1
|
Rayannavar A, Billington CJ, Tryon R, Kaye T, Gupta A, Lund TC, Lteif A, Adriatico K, Orchard PJ, Miller BS, Pillai NR. Insights From Minnesota on Newborn Screening for Adrenoleukodystrophy: A 5-Year Update. Am J Med Genet A 2025; 197:e63995. [PMID: 39803877 DOI: 10.1002/ajmg.a.63995] [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: 10/10/2024] [Revised: 12/23/2024] [Accepted: 01/04/2025] [Indexed: 04/05/2025]
Abstract
Our objectives are to report on the outcomes of adrenal insufficiency (AI) and cerebral ALD (cALD) in children diagnosed with X-linked adrenoleukodystrophy (ALD) identified by newborn screening (NBS) in Minnesota in the first 5 years following initiation of NBS in 02/2017. A retrospective chart review was conducted for children diagnosed with ALD via Minnesota NBS from 02/06/2017 through 02/06/2022. Data reviewed included newborn screening data, diagnostic very long chain fatty acid levels, ABCD1 molecular testing results, serial measurements of ACTH and cortisol, and serial brain MRI results. Thirty-two boys and 11 girls were molecularly and/or biochemically confirmed to have ALD. Of these 32 boys, six (2-7 years; median age:18 months) developed AI. Two boys developed cALD and underwent stem cell transplantation, one of whom also has been diagnosed with AI. All the pathogenic/likely pathogenic variants detected during the first 5 years had initial C26:0 lysophosphatidylcholine (C26:0 lysoPC) values over 0.3 μmol/L at the time of newborn screening. The addition of ALD to NBS in Minnesota has allowed for early detection of asymptomatic AI in six young patients and asymptomatic cALD in two patients. Data from our study shows a positive correlation between high newborn screening LysoPC levels and variant pathogenicity.
Collapse
Affiliation(s)
- Arpana Rayannavar
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- M Health Fairview Masonic Children's Hospital, Minneapolis, Minnesota, USA
| | - Charles J Billington
- M Health Fairview Masonic Children's Hospital, Minneapolis, Minnesota, USA
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rebecca Tryon
- M Health Fairview Masonic Children's Hospital, Minneapolis, Minnesota, USA
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Tory Kaye
- Minnesota Department of Health, Saint Paul, Minnesota, USA
| | - Ashish Gupta
- M Health Fairview Masonic Children's Hospital, Minneapolis, Minnesota, USA
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Troy C Lund
- M Health Fairview Masonic Children's Hospital, Minneapolis, Minnesota, USA
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Aida Lteif
- Division of Pediatric Endocrinology and Metabolism, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Katherine Adriatico
- Department of Genetics and Genomic Medicine, Children's Minnesota, Minneapolis, Minnesota, USA
| | - Paul J Orchard
- M Health Fairview Masonic Children's Hospital, Minneapolis, Minnesota, USA
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Bradley S Miller
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- M Health Fairview Masonic Children's Hospital, Minneapolis, Minnesota, USA
| | - Nishitha R Pillai
- M Health Fairview Masonic Children's Hospital, Minneapolis, Minnesota, USA
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
2
|
Dong SW, Xiao LM, Sun YH, Li GH, Xie YX, Wang MW, Wang N, Chen WJ, Chen HZ. Novel ABCD1 Variants in X-Linked Adrenoleukodystrophy. Clin Genet 2025. [PMID: 40210590 DOI: 10.1111/cge.14752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/12/2025]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a neurodegenerative disorder caused by mutations in the ABCD1 gene. We reported the clinical features and genetic findings of 17 X-ALD patients. Fifteen variants were identified, including five novel mutations: c.700dupC (p.Arg234Profs*67), c.743G>A (p.Gly248Asp), c.1469_1471delTGG (p.Val490del), c.1577C>A (p.Thr526Lys), and c.1658T>C (p.Leu553Pro), which were reported for the first time in X-ALD patients. Functional analysis confirmed the pathogenicity of novel variants at the protein and subcellular localization level. The p.Arg234Profs*67 mutant protein was undetectable, likely due to NMD-mediated mRNA degradation. This study expands the mutation spectrum and clinical profile of X-ALD, suggesting a potential correlation between the extent of protein dysfunction and disease severity.
Collapse
Affiliation(s)
- Sen-Wei Dong
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Li-Mei Xiao
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yu-Hao Sun
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Gui-He Li
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ying-Xuan Xie
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Meng-Wen Wang
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ning Wang
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Wan-Jin Chen
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Hai-Zhu Chen
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Neuroscience and Fujian Key Laboratory of Molecular Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
3
|
Engelen M, Kemp S. Newborn Screening for Adrenoleukodystrophy. JAMA Pediatr 2025; 179:365-366. [PMID: 39928328 DOI: 10.1001/jamapediatrics.2024.6771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Affiliation(s)
- Marc Engelen
- Department of Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
4
|
Zhou X, Ma CY, Zhang X, Xu X, Duan F, Kou M, Liu H, Zeng L, Guo L, Chen S, Chen L, Li Z, Luo J, Wu J, Li Z, Li Z, Sui T, Yuan P, Lin Z, Chen H, Lai L, Lian Q. Development of a rabbit model for adrenoleukodystrophy: A pilot study on gene therapy using rAAV9. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102469. [PMID: 40027885 PMCID: PMC11872110 DOI: 10.1016/j.omtn.2025.102469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a common peroxisomal disorder caused by mutations in the ABCD1 gene, leading to the accumulation of very long-chain fatty acids (VLCFAs). This progressive neurodegenerative disease manifests in three primary forms: childhood-acquired cerebral demyelination (CALD), adult myelopathy (AMN), and primary adrenal cortical insufficiency. Bone marrow transplantation effectively halts disease progression only in the early stages of CALD. A thorough investigation of the pathophysiology of X-ALD has been hampered by the lack of a reliable animal model. Valid animal models of X-ALD are urgently needed. To address this, we used CRISPR-Cas9 technology to knock out the ABCD1 gene and established a novel rabbit model of X-ALD. The mutants exhibited elevated serum levels of hexacosanoic acid (C26:0), lignoceric acid (C24:0), and an increased C26:0/C22:0 ratio, as well as significant white matter demyelination in the brain and spinal cord. We also investigated rAAV9-based gene therapy in this model and found a significant reduction in VLCFAs. This study introduces CRISPR-Cas9-mediated ABCD1 gene knockout rabbits as a novel animal model. It comprehensively evaluates the short-term outcomes and safety of rAAV-based gene therapy for X-ALD, providing a promising approach to explore the molecular and pharmacological mechanisms of the disease.
Collapse
Affiliation(s)
- Xiaoya Zhou
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Chui-Yan Ma
- Center for Translational Stem Cell Biology, Hong Kong, China; HKUMed Laboratory of Cellular Therapeutics, University of Hong Kong, Hong Kong 999077, China
| | - Xiaoxian Zhang
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Xianchuan Xu
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Fuyu Duan
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Meng Kou
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Hongsheng Liu
- Department of Radiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Liang Zeng
- Department of Pathology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Liyan Guo
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Shaoxiang Chen
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Li Chen
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Ziyue Li
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Jie Luo
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
| | - Jieying Wu
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Zhejin Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhanjun Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Tingting Sui
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Ping Yuan
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Zhijian Lin
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Liangxue Lai
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Qizhou Lian
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- CAS Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
- Center for Translational Stem Cell Biology, Hong Kong, China; HKUMed Laboratory of Cellular Therapeutics, University of Hong Kong, Hong Kong 999077, China
- Department of Surgery, The University of Hong Kong Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
5
|
Porcari GS, Collyer JW, Adang LA, Rajan DS. Current Advances and Challenges in Gene Therapies for Neurologic Disorders: A Review for the Clinician. Neurol Genet 2025; 11:e200229. [PMID: 39810751 PMCID: PMC11731373 DOI: 10.1212/nxg.0000000000200229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/12/2024] [Indexed: 01/16/2025]
Abstract
Over 300 million people globally are affected by rare diseases, many of which present predominantly with neurologic symptoms. Rare neurologic disorders pose significant diagnostic and therapeutic challenges including delayed diagnoses, limited treatment options, and a shortage of specialists. However, advancements in diagnostics, particularly next-generation sequencing and expansion of newborn screening, have significantly shortened the time to diagnosis for many of these disorders. Concurrently, the past decade has witnessed exponential development of new treatments for rare neurologic diseases, with several approved gene therapies and more trials under way. A range of targeted therapies now offers hope for not only symptomatic management but also for disease modification. As treatments transition from clinical trials to clinical practice, the responsibility of identifying and monitoring patients may increasingly fall on the general neurologists. This evolving therapeutic landscape highlights the urgent need to enhance our understanding of this new class of medications and the details on clinical eligibility and monitoring of patients with diseases that have approved gene therapies. This article provides a comprehensive overview of gene-targeted therapies currently available for neurologic disorders, with a focus on their mechanisms, challenges, and post-treatment considerations.
