1
|
Mangione CC, Frank A, Dalgard CL, Burnett BG, Flagg TP. Transcriptional reprogramming in SMA mouse hearts reveals signatures of early heart failure and dysregulated calcium signaling. Hum Mol Genet 2025:ddaf060. [PMID: 40287831 DOI: 10.1093/hmg/ddaf060] [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: 11/15/2024] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Spinal muscular atrophy (SMA) is an inherited neurodegenerative disease that leads to loss of motor neurons in the anterior horn of the spinal cord with consequent muscle atrophy. SMA results from the functional deletions of the SMN1 gene, resulting in insufficient production of the survival motor neuron (SMN) protein. It is not known why lower motor neurons are particularly sensitive to the loss of SMN function, but it is increasingly apparent that extraneuronal tissues, such as cardiac and skeletal muscle, are also affected by SMN deficiency. We have previously shown that SMN deficiency in a mouse model of spinal muscular atrophy (SMNΔ7) impairs cardiomyocyte contraction and Ca2+ handling. In this study, we performed a comparative total mRNA sequencing analysis of whole hearts isolated at an early (P5) or late (P10) stage of the disease process to investigate the mechanisms contributing to cardiac pathology in SMA. The results demonstrate transcriptional signatures consistent with heart failure, dysregulation of Ca2+ signaling, and hypoxia induced changes occurring as early as P5 and persisting through P10. Similar transcriptomic changes in skeletal muscle tissue indicate that there are likely common, cell autonomous molecular mechanisms resulting in both cardiac and skeletal muscle due to SMN deficiency. The identification of these common themes suggests a link underlying the mechanism of neuronal and non-neuronal deficits in SMA.
Collapse
Affiliation(s)
- Cecelia C Mangione
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD, 20814, United States
| | - Andrew Frank
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD, 20814, United States
| | - Clifton L Dalgard
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD, 20814, United States
| | - Barrington G Burnett
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD, 20814, United States
| | - Thomas P Flagg
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD, 20814, United States
| |
Collapse
|
2
|
Wang CH, Hsu TR, Liu MY, Wang LY, Chou IJ, Lee WT, Liang WC, Lee IC, Chen HJ, Kao SM, Ho HC, Niu DM, Hsiao KJ, Chang MY, Hsieh HM, Jong YJ. Newborn screening facilitates early theranostics and improved spinal muscular atrophy outcome: five-year real-world evidence from Taiwan. Orphanet J Rare Dis 2025; 20:197. [PMID: 40275389 PMCID: PMC12023543 DOI: 10.1186/s13023-025-03697-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Recent findings indicate that infants with spinal muscular atrophy (SMA) treated early through newborn screening (NBS) have better outcomes. This study aimed to investigate the long-term outcomes of a 5-year SMA NBS program in Taiwan. RESULTS From September 2017 to August 2022, two NBS centers screened patients for SMN1 homozygous deletion using quantitative real-time polymerase chain reaction (RT-PCR) or the Sequenom MassARRAY platform and subsequently confirmed the findings using multiplex ligation-dependent probe amplification (MLPA). Implementation of SMA NBS using RT-PCR or MassARRAY platform efficiently led to the detection of neonates with homozygous survival motor neuron 1 (SMN1) deletions at a median age of 9 (range 4-14) days. Among the 446,966 newborns screened, 22 were detected to have a homozygous deletion of SMN1, followed by MLPA confirmation. One patient initially showed negative screening results but was later confirmed to have a compound heterozygous mutation. Among the 23 confirmed cases, 8 patients had two SMN2 copies (all classified as SMA type 1), 11 patients had three SMN2 copies (including 4 with SMA type 1, 2 with SMA type 2, 3 with SMA type 3, and 2 asymptomatic cases), and 4 patients had four SMN2 copies (all asymptomatic). The mean (median) follow-up duration for 19 survivors was 4.2 (5.0) years. All patients with two SMN2 copies developed symptoms within 62 days; those with three SMN2 copies experienced disease onset within 1 year. After diagnosis, most patients were on a watch and wait to receive disease-modifying therapy (DMT) due to initial lack of insurance coverage and limitations on indications after coverage was granted. Of the 19 children who received DMT, the outcomes included 12 walkers, 1 walker requiring support, 3 sitters, 1 non-sitter, and 2 patients with SMA type 1b with two SMN2 copies who succumbed to acute respiratory failure. CONCLUSIONS This 5-year SMA NBS study using RT-PCR or the MassARRAY platform, along with an extended follow-up, demonstrates that early diagnosis and prompt treatment can enhance SMA clinical outcomes and change its natural progression in the therapeutic era. Infants with NBS who received presymptomatic DMT had better clinical outcomes than those who received symptomatic DMT.
