64%) in our patients with X-ALD. Eight novel mutations in the ABCD1 gene identified are disease-causing mutations. Brain magnetic resonance imaging and plasma VLCFA determination should be performed for the patients who present with progressive deterioration of neurological development.
Collapse
Affiliation(s)
- Shan-Shan Chu
- , Shanghai, China.,Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Ye
- , Shanghai, China. .,Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hui-Wen Zhang
- , Shanghai, China.,Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian-Shu Han
- , Shanghai, China.,Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Juan Qiu
- , Shanghai, China.,Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Lan Gao
- , Shanghai, China.,Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue-Fan Gu
- , Shanghai, China.,Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Wiesinger C, Eichler FS, Berger J. The genetic landscape of X-linked adrenoleukodystrophy: inheritance, mutations, modifier genes, and diagnosis. APPLICATION OF CLINICAL GENETICS 2015; 8:109-21. [PMID: 25999754 PMCID: PMC4427263 DOI: 10.2147/tacg.s49590] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is caused by mutations in the ABCD1 gene encoding a peroxisomal ABC transporter. In this review, we compare estimates of incidence derived from different populations in order to provide an overview of the worldwide incidence of X-ALD. X-ALD presents with heterogeneous phenotypes ranging from adrenomyeloneuropathy (AMN) to inflammatory demyelinating cerebral ALD (CALD). A large number of different mutations has been described, providing a unique opportunity for analysis of functional domains within ABC transporters. Yet the molecular basis for the heterogeneity of clinical symptoms is still largely unresolved, as no correlation between genotype and phenotype exists in X-ALD. Beyond ABCD1, environmental triggers and other genetic factors have been suggested as modifiers of the disease course. Here, we summarize the findings of numerous reports that aimed at identifying modifier genes in X-ALD and discuss potential problems and future approaches to address this issue. Different options for prenatal diagnosis are summarized, and potential pitfalls when applying next-generation sequencing approaches are discussed. Recently, the measurement of very long-chain fatty acids in lysophosphatidylcholine for the identification of peroxisomal disorders was included in newborn screening programs.
Collapse
Affiliation(s)
- Christoph Wiesinger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Florian S Eichler
- Department for Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
30
|
Kang JW, Lee SM, Koo KY, Lee YM, Nam HS, Quan Z, Kang HC. Isolated cerebellar variant of adrenoleukodystrophy with a de novo adenosine triphosphate-binding cassette D1 (ABCD1) gene mutation. Yonsei Med J 2014; 55:1157-60. [PMID: 24954351 PMCID: PMC4075381 DOI: 10.3349/ymj.2014.55.4.1157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD) shows a wide range of phenotypic expression, but clinical presentation as an isolated lesion of the cerebellar white matter and dentate nuclei has not been reported. We report an unusual presentation of X-ALD only with an isolated lesion of the cerebellar white matter and dentate nuclei. The proband, a 37-year-old man presented with bladder incontinence, slurred speech, dysmetria in all limbs, difficulties in balancing, and gait ataxia. Brain magnetic resonance imaging showed an isolated signal change of white matter around the dentate nucleus in cerebellum. With high level of very long chain fatty acid, gene study showed a de novo mutation in exon 1 at nucleotide position c.277_296dup20 (p.Ala100Cysfs*10) of the adenosine triphosphate-binding cassette D1 gene. It is advised to consider X-ALD as a differential diagnosis in patients with isolated cerebellar degeneration symptoms.