Collapse
|
6
|
Ferrer RM, Jaspers YRJ, Dijkstra IME, Breeuwsma N, van Klinken J, Romero C, Engelen M, Kemp S, Heine VM. Altered lipid profile and reduced neuronal support in human induced pluripotent stem cell-derived astrocytes from adrenoleukodystrophy patients. J Inherit Metab Dis 2025; 48:e12832. [PMID: 39704488 PMCID: PMC11660744 DOI: 10.1002/jimd.12832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
X-linked adrenoleukodystrophy (ALD) is a peroxisomal disorder resulting from pathogenic variants in the ABCD1 gene that primarily affects the nervous system and is characterized by progressive axonal degeneration in the spinal cord and peripheral nerves and leukodystrophy. Dysfunction of peroxisomal very long-chain fatty acid (VLCFA) degradation has been implicated in ALD pathology, but the impact on astrocytes, which critically support neuronal function, remains poorly understood. Fibroblasts from four ALD patients were reprogrammed to generate human-induced pluripotent stem cells (hiPSC). hiPSC-derived astrocytes were generated to study the impact of ALD on astrocytic fatty acid homeostasis. Our study reveals significant changes in the lipidome of ALD hiPSC-derived astrocytes, characterized by an enrichment of VLCFAs across multiple lipid classes, including triacylglycerols, cholesteryl esters, and phosphatidylcholines. Importantly, ALD hiPSC-derived astrocytes not only exhibit intrinsic lipid dysregulation but also affect the dendritic tree complexity of neurons in co-culture systems. These findings highlight the cell-autonomous effects of pathogenic variants in the ABCD1 protein on astrocytes and their microenvironment, shed light on potential mechanisms underlying ALD neuropathology, and underscore the critical role of astrocytes in neuronal health.
Collapse
Affiliation(s)
- Roberto Montoro Ferrer
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Pediatric NeurologyEmma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, University of AmsterdamAmsterdamThe Netherlands
- Department of Complex Trait GeneticsCentre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Yorrick R. J. Jaspers
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Inge M. E. Dijkstra
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Nicole Breeuwsma
- Department of Child and Adolescence PsychiatryEmma Children's Hospital, Amsterdam UMC Location, Vrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Jan‐Bert van Klinken
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
- Core Facility MetabolomicsAmsterdam UMC Location, University of AmsterdamAmsterdamThe Netherlands
| | - Cato Romero
- Department of Complex Trait GeneticsCentre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Child and Adolescence PsychiatryEmma Children's Hospital, Amsterdam UMC Location, Vrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| | - Marc Engelen
- Department of Pediatric NeurologyEmma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, University of AmsterdamAmsterdamThe Netherlands
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Vivi M. Heine
- Department of Complex Trait GeneticsCentre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Child and Adolescence PsychiatryEmma Children's Hospital, Amsterdam UMC Location, Vrije Universiteit Amsterdam, Amsterdam NeuroscienceAmsterdamThe Netherlands
| |
Collapse
|
7
|
Zhu G, Zhou H, Ma Y, Liu R, Nie X, Qi W, Hao L, Guo X. A case of X-Linked adrenoleukodystrophy caused by a novel mutation with singular clinical manifestation: unilateral lower limb weakness. Neurol Sci 2025; 46:473-477. [PMID: 39436520 DOI: 10.1007/s10072-024-07828-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is caused by mutations in the ABCD1 gene, leading to the accumulation of very long-chain fatty acids (VLCFAs) in plasma and tissues. It primarily affects the central nervous system white matter and the adrenal cortex. Clinical manifestations include myeloneuropathy, leukodystrophy, and adrenal insufficiency. Reliable methods for diagnosis include VLCFAs and genetic testing. We report the case of a 31-year-old male X-ALD patient who mainly presented with unilateral lower limb weakness. Adrenal insufficiency was not observed, and there was no evidence of peripheral nerve involvement in nerve conduction studies. MRI revealed only mild atrophy of thoracic spinal cord without other relevant abnormalities. Ultimately, Next-Generation Sequencing (NGS) and VLCFAs testing confirmed the diagnosis of X-ALD, and the NGS indicated a novel missense mutation.
Collapse
Affiliation(s)
- Geke Zhu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Han Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yongbo Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Rui Liu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiangtao Nie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wenjing Qi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lei Hao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xiuming Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
8
|
Vasireddy V, Maguire CA, Anderson DW, Ng C, Gong Y, Eichler F, Fourcade S, Guilera C, Onieva A, Sanchez A, Leal-Julià M, Verdés S, Dijkstra IM, Kemp S, Park H, Lutz T, Clark SW, Bosch A, Pujol A, Kozarsky K. An in vitro and in vivo efficacy evaluation of gene therapy candidate SBT101 in mouse models of adrenomyeloneuropathy and in NHPs. Mol Ther Methods Clin Dev 2024; 32:101354. [PMID: 39524975 PMCID: PMC11550353 DOI: 10.1016/j.omtm.2024.101354] [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: 04/30/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
Adrenomyeloneuropathy is a progressive neurodegenerative disease caused by pathogenic variants in the ABCD1 gene, resulting in very-long-chain fatty acid (VLCFA) accumulation that leads to dying-back axonopathy. Our candidate gene therapy, SBT101 (AAV9-human ABCD1 [hABCD1]), aims to ameliorate pathology by delivering functional copies of hABCD1 to the spinal cord. Transduced cells produce functional ABCD1 protein, thereby repairing the underlying biochemical defect. In vitro and in vivo mouse studies were conducted to assess the biochemical and functional efficacy of SBT101 and show effective delivery to target tissues involved in the disease pathology: spinal cord and dorsal root ganglia. Administration of SBT101 to mixed glial cell cultures from Abcd1-Null mice, and to male Abcd1 knockout (Abcd1 -/y ) and double-knockout (Abcd1 -/y /Abcd2 -/- ) mice led to increased hABCD1 production and reduced VLCFA. Double-knockout mice also exhibited improved grip strength. Furthermore, we conducted biodistribution and safety assessments in nonhuman primates. Six-hour intrathecal lumbar infusions demonstrated effective transduction throughout target tissues, supporting the clinical feasibility of the procedure. SBT101 was well tolerated, with no observed SBT101-related mortality or clinical signs. These findings not only provide preclinical efficacy data for SBT101 but also inform clinically relevant SBT101 dose selection for patients with adrenomyeloneuropathy.
Collapse
Affiliation(s)
| | - Casey A. Maguire
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Carrie Ng
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yi Gong
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Stéphane Fourcade
- Neurometabolic Disease Laboratory, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospital Duran i Reynalds, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBERER), Institudo de Salud Carlos III (ISCIII), Madrid, Spain
| | - Cristina Guilera
- Neurometabolic Disease Laboratory, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospital Duran i Reynalds, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBERER), Institudo de Salud Carlos III (ISCIII), Madrid, Spain
| | - Andrea Onieva
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall D’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Angela Sanchez
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall D’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Marc Leal-Julià
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall D’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Sergi Verdés
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall D’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Inge M.E. Dijkstra
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | | | - Tiffany Lutz
- SwanBio Therapeutics, Inc., Philadelphia, PA, USA
| | | | - Assumpció Bosch
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall D’Hebron Institut de Recerca (VHIR), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Institudo de Salud Carlos III (ISCIII), Madrid, Spain
| | - Aurora Pujol
- Neurometabolic Disease Laboratory, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospital Duran i Reynalds, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBERER), Institudo de Salud Carlos III (ISCIII), Madrid, Spain
- Catalan Institution of Research and Advanced Studies (ICREA), 08010 Barcelona, Catalonia, Spain
| | | |
Collapse
|
9
|
Zuo X, Chen Z. From gene to therapy: a review of deciphering the role of ABCD1 in combating X-Linked adrenoleukodystrophy. Lipids Health Dis 2024; 23:369. [PMID: 39529100 PMCID: PMC11552335 DOI: 10.1186/s12944-024-02361-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a severe genetic disorder caused by ABCD1 mutations, resulting in the buildup of very-long-chain fatty acids, leading to significant neurological decline and adrenal insufficiency. Despite advancements in understanding the mechanisms of X-ALD, its pathophysiology remains incompletely understood, complicating the development of effective treatments. This review provides a comprehensive overview of X-ALD, with a focus on the genetic and biochemical roles of ABCD1 and the impacts of its mutations. Current therapeutic approaches are evaluated, discussing their limitations, and emphasizing the need to fully elucidate the pathogenesis of X-ALD. Additionally, this review highlights the importance of international collaboration to enhance systematic data collection and advance biomarker discovery, ultimately improving patient outcomes with X-ALD.