Collapse
Affiliation(s)
- Chen-Hua Wang
- Department of Pediatrics, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Translational Research Center of Neuromuscular Diseases, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ting-Rong Hsu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mei-Ying Liu
- Neonatal Screening Center, The Chinese Foundation of Health, Taipei, Taiwan
| | - Li-Yun Wang
- Neonatal Screening Center, Taipei Institute of Pathology, Taipei, Taiwan
| | - I-Jun Chou
- Department of Pediatric Neurology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Wang-Tso Lee
- Department of Pediatric Neurology, National Taiwan University Children's Hospital, Taipei, Taiwan
- Department of Pediatrics, and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Chen Liang
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Translational Research Center of Neuromuscular Diseases, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Inn-Chi Lee
- Division of Pediatric Neurology, Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiao-Jan Chen
- Neonatal Screening Center, The Chinese Foundation of Health, Taipei, Taiwan
| | - Shu-Min Kao
- Neonatal Screening Center, The Chinese Foundation of Health, Taipei, Taiwan
| | - Hui-Chen Ho
- Neonatal Screening Center, Taipei Institute of Pathology, Taipei, Taiwan
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kwang-Jen Hsiao
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Preventive Medicine Foundation, Taipei, Taiwan
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Ming-Yuh Chang
- Pediatric Neurology, Changhua Christian Children's Hospital, Changhua, Taiwan
| | - Hui-Min Hsieh
- Department of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuh-Jyh Jong
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Translational Research Center of Neuromuscular Diseases, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Center for Neurotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.
| |
Collapse
|
3
|
Yu H, Wei C, Sun D, Zhang L, Xia Y, Zhu W. Research trends on spinal muscular atrophy from 1995 to 2023: A bibliometric analysis. Medicine (Baltimore) 2025; 104:e41801. [PMID: 40153758 PMCID: PMC11957644 DOI: 10.1097/md.0000000000041801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/20/2025] [Indexed: 03/30/2025] Open
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is a neuromuscular disease characterized by progressive muscle weakness due to motor neuron degeneration. The discovery of the survival motor neuron 1 (SMN1) gene in 1995 revolutionized SMA research, leading to significant therapeutic advancements. This bibliometric analysis aimed to explore global trends in SMA research and therapy, with a particular focus on China. METHODS A comprehensive database search identified 4506 relevant publications (3812 articles, 694 reviews) published between 1995 and 2023. Bibliometric tools were used to analyze publication trends, collaborations, and research topics. RESULTS SMA research has experienced substantial growth, with the United States leading in publications followed by the United Kingdom and Germany. China has shown increasing engagement in this field. Key research areas include genetic and molecular mechanisms, survival motor neuron gene therapy, antisense oligonucleotides, and muscle strength-promoting factors. Chinese researchers have contributed significantly to these areas, with a higher reporting frequency of SMA-related topics compared to other countries. CONCLUSION This bibliometric analysis provides a comprehensive overview of global SMA research, highlighting significant advancements, and identifying future directions. The findings offer valuable insights for researchers, clinicians, and policymakers in China to ensure alignment with global medical advancements and improve the lives of individuals affected by SMA.