Collapse
Affiliation(s)
- Joon Won Kang
- Department of Pediatrics, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, Korea
| | - Sang Mi Lee
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyo Yeon Koo
- Department of Clinical Genetics, Yonsei University College of Medicine, Seoul, Korea
| | - Young-Mock Lee
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyo Suk Nam
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Zhejiu Quan
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hoon-Chul Kang
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
31
|
Krishna SH, McKinney AM, Lucato LT. Congenital Genetic Inborn Errors of Metabolism Presenting as an Adult or Persisting Into Adulthood: Neuroimaging in the More Common or Recognizable Disorders. Semin Ultrasound CT MR 2014; 35:160-91. [DOI: 10.1053/j.sult.2013.10.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
32
|
Berger J, Forss-Petter S, Eichler FS. Pathophysiology of X-linked adrenoleukodystrophy. Biochimie 2013; 98:135-42. [PMID: 24316281 PMCID: PMC3988840 DOI: 10.1016/j.biochi.2013.11.023] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 11/22/2013] [Indexed: 12/26/2022]
Abstract
Currently the molecular basis for the clinical heterogeneity of X-linked adrenoleukodystrophy (X-ALD) is poorly understood. The genetic bases for all different phenotypic variants of X-ALD are mutations in the gene encoding the peroxisomal ATP-binding cassette (ABC) transporter, ABCD1 (formerly adrenoleukodystrophy protein, ALDP). ABCD1 transports CoA-activated very long-chain fatty acids from the cytosol into the peroxisome for degradation. The phenotypic variability is remarkable ranging from cerebral inflammatory demyelination of childhood onset, leading to death within a few years, to adults remaining pre-symptomatic through more than five decades. There is no general genotype–phenotype correlation in X-ALD. The default manifestation of mutations in ABCD1 is adrenomyeloneuropathy, a slowly progressive dying-back axonopathy affecting both ascending and descending spinal cord tracts as well as in some cases, a peripheral neuropathy. In about 60% of male X-ALD patients, either in childhood (35–40%) or in adulthood (20%), an initial, clinically silent, myelin destabilization results in conversion to a devastating, rapidly progressive form of cerebral inflammatory demyelination. Here, ABCD1 remains a susceptibility gene, necessary but not sufficient for inflammatory demyelination to occur. Although the accumulation of very long-chain fatty acids appears to be essential for the pathomechanism of all phenotypes, the molecular mechanisms underlying these phenotypes are fundamentally different. Cell autonomous processes such as oxidative stress and energy shortage in axons as well as non-cell autonomous processes involving axon–glial interactions seem pertinent to the dying-back axonopathy. Various dynamic mechanisms may underlie the initiation of inflammation, the altered immune reactivity, the propagation of inflammation, as well as the mechanisms leading to the arrest of inflammation after hematopoietic stem cell transplantation. An improved understanding of the molecular mechanisms involved in these events is required for the development of urgently needed therapeutics. Adrenomyeloneuropathy (AMN) is proposed to be the core syndrome of X-ALD. The cerebral inflammatory demyelinating form of X-ALD is independent of AMN. The same genetic basis but fundamentally different pathomechanisms lead to AMN and cerebral ALD. Genetic, epigenetic and environmental factors modulate onset and severity of AMN and cerebral ALD.
Collapse
Affiliation(s)
- J Berger
- Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria.
| | - S Forss-Petter
- Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria
| | - F S Eichler
- Department for Neurology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street ACC 708, Boston, MA 02114, USA
| |
Collapse
|
33
|
Deib G, Poretti A, Meoded A, Cohen KJ, Raymond GV, Abromowitch M, Huisman TAGM. Onset of adreno-leukodystrophy after medulloblastoma therapy: causal connection or coincidence? JIMD Rep 2013; 2:29-32. [PMID: 23430850 DOI: 10.1007/8904_2011_39] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 04/12/2011] [Accepted: 05/03/2011] [Indexed: 11/30/2022] Open
Abstract
X-linked adreno-leukodystrophy (ALD) is a peroxisomal disorder affecting the white matter of the central nervous system and the adrenal cortex. It is caused by mutations in the ABCD1 gene encoding for a peroxisomal membrane protein. The absent genotype-phenotype correlation implies a contribution by environmental factors to explain the phenotypical heterogeneity. We report on a 4-year-old boy with a biochemically confirmed diagnosis of ALD after birth. At the age of 32 months, the additional diagnosis of a medulloblastoma was made. After treatment of the medulloblastoma, he developed active areas of demyelination representing the characteristic neuroimaging features of ALD. The clinical history of our patient supports the hypothesis that external factors, like neurosurgical intervention as part of medulloblastoma treatment, may accelerate or initiate cerebral ALD-related demyelination. A postsurgical inflammatory reaction may facilitate the inclusion of abnormal fatty acids in myelin. The opening of the blood-brain barrier following neurosurgery may enhance the recognition of previously sequestered antigens considered to play a role in ALD onset. Consequently, neurosurgical disruption of the BBB can precipitate the immune-mediated inflammatory process, which progressively destroys myelin in ALD patients. Tumor-related chemotherapy and/or radiotherapy may also play a contributing role. We suggest that X-ALD patients who undergo neurosurgical intervention need close follow-up imaging to identify active demyelination early.