Collapse
Affiliation(s)
- Xinxin Zuo
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Zeyu Chen
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
10
|
Chaudhary R, Rehman M, Agarwal V, Kumar A, Kaushik AS, Srivastava S, Srivastava S, Verma R, Rajinikanth PS, Mishra V. Terra incognita of glial cell dynamics in the etiology of leukodystrophies: Broadening disease and therapeutic perspectives. Life Sci 2024; 354:122953. [PMID: 39122110 DOI: 10.1016/j.lfs.2024.122953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Neuroglial cells, also known as glia, are primarily characterized as auxiliary cells within the central nervous system (CNS). The recent findings have shed light on their significance in numerous physiological processes and their involvement in various neurological disorders. Leukodystrophies encompass an array of rare and hereditary neurodegenerative conditions that were initially characterized by the deficiency, aberration, or degradation of myelin sheath within CNS. The primary cellular populations that experience significant alterations are astrocytes, oligodendrocytes and microglia. These glial cells are either structurally or metabolically impaired due to inherent cellular dysfunction. Alternatively, they may fall victim to the accumulation of harmful by-products resulting from metabolic disturbances. In either situation, the possible replacement of glial cells through the utilization of implanted tissue or stem cell-derived human neural or glial progenitor cells hold great promise as a therapeutic strategy for both the restoration of structural integrity through remyelination and the amelioration of metabolic deficiencies. Various emerging treatment strategies like stem cell therapy, ex-vivo gene therapy, infusion of adeno-associated virus vectors, emerging RNA-based therapies as well as long-term therapies have demonstrated success in pre-clinical studies and show promise for rapid clinical translation. Here, we addressed various leukodystrophies in a comprehensive and detailed manner as well as provide prospective therapeutic interventions that are being considered for clinical trials. Further, we aim to emphasize the crucial role of different glial cells in the pathogenesis of leukodystrophies. By doing so, we hope to advance our understanding of the disease, elucidate underlying mechanisms, and facilitate the development of potential treatment interventions.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rajkumar Verma
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
11
|
Manor J, Jangam SV, Chung HL, Bhagwat P, Andrews J, Chester H, Kondo S, Srivastav S, Botas J, Moser AB, Huguenin SM, Wangler MF. Genetic analysis of the X-linked Adrenoleukodystrophy ABCD1 gene in Drosophila uncovers a role in Peroxisomal dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614586. [PMID: 39386423 PMCID: PMC11463603 DOI: 10.1101/2024.09.23.614586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a progressive neurodegenerative disorder caused by a loss-of-function (LOF) mutation in the ATP-binding cassette subfamily D member 1 (ABCD1) gene, leading to the accumulation of very long-chain fatty acids (VLCFAs). This disorder exhibits striking heterogeneity; some male patients develop an early childhood neuroinflammatory demyelination disorder, while other patients, including adult males and most affected female carriers, experience a chronic progressive myelopathy. Adrenocortical failure is observed in almost all male patients, with age of onset varying sometimes being the first diagnostic finding. The gene underlying this spectrum of disease encodes an ATP-binding cassette (ABC) transporter that localizes to peroxisomes and facilitates VLCFA transport. X-ALD is considered a single peroxisomal component defect and does not play a direct role in peroxisome assembly. Drosophila models of other peroxisomal genes have provided mechanistic insight into some of the neurodegenerative mechanisms with reduced lifespan, retinal degeneration, and VLCFA accumulation. Here, we perform a genetic analysis of the fly ABCD1 ortholog Abcd1 (CG2316). Knockdown or deficiency of Abcd1 leads to VLCFA accumulation, salivary gland defects, locomotor impairment and retinal lipid abnormalities. Interestingly, there is also evidence of reduced peroxisomal numbers. Flies overexpressing the human cDNA for ABCD1 display a wing crumpling phenotype characteristic of the pex2 loss-of-function. Surprisingly, overexpression of human ABCD1 appears to inhibit or overwhelm peroxisomal biogenesis to levels similar to null mutations in fly pex2, pex16 and pex3. Drosophila Abcd1 is therefore implicated in peroxisomal number, and overexpression of the human ABCD1 gene acts a potent inhibitor of peroxisomal biogenesis in flies.
Collapse
Affiliation(s)
- Joshua Manor
- Metabolic Disease Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Sharayu V Jangam
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Hyung-lok Chung
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA
- Department of Neurology, Weill Cornell Medical College, New York, NY, USA
| | - Pranjali Bhagwat
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Jonathan Andrews
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Hillary Chester
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Shu Kondo
- Tokyo University of Science, Faculty of Advanced Engineering, Department of Biological Science and Technology, Tokyo, Japan
| | - Saurabh Srivastav
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Juan Botas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Ann B. Moser
- Hugo W Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Suzette M. Huguenin
- Hugo W Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
12
|
Jaspers YRJ, Yska HAF, Bergner CG, Dijkstra IME, Huffnagel IC, Voermans MMC, Wever E, Salomons GS, Vaz FM, Jongejan A, Hermans J, Tryon RK, Lund TC, Köhler W, Engelen M, Kemp S. Lipidomic biomarkers in plasma correlate with disease severity in adrenoleukodystrophy. COMMUNICATIONS MEDICINE 2024; 4:175. [PMID: 39256476 PMCID: PMC11387402 DOI: 10.1038/s43856-024-00605-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND X-linked adrenoleukodystrophy (ALD) is a neurometabolic disorder caused by pathogenic variants in ABCD1 resulting very long-chain fatty acids (VLCFA) accumulation in plasma and tissues. Males can present with various clinical manifestations, including adrenal insufficiency, spinal cord disease, and leukodystrophy. Female patients typically develop spinal cord disease and peripheral neuropathy. Predicting the clinical outcome of an individual patient remains impossible due to the lack of genotype-phenotype correlation and predictive biomarkers. METHODS The availability of a large prospective cohort of well-characterized patients and associated biobank samples allowed us to investigate the relationship between lipidome and disease severity in ALD. We performed a lipidomic analysis of plasma samples from 24 healthy controls, 92 male and 65 female ALD patients. RESULTS Here we show that VLCFA are incorporated into different lipid classes, including lysophosphatidylcholines, phosphatidylcholines, triglycerides, and sphingomyelins. Our results show a strong association between higher levels of VLCFA-containing lipids and the presence of leukodystrophy, adrenal insufficiency, and severe spinal cord disease in male ALD patients. In female ALD patients, VLCFA-lipid levels correlate with X-inactivation patterns in blood mononuclear cells, and higher levels are associated with more severe disease manifestations. Finally, hematopoietic stem cell transplantation significantly reduces, but does not normalize, plasma C26:0-lysophosphatidylcholine levels in male ALD patients. Our findings are supported by the concordance of C26:0-lysophosphatidylcholine and total VLCFA analysis with the lipidomics results. CONCLUSIONS This study reveals the profound impact of ALD on the lipidome and provides potential biomarkers for predicting clinical outcomes in ALD patients.