Collapse
Affiliation(s)
- Hao Yu
- Department of Medical Genetics and Center for Rare Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine and Zhejiang Key Laboratory of Rare Diseases for Precision Medicine and Clinical Translation, Hangzhou, China
| | - Cuijie Wei
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Dan Sun
- Department of Neurology, Wuhan Children’ s Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zhang
- Biogen Biotechnology (Shanghai) Co., Ltd, Shanghai, China
| | - Yanyan Xia
- Biogen Biotechnology (Shanghai) Co., Ltd, Shanghai, China
| | - Wenhua Zhu
- Department of Neurology, Huashan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
4
|
Malekpour SA, Kalirad A, Majidian S. Inferring the Selective History of CNVs Using a Maximum Likelihood Model. Genome Biol Evol 2025; 17:evaf050. [PMID: 40100752 PMCID: PMC11950529 DOI: 10.1093/gbe/evaf050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/27/2025] [Accepted: 03/13/2025] [Indexed: 03/20/2025] Open
Abstract
Copy number variations (CNVs)-structural variations generated by deletion and/or duplication that result in a change in DNA dosage-are prevalent in nature. CNVs can drastically affect the phenotype of an organism and have been shown to be both involved in genetic disorders and be used as raw material in adaptive evolution. Unlike single-nucleotide variations, the often large and varied effects of CNVs on phenotype hinders our ability to infer their selective advantage based on the population genetics data. Here, we present a likelihood-based approach, dubbed PoMoCNV (POlymorphism-aware phylogenetic MOdel for CNVs), that estimates the evolutionary parameters such as mutation rates among different copy numbers and relative fitness loss per copy deletion at a genomic locus based on population genetics data. As a case study, we analyze the genomics data of 40 strains of Caenorhabditis elegans, representing four different populations. We take advantage of the data on chromatin accessibility to interpret the mutation rate and fitness of copy numbers, as inferred by PoMoCNV, specifically in open or closed chromatin loci. We further test the reliability of PoMoCNV by estimating the evolutionary parameters of CNVs for mutation-accumulation experiments in C. elegans with varying levels of genetic drift.
Collapse
Affiliation(s)
- Seyed Amir Malekpour
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran 19395-5746, Iran
| | - Ata Kalirad
- Department for Integrative Evolutionary Biology, Max Planck Institute for Biology Tübingen, Tübingen 72076, Germany
| | - Sina Majidian
- SIB Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne 1015, Switzerland
| |
Collapse
|
5
|
Aziz B, Arif AA, Kazi K, Kirmani S, Ansar Z, Nasir A, Ibrahim SH, Ahmed KM, Hasan Z, Khan S. Analysis of spinal muscular atrophy patients from the spinal muscular atrophy and muscular dystrophy registry of Pakistan. J Neuromuscul Dis 2025; 12:260-270. [PMID: 39973405 DOI: 10.1177/22143602241301657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BackgroundSpinal Muscular Atrophy (SMA) leads to motor neuron loss, with progressive muscle weakness and wasting. Nationwide registries for neuromuscular diseases are pivotal for assessing epidemiology, preparing for clinical trials, and for adopting standardized management guidelines.ObjectivesThis paper aims to present data gathered during the establishment of Pakistan's inaugural registry for genetically confirmed SMA cases.MethodsIn this retrospective study, 215 participants with genetically confirmed SMA were recruited. Telephonic interviews were conducted to collect data for the Muscular Disease Registry of Pakistan that was analyzed using STATA version 17.0.ResultsSMA type 1 was the most common type (71.2%, n = 153). Amongst patients who were tested for survival motor neuron (SMN2) copies, the majority (84.4%, n = 168) had two SMN2 copies. SMA types were significantly associated with the ability to sit (p < 0.001) and walk (p < 0.001), and usage of a wheelchair (p = 0.0054). SMN2 copy numbers were significantly associated with the ability to sit (p = 0.020) and walk (p = 0.031).ConclusionsThis study highlights the high prevalence of SMA genotypes and phenotypes associated with severe disease in our population. Our findings reiterate the challenging prognosis for Pakistani children with SMA and underscore the necessity of the development of nationwide newborn screening programs and making treatments available.