Collapse
Affiliation(s)
- G Deib
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Neil E, Sivaswamy L. Case 1: A six-year-old with hyperactivity. Paediatr Child Health 2013; 18:91-2. [DOI: 10.1093/pch/18.2.91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2012] [Indexed: 11/13/2022] Open
|
35
|
Abstract
Leukodystrophies comprise a broad group of progressive, inherited disorders affecting mainly myelin. They often present after a variable period of normalcy with a variety of neurologic problems. Though the ultimate diagnosis is not found in many patients with leukodystrophies, distinctive features unique to them aid in diagnosis, treatment and prognostication. The clinical characteristics, etiologies, diagnostic testing and treatment options are reviewed in detail for some of the major leukodystrophies: X-linked adrenoleukodystrophy, Krabbe disease, metachromatic leukodystrophy, Pelizaeus-Merzbacher disease, Alexander disease, Canavan disease, megalencephalic leukoencephalopathy with subcortical cysts and vanishing white matter disease.
Collapse
Affiliation(s)
- Seth J Perlman
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | | |
Collapse
|
36
|
Berger J, Pujol A, Aubourg P, Forss-Petter S. Current and future pharmacological treatment strategies in X-linked adrenoleukodystrophy. Brain Pathol 2010; 20:845-56. [PMID: 20626746 DOI: 10.1111/j.1750-3639.2010.00393.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mutations in the ABCD1 gene cause the clinical spectrum of the neurometabolic disorder X-linked adrenoleukodystrophy/adrenomyeloneuropathy (X-ALD/AMN). Currently, the most efficient therapeutic opportunity for patients with the cerebral form of X-ALD is hematopoietic stem cell transplantation and possibly gene therapy of autologous hematopoietic stem cells. Both treatments, however, are only accessible to a subset of X-ALD patients, mainly because of the lack of markers that can predict the onset of cerebral demyelination. Moreover, for female or male X-ALD patients with AMN, currently only unsatisfying therapeutic opportunities are available. Thus, this review focuses on current and urgently needed future pharmacological therapies. The treatment of adrenal and gonadal insufficiency is well established, whereas applications of immunomodulatory and immunosuppressive drugs have failed to prevent progression of cerebral neuroinflammation. The use of Lorenzo's oil and the inefficacy of lovastatin to normalize very-long-chain fatty acids in clinical trials as well as currently experimental and therefore possible future therapeutic strategies are reviewed. The latter include pharmacological gene therapy mediated by targeted upregulation of ABCD2, the closest homolog of ABCD1, antioxidative drug treatment, small molecule histone deacetylase inhibitors such as butyrates and valproic acid, and other neuroprotective attempts.
Collapse
Affiliation(s)
- Johannes Berger
- Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| | | | | | | |
Collapse
|
37
|
Morell BK, Teichler J, Budak K, Vollenweider J, Pavlicek V. X-linked adrenoleukodystrophy presenting as Addison's disease. BMJ Case Rep 2010; 2010:2010/may13_1/bcr1120092419. [PMID: 22753300 DOI: 10.1136/bcr.11.2009.2419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
We report the case of a young man with a history of attention deficit/hyperactivity disorder and mild cognitive impairment who presented with chronic fatigue, anorexia and progressive darkening of the skin. On laboratory testing, severely depressed concentrations of morning cortisol, along with highly elevated values of adrenocorticotropic hormone (ACTH) revealed primary adrenal insufficiency as the primary cause of the patient's symptomatology. Imaging of the brain showed altered signal intensities in the parieto-occipital regions of the brain. The demonstration of increased very long chain fatty acids (VLCFA) established the diagnosis of adolescent X-linked adrenoleukodystrophy (X-ALD). Presenting at an advanced yet slowly progressive stage the patient was not a suitable candidate for haematopoietic stem cell transplantation (HSCT), and treatment focused on hormone replacement therapy, family counselling and supportive care. On follow-up visits within the following year, fatigue had diminished and there was no evidence of progressive neurological deficits. However, exacerbation of the psychiatric symptomatology resulted in admittance to a psychiatric ward.