Collapse
Affiliation(s)
- Yorrick R J Jaspers
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Hemmo A F Yska
- Department of Pediatric Neurology, Amsterdam UMC location University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Caroline G Bergner
- Department of Neurology, Leukodystrophy Outpatient Clinic, Leipzig University Medical Center, Leipzig, Germany
| | - Inge M E Dijkstra
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Irene C Huffnagel
- Department of Pediatric Neurology, Amsterdam UMC location University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Marije M C Voermans
- Department of Pediatric Neurology, Amsterdam UMC location University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Eric Wever
- Bioinformatics Laboratory, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Gajja S Salomons
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatrics, Amsterdam UMC location University of Amsterdam, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Aldo Jongejan
- Bioinformatics Laboratory, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Jill Hermans
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Rebecca K Tryon
- Department of Pediatrics, Division of Bone Marrow Transplantation, University of Minnesota Children's Hospital, Minneapolis, MN, USA
| | - Troy C Lund
- Department of Pediatrics, Division of Bone Marrow Transplantation, University of Minnesota Children's Hospital, Minneapolis, MN, USA
| | - Wolfgang Köhler
- Department of Neurology, Leukodystrophy Outpatient Clinic, Leipzig University Medical Center, Leipzig, Germany
| | - Marc Engelen
- Department of Pediatric Neurology, Amsterdam UMC location University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands.
| |
Collapse
|
13
|
Granadeiro L, Zarralanga VE, Rosa R, Franquinho F, Lamas S, Brites P. Ataxia with giant axonopathy in Acbd5-deficient mice halted by adeno-associated virus gene therapy. Brain 2024; 147:1457-1473. [PMID: 38066620 DOI: 10.1093/brain/awad407] [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: 08/07/2023] [Revised: 10/27/2023] [Accepted: 11/07/2023] [Indexed: 04/06/2024] Open
Abstract
Acyl-CoA binding domain containing 5 (ACBD5) is a critical player in handling very long chain fatty acids (VLCFA) en route for peroxisomal β-oxidation. Mutations in ACBD5 lead to the accumulation of VLCFA and patients present retinal dystrophy, ataxia, psychomotor delay and a severe leukodystrophy. Using CRISPR/Cas9, we generated and characterized an Acbd5 Gly357* mutant allele. Gly357* mutant mice recapitulated key features of the human disorder, including reduced survival, impaired locomotion and reflexes, loss of photoreceptors, and demyelination. The ataxic presentation of Gly357* mice involved the loss of cerebellar Purkinje cells and a giant axonopathy throughout the CNS. Lipidomic studies provided evidence for the extensive lipid dysregulation caused by VLCFA accumulation. Following a proteomic survey, functional studies in neurons treated with VLCFA unravelled a deregulated cytoskeleton with reduced actin dynamics and increased neuronal filopodia. We also show that an adeno-associated virus-mediated gene delivery ameliorated the gait phenotypes and the giant axonopathy, also improving myelination and astrocyte reactivity. Collectively, we established a mouse model with significance for VLCFA-related disorders. The development of relevant neuropathological outcomes enabled the understanding of mechanisms modulated by VLCFA and the evaluation of the efficacy of preclinical therapeutic interventions.
Collapse
Affiliation(s)
- Luis Granadeiro
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S and Instituto de Biologia Molecular e Celular - IBMC, 4200-135 Porto, Portugal
- Graduate Program in Molecular and Cell Biology, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal
| | - Violeta Enríquez Zarralanga
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S and Instituto de Biologia Molecular e Celular - IBMC, 4200-135 Porto, Portugal
| | - Ricardo Rosa
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S and Instituto de Biologia Molecular e Celular - IBMC, 4200-135 Porto, Portugal
| | - Filipa Franquinho
- Animal Facility, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S, 4200-135 Porto, Portugal
| | - Sofia Lamas
- Animal Facility, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S, 4200-135 Porto, Portugal
| | - Pedro Brites
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S and Instituto de Biologia Molecular e Celular - IBMC, 4200-135 Porto, Portugal
| |
Collapse
|
14
|
Yska HAF, Engelen M, Bugiani M. The pathology of X-linked adrenoleukodystrophy: tissue specific changes as a clue to pathophysiology. Orphanet J Rare Dis 2024; 19:138. [PMID: 38549180 PMCID: PMC10976706 DOI: 10.1186/s13023-024-03105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/23/2024] [Indexed: 04/02/2024] Open
Abstract
Although the pathology of X-linked adrenoleukodystrophy (ALD) is well described, it represents the end-stage of neurodegeneration. It is still unclear what cell types are initially involved and what their role is in the disease process. Revisiting the seminal post-mortem studies from the 1970s can generate new hypotheses on pathophysiology. This review describes (histo)pathological changes of the brain and spinal cord in ALD. It aims at integrating older works with current insights and at providing an overarching theory on the pathophysiology of ALD. The data point to an important role for axons and glia in the pathology of both the myelopathy and leukodystrophy of ALD. In-depth pathological analyses with new techniques could help further unravel the sequence of events behind the pathology of ALD.
Collapse
Affiliation(s)
- Hemmo A F Yska
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Marc Engelen
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Pathology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Tang C, Tang F, Cai Y, Tan M, Liu S, Xie T, Jiang X, Huang Y. A pilot study of newborn screening for X-linked adrenoleukodystrophy based on liquid chromatography-tandem mass spectrometry method for detection of C26:0-lysophosphatidylcholine in dried blood spots: Results from 43,653 newborns in a southern Chinese population. Clin Chim Acta 2024; 552:117653. [PMID: 37977233 DOI: 10.1016/j.cca.2023.117653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND X-linked adrenoleukodystrophy (X-ALD) is a rare X-linked disease caused by mutations of the ABCD1 gene. C26:0-lysophosphatidylcholine (C26:0-LPC) has been proved to be an accurate biomarker for X-ALD. This study aims to propose an effective method for screening of X-ALD and to evaluate the performance of the newborn screening (NBS) assay for X-ALD in Guangzhou. METHODS C26:0-LPC in dried blood spots (DBS) was extracted by methanol solution containing isotope-labelled internal standard (C26:0-d4-LPC) and analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). The sensitivity of the method was assessed in eight male X-ALD patients, two female carriers and 583 healthy controls. The method was conducted on 43,653 newborns. Next generation sequencing was performed on screen-positive samples. Plasma analysis of very long-chain fatty acids and genetic counselling were performed by way of follow-up. RESULTS Elevated C26:0-LPC were 100% sensitive for screening of X-ALD. Of 43,653 newborns, 32 (18 males, 14 females) screened positive. Of these, 14 (43.7%) were identified ABCD1 variants, including seven hemizygous males and seven heterozygous females, and two (6.3%) were diagnosed with other peroxisomal disorders. CONCLUSION The LC-MS/MS method for screening of X-ALD can identify males, heterozygous females and other peroxisomal disorders. The incidence of X-ALD in Guangzhou is not low.
Collapse
Affiliation(s)
- Chengfang Tang
- Guangzhou Newborn Screening Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Fang Tang
- Guangzhou Newborn Screening Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Yanna Cai
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Minyi Tan
- Guangzhou Newborn Screening Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Sichi Liu
- Guangzhou Newborn Screening Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Ting Xie
- Guangzhou Newborn Screening Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Xiang Jiang
- Guangzhou Newborn Screening Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Yonglan Huang
- Guangzhou Newborn Screening Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China.
| |
Collapse
|
16
|
Videbæk C, Melgaard L, Lund AM, Grønborg SW. Newborn screening for adrenoleukodystrophy: International experiences and challenges. Mol Genet Metab 2023; 140:107734. [PMID: 37979237 DOI: 10.1016/j.ymgme.2023.107734] [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: 08/28/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
X-linked adrenoleukodystrophy (XALD) is the most common leukodystrophy. It has an estimated incidence of around 1/17.000, and a variable phenotype. Following the passage of Aidens Law, New York became the first state to implement a newborn screening for XALD in 2013. Since then, 38 American states, Taiwan, and the Netherlands have included XALD in their NBS program, and Japan and Italy have ongoing pilot studies. Screening for XALD allows for early, potentially lifesaving treatment of adrenal insufficiency and cerebral demyelination but is also a complex subject, due to our limited understanding of the natural history and lack of prognostic biomarkers. Screening protocols and algorithms vary between countries and states, and results and experiences gained so far are important for the future implementation of XALD NBS in other countries. In this review, we have examined the algorithms, methodologies, and outcomes used, as well as how common challenges are addressed in countries/states that have experience using NBS for XALD. We identified 14 peer-reviewed reports on NBS for XALD. All studies presented methods for detecting XALD at birth by NBS using a combination of mass spectrometry and ABCD1 gene sequencing. This has allowed for early surveillance of presymptomatic XALD patients, and the possibility for early detection and timely treatment of XALD manifestations. Obstacles to NBS for XALD include how to deal with variants of unknown significance, whether to screen females, and the ethical concerns of an NBS for a disease where we have limited understanding of natural history and phenotype/genotype correlation.