Collapse
Affiliation(s)
- Bisma Aziz
- Department of Medicine, Aga Khan University Hospital, Karachi, Pakistan
| | - Ahmed A Arif
- Department of Public Health Sciences, University of North Carolina at Charlotte, USA
| | - Kulsum Kazi
- Department of Medicine, Aga Khan University Hospital, Karachi, Pakistan
| | - Salman Kirmani
- Chair and Director, Center of Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Zeeshan Ansar
- Department of Pathology & Lab Medicine, Aga Khan University Hospital, Karachi, Pakistan
| | - Asghar Nasir
- Department of Pathology & Lab Medicine, Aga Khan University Hospital, Karachi, Pakistan
| | - Shahnaz Hamid Ibrahim
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Pakistan
| | | | - Zahra Hasan
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| | - Sara Khan
- Department of Medicine, Aga Khan University Hospital, Karachi, Pakistan
| |
Collapse
|
6
|
Dong Y, Zhang S, Wang H, Jia X, Yu C, Li W, Ma X, Yu X, Li D, Shu J, Cai C. Delayed Diagnosis of Spinal Muscular Atrophy in Two Chinese Families due to Novel SMN1 Deletions. Am J Med Genet A 2025; 197:e63917. [PMID: 39497486 DOI: 10.1002/ajmg.a.63917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 09/20/2024] [Accepted: 10/12/2024] [Indexed: 02/13/2025]
Abstract
Autosomal recessive spinal muscular atrophy (SMA) is a leading cause of infant and child mortality, with homozygous deletion in exon 7 of the SMN1 gene being a major genetic cause. However, routine genetic testing methods may overlook structural variants outside of exon 7, potentially leading to misdiagnosis of SMA patients. Here, we reported two Chinese SMA patients who primarily exhibited developmental delays. Physical examinations revealed markedly reduced muscle strength and tone in their extremities, and electromyography suggested extensive neurogenic damage in the anterior horn of the spinal cord. The MLPA results indicated a single copy number of SMN1 in both patients, which is inconsistent with the typical genetic pattern of SMA. Through RNA sequencing and ultra-long read sequencing, we ultimately identified a rare structural variant involving the deletion of exons 2a-5 in both unrelated patients. This confirmed the presence of compound heterozygous variants in the SMN1 gene as the actual genetic cause. To our knowledge, this is the first case where a combination of RNA sequencing and ultra-long read sequencing has been used to diagnose SMA. We demonstrated the significant value of RNA sequencing in cases where children are highly suspected of having SMA but present negative results in routine genetic testing. This underscores the crucial role of accurate genetic testing methods in achieving early diagnosis of SMA.
Collapse
Affiliation(s)
- Yan Dong
- Graduate College, Tianjin Medical University, Tianjin, China
- Tianjin Children's Hospital, Children's Hospital of Tianjin University, Tianjin, China
| | - Shuyue Zhang
- Graduate College, Tianjin Medical University, Tianjin, China
- Tianjin Children's Hospital, Children's Hospital of Tianjin University, Tianjin, China
| | - Hong Wang
- Tianjin Children's Hospital, Children's Hospital of Tianjin University, Tianjin, China
- Department of Neurology, Tianjin Children's Hospital, Tianjin, China
| | - Xiaodong Jia
- Key Laboratory of Multi-Omics Precision Diagnosis Technology for Neurological Diseases in Tianjin, Tianjin Kingmed Center for Clinical Laboratory, Tianjin, China
- Tianjin Kingmed Center for Clinical Laboratory, Tianjin, China
| | - Changshun Yu
- Key Laboratory of Multi-Omics Precision Diagnosis Technology for Neurological Diseases in Tianjin, Tianjin Kingmed Center for Clinical Laboratory, Tianjin, China
- Tianjin Kingmed Center for Clinical Laboratory, Tianjin, China
| | - Weiran Li
- Graduate College, Tianjin Medical University, Tianjin, China
- Tianjin Children's Hospital, Children's Hospital of Tianjin University, Tianjin, China
| | - Ximeng Ma
- Graduate College, Tianjin Medical University, Tianjin, China
- Tianjin Children's Hospital, Children's Hospital of Tianjin University, Tianjin, China
| | - Xiaoli Yu
- Tianjin Children's Hospital, Children's Hospital of Tianjin University, Tianjin, China
- Department of Neurology, Tianjin Children's Hospital, Tianjin, China
| | - Dong Li
- Tianjin Children's Hospital, Children's Hospital of Tianjin University, Tianjin, China
- Department of Neurology, Tianjin Children's Hospital, Tianjin, China
| | - Jianbo Shu
- Tianjin Children's Hospital, Children's Hospital of Tianjin University, Tianjin, China
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital, Tianjin, China
- Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, China