Collapse
Affiliation(s)
- Bernhard Kaspar Morell
- Kantonsspital Münsterlingen, Internal Medicine, Postfach, Münsterlingen, 8596, Switzerland.
| | | | | | | | | |
Collapse
|
38
|
Liu J, Sabeva NS, Bhatnagar S, Li XA, Pujol A, Graf GA. ABCD2 is abundant in adipose tissue and opposes the accumulation of dietary erucic acid (C22:1) in fat. J Lipid Res 2010; 51:162-8. [PMID: 19556607 DOI: 10.1194/jlr.m900237-jlr200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ATP binding cassette transporter, ABCD2 (D2), is a peroxisomal protein whose mRNA has been detected in the adrenal, brain, liver, and fat. Although the role of this transporter in neural tissues has been studied, its function in adipose tissue remains unexplored. The level of immunoreactive D2 in epididymal fat is >50-fold of that found in brain or adrenal. D2 is highly enriched in adipocytes and is upregulated during adipogenesis but is not essential for adipocyte differentiation or lipid accumulation in day 13.5 mouse embryonic fibroblasts isolated from D2-deficient (D2(-/-)) mice. Although no differences were appreciated in differentiation percentage, total lipid accumulation was greater in D2(-/-) adipocytes compared with the wild type. These results were consistent with in vivo observations in which no significant differences in adiposity or adipocyte diameter between wild-type and D2(-/-) mice were observed. D2(-/-) adipose tissue showed an increase in the abundance of 20:1 and 22:1 fatty acids. When mice were challenged with a diet enriched in erucic acid (22:1), this lipid accumulated in the adipose tissue in a gene-dosage-dependent manner. In conclusion, D2 is a sterol regulatory element binding protein target gene that is highly abundant in fat and opposes the accumulation of dietary lipids generally absent from the triglyceride storage pool within adipose tissue.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Pharmaceutical Sciences and Cardiovascular Research Center, Lexington, Kentucky, USA
| | | | | | | | | | | |
Collapse
|
39
|
Raymond GV, Seidman R, Monteith TS, Kolodny E, Sathe S, Mahmood A, Powers JM. Head trauma can initiate the onset of adreno-leukodystrophy. J Neurol Sci 2009; 290:70-4. [PMID: 19945717 DOI: 10.1016/j.jns.2009.11.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 10/14/2009] [Accepted: 11/10/2009] [Indexed: 11/17/2022]
Abstract
X-linked adreno-leukodystrophy and its adult variant, adrenomyeloneuropathy, are caused by mutations in ABCD1 that encodes a peroxisomal membrane protein of unknown physiological significance. In spite of identical mutations, they can have markedly divergent neurological and neuropathologic characteristics. Adreno-leukodystrophy classically presents in normal boys with mild neuropsychiatric features, which progress to frank neurological signs, the vegetative state and death in approximately three years. Adrenomyeloneuropathy typically affects young men with spastic paraparesis and sensory ataxia that can progress over decades. The neuropathologic correlate for adreno-leukodystrophy is severe inflammatory demyelination of posterior cerebral white matter, while a chronic distal axonopathy of spinal cord and peripheral nerve occurs in adrenomyeloneuropathy. Consequently, both modifier genes and environmental factors have been implicated in their pathogeneses. We report five cases of adreno-leukodystrophy whose onsets were initiated by moderate to severe head trauma, two of whom were conversions from adrenomyeloneuropathy. Their clinical courses were rapidly incapacitating, short (i.e., weeks to a few years) and fatal due to marked cerebral inflammatory demyelination. These cases, in concert with several previous reports, indicate that head trauma is one environmental factor that can have a profoundly deleterious effect on those genetically at risk for, or with milder clinical phenotypes of, this disease. Avoidance of potential head trauma and a rapid response to episodes of moderate to severe head trauma in this patient population seem prudent.