Collapse
Affiliation(s)
- Cecilie Videbæk
- Centre for Inherited Metabolic Diseases, Departments of Clinical Genetics and Paediatrics, Copenhagen University Hospital, Rigshospitalet, Denmark.
| | - Lars Melgaard
- Danish Center for Neonatal Screening, Clinical Mass Spectrometry, Statens Serum Institut, Denmark
| | - Allan M Lund
- Centre for Inherited Metabolic Diseases, Departments of Clinical Genetics and Paediatrics, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Sabine Weller Grønborg
- Centre for Inherited Metabolic Diseases, Departments of Clinical Genetics and Paediatrics, Copenhagen University Hospital, Rigshospitalet, Denmark
| |
Collapse
|
17
|
Waterham HR, Koster J, Ebberink MS, Ješina P, Zeman J, Nosková L, Kmoch S, Devic P, Cheillan D, Wanders RJA, Ferdinandusse S. Autosomal dominant Zellweger spectrum disorder caused by de novo variants in PEX14 gene. Genet Med 2023; 25:100944. [PMID: 37493040 DOI: 10.1016/j.gim.2023.100944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
PURPOSE Zellweger spectrum disorders (ZSDs) are known as autosomal recessive disorders caused by defective peroxisome biogenesis due to bi-allelic pathogenic variants in any of at least 13 different PEX genes. Here, we report 2 unrelated patients who present with an autosomal dominant ZSD. METHODS We performed biochemical and genetic studies in blood and skin fibroblasts of the patients and demonstrated the pathogenicity of the identified PEX14 variants by functional cell studies. RESULTS We identified 2 different single heterozygous de novo variants in the PEX14 genes of 2 patients diagnosed with ZSD. Both variants cause messenger RNA mis-splicing, leading to stable expression of similar C-terminally truncated PEX14 proteins. Functional studies indicated that the truncated PEX14 proteins lost their function in peroxisomal matrix protein import and cause increased degradation of peroxisomes, ie, pexophagy, thus exerting a dominant-negative effect on peroxisome functioning. Inhibition of pexophagy by different autophagy inhibitors or genetic knockdown of the peroxisomal autophagy receptor NBR1 resulted in restoration of peroxisomal functions in the patients' fibroblasts. CONCLUSION Our finding of an autosomal dominant ZSD expands the genetic repertoire of ZSDs. Our study underscores that single heterozygous variants should not be ignored as possible genetic cause of diseases with an established autosomal recessive mode of inheritance.
Collapse
Affiliation(s)
- Hans R Waterham
- Amsterdam UMC - AMC, Department of Laboratory Medicine, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands; Amsterdam Reproduction & Development, Amsterdam, The Netherlands; United for Metabolic Diseases, The Netherlands.
| | - Janet Koster
- Amsterdam UMC - AMC, Department of Laboratory Medicine, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands
| | - Merel S Ebberink
- Amsterdam UMC - AMC, Department of Laboratory Medicine, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands
| | - Pavel Ješina
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague 2, Czech Republic
| | - Jiri Zeman
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague 2, Czech Republic
| | - Lenka Nosková
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague 2, Czech Republic
| | - Stanislav Kmoch
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General Faculty Hospital, Prague 2, Czech Republic
| | - Perrine Devic
- Centre Hospitalier Universitaire de Lyon, CHU Lyon·U 301, Hopital Neurologique, Bron, France
| | - David Cheillan
- Service Biochimie et Biologie Moléculaire Grand Est, UM Pathologies Métaboliques, Erythrocytaires et Dépistage Périnatal, Centre de Biologie et de Pathologie Est, Groupement Hospitalier Est - Hospices Civils de Lyon, Bron Cedex, France
| | - Ronald J A Wanders
- Amsterdam UMC - AMC, Department of Laboratory Medicine, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands; Amsterdam Reproduction & Development, Amsterdam, The Netherlands; United for Metabolic Diseases, The Netherlands
| | - Sacha Ferdinandusse
- Amsterdam UMC - AMC, Department of Laboratory Medicine, Laboratory Genetic Metabolic Diseases, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Kemp S, Orsini JJ, Ebberink MS, Engelen M, Lund TC. VUS: Variant of uncertain significance or very unclear situation? Mol Genet Metab 2023; 140:107678. [PMID: 37574344 DOI: 10.1016/j.ymgme.2023.107678] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023]
Abstract
The advancements in population screening, including newborn screening, enables the identification of disease-causing variants and timely initiation of treatment. However, screening may also identify mild variants, non-disease variants, and variants of uncertain significance (VUS). The identification of a VUS poses a challenge in terms of diagnostic uncertainty and confusion. X-linked adrenoleukodystrophy (ALD) serves as an illustrative example of this complex issue. ALD is a monogenic neurometabolic disease with a complex clinical presentation and a lack of predictive tests for clinical severity. Despite the success of ALD newborn screening, a significant proportion (62%) of missense variants identified through newborn screening exhibit uncertainty regarding their pathogenicity. Resolving this issue requires ongoing efforts to accurately classify variants and refine screening protocols. While it is undisputable that ALD newborn screening greatly benefits boys with the disease, the identification of VUS underscores the need for continuous research and collaboration in improving screening practices.
Collapse
Affiliation(s)
- Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC location University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands.
| | - Joseph J Orsini
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Merel S Ebberink
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC location University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Marc Engelen
- Department of Pediatric Neurology, Amsterdam UMC location University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Troy C Lund
- Department of Pediatrics, Blood and Marrow Transplant Program, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
19
|
Mohan S, Mayers M, Weaver M, Baudet H, De Biase I, Goldstein J, Mao R, McGlaughon J, Moser A, Pujol A, Suchy S, Yuzyuk T, Braverman NE. Evaluating the strength of evidence for genes implicated in peroxisomal disorders using the ClinGen clinical validity framework and providing updates to the peroxisomal disease nomenclature. Mol Genet Metab 2023; 139:107604. [PMID: 37236006 PMCID: PMC10484331 DOI: 10.1016/j.ymgme.2023.107604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/09/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023]
Abstract
Peroxisomal disorders are heterogeneous in nature, with phenotypic overlap that is indistinguishable without molecular testing. Newborn screening and gene sequencing for a panel of genes implicated in peroxisomal diseases are critical tools for the early and accurate detection of these disorders. It is therefore essential to evaluate the clinical validity of the genes included in sequencing panels for peroxisomal disorders. The Peroxisomal Gene Curation Expert Panel (GCEP) assessed genes frequently included on clinical peroxisomal testing panels using the Clinical Genome Resource (ClinGen) gene-disease validity curation framework and classified gene-disease relationships as Definitive, Strong, Moderate, Limited, Disputed, Refuted, or No Known Disease Relationship. Subsequent to gene curation, the GCEP made recommendations to update the disease nomenclature and ontology in the Monarch Disease Ontology (Mondo) database. Thirty-six genes were assessed for the strength of evidence supporting their role in peroxisomal disease, leading to 36 gene-disease relationships, after two genes were removed for their lack of a role in peroxisomal disease and two genes were curated for two different disease entities each. Of these, 23 were classified as Definitive (64%), one as Strong (3%), eight as Moderate (23%), two as Limited (5%), and two as No known disease relationship (5%). No contradictory evidence was found to classify any relationships as Disputed or Refuted. The gene-disease relationship curations are publicly available on the ClinGen website (https://clinicalgenome.org/affiliation/40049/). The changes to peroxisomal disease nomenclature are displayed on the Mondo website (http://purl.obolibrary.org/obo/MONDO_0019053). The Peroxisomal GCEP-curated gene-disease relationships will inform clinical and laboratory diagnostics and enhance molecular testing and reporting. As new data will emerge, the gene-disease classifications asserted by the Peroxisomal GCEP will be re-evaluated periodically.