| | - Chunquan Cai
- Tianjin Children's Hospital, Children's Hospital of Tianjin University, Tianjin, China
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital, Tianjin, China
- Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, China
| |
Collapse
|
7
|
de Almeida MMA, De Repentigny Y, Gagnon S, Sutton ER, Kothary R. Impact of liver-specific survival motor neuron (SMN) depletion on central nervous system and peripheral tissue pathology. eLife 2025; 13:RP99141. [PMID: 39976226 PMCID: PMC11841985 DOI: 10.7554/elife.99141] [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] [Indexed: 02/21/2025] Open
Abstract
Spinal muscular atrophy (SMA) is caused by mutations in the Survival Motor Neuron 1 (SMN1) gene. While traditionally viewed as a motor neuron disorder, there is involvement of various peripheral organs in SMA. Notably, fatty liver has been observed in SMA mouse models and SMA patients. Nevertheless, it remains unclear whether intrinsic depletion of SMN protein in the liver contributes to pathology in the peripheral or central nervous systems. To address this, we developed a mouse model with a liver-specific depletion of SMN by utilizing an Alb-Cre transgene together with one Smn2B allele and one Smn1 exon 7 allele flanked by loxP sites. Initially, we evaluated phenotypic changes in these mice at postnatal day 19 (P19), when the severe model of SMA, the Smn2B/- mice, exhibit many symptoms of the disease. The liver-specific SMN depletion does not induce motor neuron death, neuromuscular pathology or muscle atrophy, characteristics typically observed in the Smn2B/- mouse at P19. However, mild liver steatosis was observed, although no changes in liver function were detected. Notably, pancreatic alterations resembled that of Smn2B/-mice, with a decrease in insulin-producing β-cells and an increase in glucagon-producingα-cells, accompanied by a reduction in blood glucose and an increase in plasma glucagon and glucagon-like peptide (GLP-1). These changes were transient, as mice at P60 exhibited recovery of liver and pancreatic function. While the mosaic pattern of the Cre-mediated excision precludes definitive conclusions regarding the contribution of liver-specific SMN depletion to overall tissue pathology, our findings highlight an intricate connection between liver function and pancreatic abnormalities in SMA.
Collapse
Affiliation(s)
- Monique Marylin Alves de Almeida
- Regenerative Medicine Program, Ottawa Hospital Research InstituteOttawaCanada
- Centre for Neuromuscular Disease, University of OttawaOttawaCanada
| | - Yves De Repentigny
- Regenerative Medicine Program, Ottawa Hospital Research InstituteOttawaCanada
| | - Sabrina Gagnon
- Regenerative Medicine Program, Ottawa Hospital Research InstituteOttawaCanada
| | - Emma R Sutton
- Regenerative Medicine Program, Ottawa Hospital Research InstituteOttawaCanada
- Centre for Neuromuscular Disease, University of OttawaOttawaCanada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research InstituteOttawaCanada
- Centre for Neuromuscular Disease, University of OttawaOttawaCanada
- Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
- Department of Medicine, University of OttawaOttawaCanada
| |
Collapse
|
8
|
Schroth MK, Deans J, Bharucha Goebel DX, Burnette WB, Darras BT, Elsheikh BH, Felker MV, Klein A, Krueger J, Proud CM, Veerapandiyan A, Graham RJ. Spinal Muscular Atrophy Update in Best Practices: Recommendations for Treatment Considerations. Neurol Clin Pract 2025; 15:e200374. [PMID: 39399564 PMCID: PMC11464225 DOI: 10.1212/cpj.0000000000200374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/18/2024] [Indexed: 10/15/2024]
Abstract
Background and Objectives Spinal muscular atrophy (SMA) is an autosomal recessive disorder caused by biallelic variants of the Survival Motor Neuron 1 gene (SMN1) that affects approximately 1 in 15,000 live births. Availability of 3 SMN-enhancing treatments for SMA has led to urgency to review how clinicians and patients use these treatments for SMA, while additional research and real-world data and experience are being collected. This work describes important factors to assist with decision-making for SMN-enhancing treatments. Methods A systematic literature review was conducted on SMN-enhancing treatments for SMA and related studies. A working group of American and European health care providers with expertise in SMA care identified barriers and developed recommendations through a modified Delphi technique with serial surveys and feedback through virtual meetings to fill gaps for information where evidence is limited. A community working group of an individual living with SMA and caregivers