Collapse
|
40
|
Singh I, Singh AK, Contreras MA. Peroxisomal dysfunction in inflammatory childhood white matter disorders: an unexpected contributor to neuropathology. J Child Neurol 2009; 24:1147-57. [PMID: 19605772 PMCID: PMC3077730 DOI: 10.1177/0883073809338327] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The peroxisome, an ubiquitous subcellular organelle, plays an important function in cellular metabolism, and its importance for human health is underscored by the identification of fatal disorders caused by genetic abnormalities. Recent findings indicate that peroxisomal dysfunction is not only restricted to inherited peroxisomal diseases but also to disease processes associated with generation of inflammatory mediators that downregulate cellular peroxisomal homeostasis. Evidence indicates that leukodystrophies (i.e. X-linked adrenoleukodystrophy, globoid cell leukodystrophy, and periventricular leukomalacia) may share common denominators in the development and progression of the inflammatory process and thus in the dysfunctions of peroxisomes. Dysfunctions of peroxisomes may therefore contribute in part to white matter disease and to the mental and physical disabilities that develop in patients affected by these diseases.
Collapse
Affiliation(s)
- Inderjit Singh
- Department of Pediatrics, Division of Developmental Neurogenetics, Charles Darby Children's Research Institute, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | | | |
Collapse
|
41
|
Abstract
Most of the studies indicate that there is as yet no complete cure for X-ALD. However, methods of the treatment seem to slow rather than treat the disease. One method is the use of Lorenzo's oil in conjunction with a low fat diet, which may help in cerebral X-ALD. X-ALD is in very close resemblance to another neurodegenerative disease, amyotrophic lateral sclerosis (ALS). One of the believed pathomechanisms of ALS is oxidative stress; therefore, this article's emphasis on the role of reactive oxygen species in X-ALD. The aim of the present study was to review the literature concerning the advances in the treatment of X-adrenoleukodystrophy (X-ALD, OMIM # 300100) in the last two decades and to shed more light on the link between oxidative stress and X-ALD. This review article may point to a deficit in reactive oxygen species (ROS) scavenging and/or ROS overproduction being involved in the aetiopathology of these neurodegenerative diseases. Consequently, one of the useful neuronal rescue strategies could be the treatment with antioxidant agents.
Collapse
Affiliation(s)
- Mohamed A Al-Omar
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
42
|
Di Benedetto R, Denti MA, Salvati S, Attorri L, Di Biase A. PMP70 knock-down generates oxidative stress and pro-inflammatory cytokine production in C6 glial cells. Neurochem Int 2008; 54:37-42. [PMID: 18992293 DOI: 10.1016/j.neuint.2008.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Revised: 10/03/2008] [Accepted: 10/08/2008] [Indexed: 11/28/2022]
Abstract
By using RNA interference (RNAi) in rat C6 glial cells, we previously generated the cell line abcd3kd in which the peroxisomal half-transporter PMP70 was stably knocked-down. The observations that abcd3kd cells had peroxisomal beta-oxidation impairment and an increase of hexacosenoic acid in cholesterol ester fraction, indicated an overlapping function of PMP70 with adrenoleukodystrophy protein (ALDP), the peroxisomal half-transporters involved in X-linked adrenoleukodystrophy (X-ALD). The objective of the present study was to investigate whether PMP70 could affect some oxidative and inflammatory parameters, since many findings indicate oxidative damage in the brain of ALD patients and inflammation is a hallmark of the cerebral forms of X-ALD. We thus measured parameters indicative of oxidative stress, the expression or activity of antioxidant enzymes, and the production of some pro-inflammatory cytokines. Our results show that, due to inducible nitric oxide synthase up-regulation, abcd3kd cell line produces higher levels of nitrites than native C6 cells. The enhanced production of superoxide and thiobarbituric acid-reactive substances, the increased expression of mitochondrial superoxide dismutase, and the reduction of catalase and glutathione peroxidase activities confirm the presence of an oxidative process. We then measured the concentrations of TNFalpha, IFNgamma, and IL-12 and we observed that abcd3kd cells produce higher amounts of pro-inflammatory cytokines compared to native C6 cells. By using neutralizing antibodies against IL-12, not only inflammatory parameters significantly decrease, but nitrite and superoxide production is also affected. This demonstrates that oxidative status of abcd3kd cells is not a direct PMP70 knock-down consequence, but depends on IL-12 release. The scenery induced by the knock-down of PMP70 in C6 cells recall the oxidative and inflammatory status observed in human X-ALD and thus reinforce the idea that PMP70 could affect the clinical course of the disease.