Collapse
Affiliation(s)
- Shruthi Mohan
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Megan Mayers
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Meredith Weaver
- American College of Medical Genetics and Genomics, Bethesda, MD, USA
| | - Heather Baudet
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | | | - Jennifer Goldstein
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Rong Mao
- ARUP Laboratories, Salt Lake City, UT, USA
| | | | - Ann Moser
- Kennedy Krieger Institute, Baltimore, MD, USA
| | - Aurora Pujol
- Bellvitge Biomedical Research Institute (IDIBELL Instituto de Investigación Biomédica de Bellvitge), Barcelona, Spain
| | | | | | - Nancy E Braverman
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
20
|
Luo XM, Liu LY, Wang QH, Wang YY, Wang J, Yang XY, Li SJ, Zou LP. Exploratory study of autophagy inducer sirolimus for childhood cerebral adrenoleukodystrophy. Front Pediatr 2023; 11:1187078. [PMID: 37360358 PMCID: PMC10289280 DOI: 10.3389/fped.2023.1187078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/05/2023] [Indexed: 06/28/2023] Open
Abstract
Objectives X-linked adrenoleukodystrophy (ALD) is a peroxisomal disease caused by mutations in the ABCD1 gene. Childhood cerebral ALD (CCALD) is characterized by inflammatory demyelination, rapidly progressing, often fatal. Hematopoietic stem cell transplant only delays disease progression in patients with early-stage cerebral ALD. Based on emergency humanitarianism, this study aims to investigate the safety and efficacy of sirolimus in the treatment of patients with CCALD. Methods This was a prospective, single-center, one-arm clinical trial. We enrolled patients with CCALD, and all enrolled patients received sirolimus treatment for three months. Adverse events were monitored and recorded to evaluate the safety. The efficacy was evaluated using the neurologic function scale (NFS), Loes score, and white matter hyperintensities. Results A total of 12 patients were included and all presented with CCALD. Four patients dropped out and a total of eight patients in the advanced stage completed a 3-month follow-up. There were no serious adverse events, and the common adverse events were hypertonia and oral ulcers. After sirolimus treatment, three of the four patients with an initial NFS > 10 showed improvements in their clinical symptoms. Loes scores decreased by 0.5-1 point in two of eight patients and remained unchanged in one patient. Analysis of white matter hyperintensities revealed a significant decrease in signal intensity (n = 7, p = 0.0156). Conclusions Our study suggested that autophagy inducer sirolimus is safe for CCALD. Sirolimus did not improve clinical symptoms of patients with advanced CCALD significantly. Further study with larger sample size and longer follow-up is needed to confirm the drug efficacy.Clinical Trial registration: https://www.chictr.org.cn/historyversionpuben.aspx, identifier ChiCTR1900021288.
Collapse
Affiliation(s)
- Xiao-Mei Luo
- Senior Department of Pediatrics, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Li-Ying Liu
- Department of Pediatrics, Xuanwu Hospital Capital Medical University, Beijing, China
- Department of Neurology, Beijing Jingdu Children's Hospital, Beijing, China
| | - Qiu-Hong Wang
- Senior Department of Pediatrics, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Yang-Yang Wang
- Senior Department of Pediatrics, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Jing Wang
- Senior Department of Pediatrics, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Xiao-Yan Yang
- Senior Department of Pediatrics, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Shi-Jun Li
- Department of Radiology, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Li-Ping Zou
- Senior Department of Pediatrics, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
21
|
Dohr KA, Tokic S, Gastager-Ehgartner M, Stojakovic T, Dumic M, Plecko B, Dumic KK. Two Single Nucleotide Deletions in the ABCD1 Gene Causing Distinct Phenotypes of X-Linked Adrenoleukodystrophy. Int J Mol Sci 2023; 24:ijms24065957. [PMID: 36983033 PMCID: PMC10051867 DOI: 10.3390/ijms24065957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a rare inborn error of the peroxisomal metabolism caused by pathologic variants in the ATP-binding cassette transporter type D, member 1 (ABCD1) gene located on the X-chromosome. ABCD1 protein, also known as adrenoleukodystrophy protein, is responsible for transport of the very long chain fatty acids (VLCFA) from cytoplasm into the peroxisomes. Therefore, altered function or lack of the ABCD1 protein leads to accumulation of VLCFA in various tissues and blood plasma leading to either rapidly progressive leukodystrophy (cerebral ALD), progressive adrenomyeloneuropathy (AMN), or isolated primary adrenal insufficiency (Addison's disease). We report two distinct single nucleotide deletions in the ABCD1 gene, c.253delC [p.Arg85Glyfs*18] in exon 1, leading to both cerebral ALD and to AMN phenotype in one family, and c.1275delA [p.Phe426Leufs*15] in exon 4, leading to AMN and primary adrenal insufficiency in a second family. For the latter variant, we demonstrate reduced mRNA expression and a complete absence of the ABCD1 protein in PBMC. Distinct mRNA and protein expression in the index patient and heterozygous carriers does not associate with VLCFA concentration in plasma, which is in line with the absence of genotype-phenotype correlation in X-ALD.
Collapse
Affiliation(s)
- Katrin A Dohr
- Research Unit of Analytical Mass Spectrometry, Cell Biology and Biochemistry of Inborn Errors of Metabolism, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Silvija Tokic
- Research Unit of Analytical Mass Spectrometry, Cell Biology and Biochemistry of Inborn Errors of Metabolism, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Magdalena Gastager-Ehgartner
- Research Unit of Analytical Mass Spectrometry, Cell Biology and Biochemistry of Inborn Errors of Metabolism, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, 8036 Graz, Austria
| | - Miroslav Dumic
- Department of Paediatric Endocrinology and Diabetes, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Barbara Plecko
- Division of General Paediatrics, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Katja K Dumic
- Department of Paediatric Endocrinology and Diabetes, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
22
|
Chang C, Gupta AO, Orchard PJ, Nascene DR, Kierstein J, Tryon RK, Lund TC. A case series of adrenoleukodystrophy in children conceived through in vitro fertilization with an egg donor. F S Rep 2023; 4:24-28. [PMID: 36959964 PMCID: PMC10028476 DOI: 10.1016/j.xfre.2022.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Objective To report 3 cases of adrenoleukodystrophy (ALD) in children conceived by in vitro fertilization (IVF) and egg donation. Design A case report. Patients Patients aged 4-5 years old, evaluated by the University of Minnesota Leukodystrophy Center, who were diagnosed with ALD after being conceived by IVF with oocytes provided by the same donor. Interventions One patient received a hematopoietic stem cell transplant from a human leukocyte antigen-matched donor, and 1 patient received autologous lentiviral corrected hematopoietic cells. The disease state in 1 patient was unfortunately too advanced for effective treatment to be administered. Main Outcome Measures Progression of disease after diagnosis or treatment was observed by cerebral magnetic resonance imaging and monitoring the development or advancement of any cognitive, adaptive, and motor deficits. Results Patients who received a transplant for ALD successfully experienced little to no disease progression at least 6 months to 1 year after treatment. Conclusions These 3 cases of transmission of ALD through oocyte donation and IVF highlight the potential need to implement more comprehensive genetic screening of gamete donors to prevent the transfer of rare but severe genetic diseases through IVF. Further, these cases highlight limitations in carrier screening guidelines that limit reportable variants to pathogenic and likely pathogenic variants.
Collapse
Affiliation(s)
- Crystal Chang
- Division of Pediatric Blood and Marrow Transplantation and Cellular Therapy Program, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Ashish O. Gupta
- Division of Pediatric Blood and Marrow Transplantation and Cellular Therapy Program, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Paul J. Orchard
- Division of Pediatric Blood and Marrow Transplantation and Cellular Therapy Program, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | - David R. Nascene
- Department of Radiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Janell Kierstein
- Department of Genetics and Metabolism, Children’s Hospital Colorado, Aurora, Colorado
| | - Rebecca K. Tryon
- Division of Pediatric Blood and Marrow Transplantation and Cellular Therapy Program, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
- Department of Genetics, M Health Fairview, Minneapolis, Minnesota
| | - Troy C. Lund
- Division of Pediatric Blood and Marrow Transplantation and Cellular Therapy Program, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
- Reprint requests: Troy C. Lund, M.D., Ph.D., Division of Pediatric Blood and Marrow Transplant, Metabolic Program, University of Minnesota, 420 Delaware St SE, Minneapolis, Minnesota 55455.