Collapse
Affiliation(s)
- Rita Di Benedetto
- Department of Food Science, Nutrition and Health, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | |
Collapse
|
43
|
Moser HW, Mahmood A, Raymond GV. X-linked adrenoleukodystrophy. ACTA ACUST UNITED AC 2007; 3:140-51. [PMID: 17342190 DOI: 10.1038/ncpneuro0421] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Accepted: 12/15/2006] [Indexed: 12/26/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is caused by a defect in the gene ABCD1, which maps to Xq28 and codes for a peroxisomal membrane protein that is a member of the ATP-binding cassette transporter superfamily. X-ALD is panethnic and affects approximately 1:20,000 males. Phenotypes include the rapidly progressive childhood, adolescent, and adult cerebral forms; adrenomyeloneuropathy, which presents as slowly progressive paraparesis in adults; and Addison disease without neurologic manifestations. These phenotypes are frequently misdiagnosed, respectively, as attention-deficit hyperactivity disorder (ADHD), multiple sclerosis, or idiopathic Addison disease. Approximately 50% of female carriers develop a spastic paraparesis secondary to myelopathic changes similar to adrenomyeloneuropathy. Assays of very long chain fatty acids in plasma, cultured chorion villus cells and amniocytes, and mutation analysis permit presymptomatic and prenatal diagnosis, as well as carrier identification. The timely use of these assays is essential for genetic counseling and therapy. Early diagnosis and treatment can prevent overt Addison disease, and significantly reduce the frequency of the severe childhood cerebral phenotype. A promising new method for mass newborn screening has been developed, the implementation of which will have a profound effect on the diagnosis and therapy of X-ALD.
Collapse
Affiliation(s)
- Hugo W Moser
- Neurogenetics Research Center, Kennedy Krieger Institute, 707 North Broadway, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
44
|
Berger J, Gärtner J. X-linked adrenoleukodystrophy: clinical, biochemical and pathogenetic aspects. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1763:1721-32. [PMID: 16949688 DOI: 10.1016/j.bbamcr.2006.07.010] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Accepted: 07/24/2006] [Indexed: 11/17/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a clinically heterogeneous disorder ranging from the severe childhood cerebral form to asymptomatic persons. The overall incidence is 1:16,800 including hemizygotes as well as heterozygotes. The principal molecular defect is due to inborn mutations in the ABCD1 gene encoding the adrenoleukodystrophy protein (ALDP), a transporter in the peroxisome membrane. ALDP is involved in the transport of substrates from the cytoplasm into the peroxisomal lumen. ALDP defects lead to characteristic accumulation of saturated very long-chain fatty acids, the diagnostic disease marker. The pathogenesis is unclear. Different molecular mechanisms seem to induce inflammatory demyelination, neurodegeneration and adrenocortical insufficiency involving the primary ABCD1 defect, environmental factors and modifier genes. Important information has been derived from the X-ALD mouse models; species differences however complicate the interpretation of results. So far, bone marrow transplantation is the only effective long-term treatment for childhood cerebral X-ALD, however, only when performed at an early-stage of disease. Urgently needed novel therapeutic strategies are under consideration ranging from dietary approaches to gene therapy.
Collapse
Affiliation(s)
- Johannes Berger
- Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria.
| | | |
Collapse
|
45
|
Abstract
Current therapies for X-linked adrenoleukodystrophy (X-ALD) include replacement therapy with adrenal steroids, which is mandatory for all patients with impaired adrenal function but does not alter neurological progression significantly; dietary therapy with "Lorenzo's Oil," which appears to have a preventive effect in asymptomatic boys whose brain MRI is normal; and hematopoietic stem cell transplantation in patients in the early stage of the cerebral inflammatory phenotype. Application of these interventions requires careful assessment of the patients' phenotype, which often changes over time. Family screening provides important opportunities for disease prevention.