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW The present review summarizes recent advances in the diagnosis and management of patients with X-linked adrenoleukodystrophy (ALD). RECENT FINDINGS Although ALD screening has been on the list of Recommended Uniform Screening Panel since 2016, only 30 states in the United States are currently testing their newborns for this disease. Hematopoietic stem cell transplant (HSCT) remains the only successful treatment option available for early cerebral ALD but does not reverse neurological changes or affect the course of adrenal insufficiency. There remains a significant knowledge gap in our understanding and treatment of this disease. Novel therapies such as gene therapy and gene editing have shown promising results in animal models and are exciting potential treatment options for the future.Recently, the American Academy of Neurologists released their consensus guidelines on the diagnosis, surveillance, and management of ALD. SUMMARY Early diagnosis and HSCT are key to improving the morbidity and mortality associated with ALD. The implementation of universal newborn screening for ALD and rigorous investigations of novel diagnostic and therapeutic agents is the need of the hour.
Collapse
|
24
|
Zheng F, Lin Z, Hu Y, Shi X, Zhao Q, Lin Z. Identification of a Novel Non-Canonical Splice-Site Variant in ABCD1. J Clin Med 2023; 12:jcm12020473. [PMID: 36675402 PMCID: PMC9863105 DOI: 10.3390/jcm12020473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/09/2023] Open
Abstract
Cerebral adrenoleukodystrophy (CALD) is a fatal genetic disease characterized by rapid, devastating neurological decline, with a narrow curative treatment window in the early stage. Non-canonical splice-site (NCSS) variants can easily be missed during genomic DNA analyses, and only a few of them in ABCD1 have been explored. Here, we studied a Chinese patient with clinical features similar to those of early-stage CALD but with a negative molecular diagnosis and a sibling who had presumably died of CALD. Trio-based whole-exome sequencing (trio-WES) and RNA sequencing (RNA-Seq) revealed a novel hemizygote NCSS variant c.901-25_901-9 del in ABCD1 intron 1, resulting in a complex splicing pattern. The in vitro minigene assay revealed that the c.901-25_901-9 del construct contained two aberrant transcripts that caused skipping of exon 2 and a small 48-bp deletion on left of the same exon. We identified a novel NCSS variant, that extends the spectrum of the known ABCD1 variants, and demonstrated the pathogenicity of this gene variant. Our findings highlight the importance of combining RNA-Seq and WES techniques for prompt diagnosis of leukodystrophy with NCSS variants.
Collapse
Affiliation(s)
- Feixia Zheng
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhongdong Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Ying Hu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xulai Shi
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Qianlei Zhao
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhenlang Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Wenzhou Key Laboratory of Perinatal Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Correspondence: ; Tel.: +86-13-80-668-9800
| |
Collapse
|
25
|
Albersen M, van der Beek SL, Dijkstra IME, Alders M, Barendsen RW, Bliek J, Boelen A, Ebberink MS, Ferdinandusse S, Goorden SMI, Heijboer AC, Jansen M, Jaspers YRJ, Metgod I, Salomons GS, Vaz FM, Verschoof-Puite RK, Visser WF, Dekkers E, Engelen M, Kemp S. Sex-specific newborn screening for X-linked adrenoleukodystrophy. J Inherit Metab Dis 2023; 46:116-128. [PMID: 36256460 PMCID: PMC10092852 DOI: 10.1002/jimd.12571] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 02/07/2023]
Abstract
Males with X-linked adrenoleukodystrophy (ALD) are at high risk for developing adrenal insufficiency and/or progressive leukodystrophy (cerebral ALD) at an early age. Pathogenic variants in ABCD1 result in elevated levels of very long-chain fatty acids (VLCFA), including C26:0-lysophosphatidylcholine (C26:0-LPC). Newborn screening for ALD enables prospective monitoring and timely therapeutic intervention, thereby preventing irreversible damage and saving lives. The Dutch Health Council recommended to screen only male newborns for ALD without identifying untreatable conditions associated with elevated C26:0-LPC, like Zellweger spectrum disorders and single peroxisomal enzyme defects. Here, we present the results of the SCAN (Screening for ALD in the Netherlands) study which is the first sex-specific newborn screening program worldwide. Males with ALD are identified based on elevated C26:0-LPC levels, the presence of one X-chromosome and a variant in ABCD1, in heel prick dried bloodspots. Screening of 71 208 newborns resulted in the identification of four boys with ALD who, following referral to the pediatric neurologist and confirmation of the diagnosis, enrolled in a long-term follow-up program. The results of this pilot show the feasibility of employing a boys-only screening algorithm that identifies males with ALD without identifying untreatable conditions. This approach will be of interest to countries that are considering ALD newborn screening but are reluctant to identify girls with ALD because for girls there is no direct health benefit. We also analyzed whether gestational age, sex, birth weight and age at heel prick blood sampling affect C26:0-LPC concentrations and demonstrate that these covariates have a minimal effect.
Collapse
Affiliation(s)
- Monique Albersen
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam UMC location University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Samantha L van der Beek
- Reference Laboratory for Neonatal Screening, Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Inge M E Dijkstra
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Mariëlle Alders
- Department of Human Genetics, Amsterdam UMC location University of Amsterdam, Amsterdam Reproduction & Development, Amsterdam, The Netherlands
| | - Rinse W Barendsen
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Jet Bliek
- Department of Human Genetics, Amsterdam UMC location University of Amsterdam, Amsterdam Reproduction & Development, Amsterdam, The Netherlands
| | - Anita Boelen
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam UMC location University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Merel S Ebberink
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Susan M I Goorden
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Annemieke C Heijboer
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam UMC location University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Mandy Jansen
- Department for Vaccine Supply and Prevention Programs, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Yorrick R J Jaspers
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Ingrid Metgod
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam UMC location University of Amsterdam, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Gajja S Salomons
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Department of Pediatric Neurology, Amsterdam UMC location University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Rendelien K Verschoof-Puite
- Department for Vaccine Supply and Prevention Programs, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Wouter F Visser
- Reference Laboratory for Neonatal Screening, Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Eugènie Dekkers
- Center for Population Screening, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Marc Engelen
- Department of Pediatric Neurology, Amsterdam UMC location University of Amsterdam, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Wang P, Du X, Shen Q, Jiang W, Shen C, Wang H, Zhou S, Wang Y, Qian X, Zhai X. Unrelated umbilical cord blood transplantation for children with hereditary leukodystrophy: A retrospective study. Front Neurol 2022; 13:999919. [PMID: 36247778 PMCID: PMC9561100 DOI: 10.3389/fneur.2022.999919] [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: 07/21/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022] Open
Abstract
Objective To analyze the efficiency of unrelated umbilical cord blood transplantation (UCBT) in the treatment of hereditary leukodystrophy following busulfan- and cyclophosphamide-based myeloablative chemotherapy. Methods A retrospective study was performed in patients with hereditary leukodystrophy who underwent UCBT after myeloablative chemotherapy between April 2015 and March 2020. Results The study cohort included 12 pediatric patients (ten males), nine with cerebral adrenoleukodystrophy (ALD) and three with juvenile globoid cell leukodystrophy (GLD). All received HLA-matched or partially mismatched unrelated UCBT. There were no cases of graft rejection. Median neutrophil engraftment time was 20 days [12–33 days] and median platelet engraftment time was 29 days [14–65 days]. Median follow-up was 36 months [1–86 months], and the overall survival rate for patients with cerebral ALD and juvenile GLD after UCBT was 77.8% (7/9) and 100% (3/3), respectively. In patients with ALD, although lipid profiles (serum very-long-chain fatty acid) were improved post-UCBT, six patients demonstrated worse neurologic function score and performance status post-UCBT, and six patients had higher Loes scores at last follow-up compared with baseline. In patients with juvenile GLD, all patients showed stable neurologic function score and performance status despite the Loes score of one patient increased slightly after transplantation. Conclusion In patients with cerebral ALD, patients with no or mild neurological symptoms can benefit from UCBT, while UCBT cannot reverse advanced disease. In patients with juvenile GLD, UCBT is safe and contributes to stabilize neurological function.
Collapse
Affiliation(s)
- Ping Wang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Xiaonan Du
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Quanli Shen
- Department of Radiology, Children's Hospital of Fudan University, Shanghai, China
| | - Wenjin Jiang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Chen Shen
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Hongsheng Wang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Shuizhen Zhou
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Yi Wang
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Xiaowen Qian
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
- Xiaowen Qian
| | - Xiaowen Zhai
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai, China
- *Correspondence: Xiaowen Zhai
| |
Collapse
|
27
|
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
|
28
|
Chen HA, Hsu RH, Chen PW, Lee NC, Chiu PC, Hwu WL, Chien YH. High incidence of null variants identified from newborn screening of X-linked adrenoleukodystrophy in Taiwan. Mol Genet Metab Rep 2022; 32:100902. [PMID: 36046390 PMCID: PMC9421440 DOI: 10.1016/j.ymgmr.2022.100902] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/15/2022] Open
Abstract
Background Adrenoleukodystrophy (ALD) is an X-linked peroxisomal disorder caused by variants in the ABCD1 gene and can lead to Addison disease, childhood cerebral ALD, or adrenomyeloneuropathy. Presymptomatic hematopoietic stem cell transplantation is the only curative treatment for the disease and requires early detection through newborn screening (NBS) and close follow-up. Methods An NBS program for ALD was performed by a two-tiered dried blood spot (DBS) lysophosphatidylcholine C26:0 (C26:0-LPC) concentration analysis. ABCD1 sequencing was eventually added as a third-tier test, and whole exome sequencing was used to confirm the diagnosis of all peroxisomal diseases. Affected newborns were followed-up for adrenal insufficiency and cerebral white matter abnormalities. Results We identified 12 males and 10 females with ABCD1 variants, and 3 patients with Zellweger syndrome from 320,528 newborns. Eight (36.4%) ABCD1 variants identified in the current study were null variants, but there were no hotspots or founder effect. During a median follow-up period of 2.28 years, two (16.7%) male patients with ABCD1 variants developed Addison's disease. Extended family screening revealed one 28-year-old asymptomatic hemizygous father of a null variant (c.678delC). Among the three with Zellweger syndrome, one died at the age of 3 months, one showed developmental delay at the age of 1 year, and one was lost to follow-up. Conclusion Screening for ALD has been added to the NBS program in Taiwan with a high degree of success. The screening algorithm revealed a high proportion of null variants in cases found by NBS in Taiwan, a subset of patients who may have earlier disease onset. We also demonstrate the feasibility of combining the diagnosis of ALD and other peroxisomal disorders into one screening algorithm. We report our screening results of a successful newborn screening for adrenoleukodystrophy in Taiwan since November 2016. C26:0-LPC levels at newborn screening tended to be higher in males with null variants than those with missense variants. A higher proportion of ALD patients carry null variants in Taiwan,and may have earlier onset or more severe phenotypes.
Collapse
Affiliation(s)
- Hui-An Chen
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Rai-Hseng Hsu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Pin-Wen Chen
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ni-Chung Lee
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Pao-Chin Chiu
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wuh-Liang Hwu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Yin-Hsiu Chien
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Corresponding author at: Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
29
|
Volmrich AM, Cuénant LM, Forghani I, Hsieh SL, Shapiro LT. ABCD1 Gene Mutations: Mechanisms and Management of Adrenomyeloneuropathy. Appl Clin Genet 2022; 15:111-123. [PMID: 35983253 PMCID: PMC9381027 DOI: 10.2147/tacg.s359479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/06/2022] [Indexed: 01/05/2023] Open
Abstract
Pathogenic variants in the ABCD1 gene on the X chromosome may result in widely heterogenous phenotypes, including adrenomyeloneuropathy (AMN). Affected males typically present in their third or fourth decade of life with progressive lower limb weakness and spasticity, and may develop signs and symptoms of adrenal insufficiency and/or cerebral demyelination. Heterozygous females may be asymptomatic, but may develop a later-onset and more slowly progressive spastic paraparesis. In this review, we describe the clinical presentation of AMN, as well as its diagnosis and management. The role of rehabilitative therapies and options for management of spasticity are highlighted.
Collapse
Affiliation(s)
- Alyssa M Volmrich
- Department of Physical Medicine & Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lauren M Cuénant
- Department of Physical Medicine & Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Irman Forghani
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sharon L Hsieh
- MD/MPH Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lauren T Shapiro
- Department of Physical Medicine & Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
- Correspondence: Lauren T Shapiro, Department of Physical Medicine & Rehabilitation; University of Miami Miller School of Medicine, P.O. Box 016960 (C-206), Miami, FL, 33101, USA, Tel +1 305 243-6605, Fax +1 305 243-4650, Email
| |
Collapse
|
30
|
Chen ZP, Xu D, Wang L, Mao YX, Li Y, Cheng MT, Zhou CZ, Hou WT, Chen Y. Structural basis of substrate recognition and translocation by human very long-chain fatty acid transporter ABCD1. Nat Commun 2022; 13:3299. [PMID: 35676282 PMCID: PMC9177597 DOI: 10.1038/s41467-022-30974-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 05/26/2022] [Indexed: 11/08/2022] Open
Abstract
Human ABC transporter ABCD1 transports very long-chain fatty acids from cytosol to peroxisome for β-oxidation, dysfunction of which usually causes the X-linked adrenoleukodystrophy (X-ALD). Here, we report three cryogenic electron microscopy structures of ABCD1: the apo-form, substrate- and ATP-bound forms. Distinct from what was seen in the previously reported ABC transporters, the two symmetric molecules of behenoyl coenzyme A (C22:0-CoA) cooperatively bind to the transmembrane domains (TMDs). For each C22:0-CoA, the hydrophilic 3'-phospho-ADP moiety of CoA portion inserts into one TMD, with the succeeding pantothenate and cysteamine moiety crossing the inter-domain cavity, whereas the hydrophobic fatty acyl chain extends to the opposite TMD. Structural analysis combined with biochemical assays illustrates snapshots of ABCD1-mediated substrate transport cycle. It advances our understanding on the selective oxidation of fatty acids and molecular pathology of X-ALD.
Collapse
Affiliation(s)
- Zhi-Peng Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Da Xu
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Liang Wang
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Yao-Xu Mao
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Yang Li
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Meng-Ting Cheng
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Cong-Zhao Zhou
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Wen-Tao Hou
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Yuxing Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
31
|
Baker CV, Cady Keller A, Lutz R, Eveans K, Baumert K, DiPerna JC, Rizzo WB. Newborn Screening for X-Linked Adrenoleukodystrophy in Nebraska: Initial Experiences and Challenges. Int J Neonatal Screen 2022; 8:ijns8020029. [PMID: 35645283 PMCID: PMC9149921 DOI: 10.3390/ijns8020029] [Citation(s) in RCA: 6] [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: 03/08/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a neurodegenerative disease caused by pathogenic variants in ABCD1 resulting in defective peroxisomal oxidation of very long-chain fatty acids. Most male patients develop adrenal insufficiency and one of two neurologic phenotypes: a rapidly progressive demyelinating disease in mid-childhood (childhood cerebral X-ALD, ccALD) or an adult-onset spastic paraparesis (adrenomyeloneuropathy, AMN). The neurodegenerative course of ccALD can be halted if patients are treated with hematopoietic stem cell transplantation at the earliest onset of white matter disease. Newborn screening for X-ALD can be accomplished by measuring C26:0-lysophosphatidylcholine in dried blood spots. In Nebraska, X-ALD newborn screening was instituted in July 2018. Over a period of 3.3 years, 82,920 newborns were screened with 13 positive infants detected (4 males, 9 females), giving a birth prevalence of 1:10,583 in males and 1:4510 in females. All positive newborns had DNA variants in ABCD1. Lack of genotype-phenotype correlations, absence of predictive biomarkers for ccALD or AMN, and a high proportion of ABCD1 variants of uncertain significance are unique challenges in counseling families. Surveillance testing for adrenal and neurologic disease in presymptomatic X-ALD males will improve survival and overall quality of life.
Collapse
Affiliation(s)
- Craig V. Baker
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.V.B.); (A.C.K.); (R.L.)
| | - Alyssa Cady Keller
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.V.B.); (A.C.K.); (R.L.)
| | - Richard Lutz
- Munroe-Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.V.B.); (A.C.K.); (R.L.)
| | - Karen Eveans
- Nebraska Newborn Screening Program, Department of Health and Human Services, Lincoln, NE 68509, USA; (K.E.); (K.B.)
| | - Krystal Baumert
- Nebraska Newborn Screening Program, Department of Health and Human Services, Lincoln, NE 68509, USA; (K.E.); (K.B.)
| | | | - William B. Rizzo
- Department of Pediatrics and Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence: ; Tel.: +1-402-559-2560
| |
Collapse
|