Collapse
Affiliation(s)
- Hugo W Moser
- Kennedy Krieger Institute, Johns Hopkins University, 707 North Broadway, Baltimore, MD 21205, USA.
| |
Collapse
|
46
|
|
47
|
LEWIS RICHARDA, SUMNER AUSTINJ. Electrophysiologic Features of Inherited Demyelinating Neuropathies: A Reappraisal. Ann N Y Acad Sci 2006; 883:321-335. [DOI: 10.1111/j.1749-6632.1999.tb08594.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
48
|
Heinzer AK, McGuinness MC, Lu JF, Stine OC, Wei H, Van der Vlies M, Dong GX, Powers J, Watkins PA, Smith KD. Mouse models and genetic modifiers in X-linked adrenoleukodystrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2004; 544:75-93. [PMID: 14713218 DOI: 10.1007/978-1-4419-9072-3_12] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Ann K Heinzer
- The Kennedy Krieger Institute, The Johns Hopkins School of Medicine, 707 North Broadway, Baltimore, MD, 21205, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Vargas CR, Wajner M, Sirtori LR, Goulart L, Chiochetta M, Coelho D, Latini A, Llesuy S, Bello-Klein A, Giugliani R, Deon M, Mello CF. Evidence that oxidative stress is increased in patients with X-linked adrenoleukodystrophy. Biochim Biophys Acta Mol Basis Dis 2004; 1688:26-32. [PMID: 14732478 DOI: 10.1016/j.bbadis.2003.10.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a hereditary disorder of peroxisomal metabolism biochemically characterized by the accumulation of very long chain fatty acids (VLCFA), particularly hexacosanoic acid (C26:0) and tetracosanoic acid (C24:0) in different tissues and in biological fluids. The disease is clinically characterized by central and peripheral demyelination and adrenal insufficiency, which is closely related to the increased concentrations of these fatty acids. However, the mechanisms underlying the brain damage in X-ALD are poorly known. Considering that free radical generation is involved in various neurodegenerative disorders, like Parkinson disease, multiple sclerosis and Alzheimer's disease, in the present study we evaluated various oxidative stress parameters, namely chemiluminescence, thiobarbituric acid reactive species (TBA-RS), total radical-trapping antioxidant potential (TRAP), and total antioxidant reactivity (TAR) in plasma of X-ALD patients, as well as the activities of the antioxidant enzymes catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPx) in erythrocytes and fibroblasts from these patients. It was verified a significant increase of plasma chemiluminescence and TBA-RS, reflecting induction of lipid peroxidation, as well as a decrease of plasma TAR, indicating a deficient capacity to rapidly handle an increase of reactive species. We also observed a significant increase of erythrocytes GPx activity and of catalase and SOD activities in fibroblasts from the patients studied. It is therefore proposed that oxidative stress may be involved in pathophysiology of X-ALD.
Collapse
Affiliation(s)
- C R Vargas
- Department of Clinical Analysis, Pharmacy Faculty, UFRGS, Rua Ramiro Barcelos, 2350 CEP 90.035-003, Porto Alegre, RS, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Burns TM, Ryan MM, Darras B, Jones HR. Current therapeutic strategies for patients with polyneuropathies secondary to inherited metabolic disorders. Mayo Clin Proc 2003; 78:858-68. [PMID: 12839082 DOI: 10.4065/78.7.858] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Supportive care, symptomatic treatment, and patient education should be provided for patients with inherited or acquired polyneuropathies. In addition, specific treatment is available for many of the acquired polyneuropathies. Genetic counseling is valuable for many patients with inherited polyneuropathies, but only rarely is specific treatment an option for these patients. However, specific treatments are available for many of the rare and devastating systemic disorders associated with polyneuropathies. Thus, clinicians must promptly diagnose these inherited disorders so that specific treatment may be initiated. The clinical features of these rare inherited disorders are emphasized.
Collapse
Affiliation(s)
- Ted M Burns
- Department of Neurology, University of Virginia, Charlottesville 22908, USA
| | | | | | | |
Collapse
|