provided insight and perspective on SMA treatments and support through a virtual meeting to guide recommendations. Results The health care provider working group and the community working group agreed that when determining whether to start, change, add, or discontinue a treatment, essential considerations include patient and family/caregiver perspective, and treatment safety and side effects. When initiating treatment for patients newly diagnosed with SMA, important patient characteristics are age and Survival Motor Neuron 2 gene (SMN2) copy number. Furthermore, when initiating, changing, or adding treatment, current clinical status and comorbidities drive decision-making. When considering a medication or treatment plan change, unless there is an urgent indication, a treatment and associated patient outcomes should be monitored for a minimum of 6-12 months. When determining a treatment plan with an adolescent or adult with SMA, consider factors such as quality of life, burden vs benefit of treatment, and reproductive issues. Access to care coordination and interdisciplinary/multidisciplinary care are essential to treatment success. Discussion Sharing information about current knowledge of treatments and shared decision-making between health care providers and patients living with SMA and caregivers are essential to overcoming barriers to providing SMN-enhancing treatments.
Collapse
Affiliation(s)
- Mary K Schroth
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - Jennifer Deans
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - Diana X Bharucha Goebel
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - W Bryan Burnette
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - Basil T Darras
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - Bakri H Elsheikh
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - Marcia V Felker
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - Andrea Klein
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - Jena Krueger
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - Crystal M Proud
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - Aravindhan Veerapandiyan
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| | - Robert J Graham
- Cure SMA (MKS); Clinical Care Education (JD), Cure SMA; Neurology and Pediatrics (DBG), Children's National, and National Institute of Neurological Diseases and Stroke, National Institutes of Health; Division of Neurology (WBB), Department of Pediatrics, Vanderbilt University; Department of Neurology (BTD), Boston Children's Hospital, Harvard Medical School; Department of Neurology (BHE), The Ohio State University Wexner Medical Center; Child Neurology (MVF), Indiana University; Division of Neuropediatrics (AK), Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital; Pediatric Neuromuscular (JK), Helen DeVos Children's Hospital; Neurology (CMP), Children's Hospital of the King's Daughters; Department of Pediatrics (AV), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital; and Department of Anesthesiology (RJG), Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School
| |
Collapse
|
9
|
De Felipe B, Delgado-Pecellin C, Lopez-Lobato M, Olbrich P, Blanco-Lobo P, Marquez-Fernandez J, Salamanca C, Mendoza B, Castro-Serrano R, Duque C, Moreno-Prieto M, Madruga-Garrido M, Lucena JM, Fernandez RM, Ruiz-Camacho M, Varona A, Neth O. Neonatal Screening for Spinal Muscular Atrophy and Severe T- and B-Cell Lymphopenias in Andalusia: A Prospective Study. Int J Neonatal Screen 2025; 11:11. [PMID: 39982345 PMCID: PMC11843956 DOI: 10.3390/ijns11010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/22/2025] Open
Abstract
Spinal muscular atrophy (SMA) and severe T- and/or B-cell lymphopenias (STBCL) in the form of severe combined immunodeficiencies (SCID) or X-linked agammaglobulinemia (XLA) are rare but potentially fatal pathologies. In January 2021, we initiated the first pilot study in Spain to evaluate the efficacy of a very early detection technique for SMA and SCID. RT-PCR was performed on prospectively collected dried blood spots (DBSs) from newborns in Western Andalusia (Spain). Internal and external controls (SCID, XLA and SMA) were included. The determination of SMA was relative (positive/negative) and that of TRECs and KRECs was quantitative (copies/punch). A total of 14.035 prospective samples were analysed. All controls were correctly identified while no cases of SMA or SCID/XLA were prospectively identified. DBS analysis of infants with suspected SMA or STBCL that presented to our centre showed pathological values in two cases each for SMA and SCID and one for XLA, all of them being subsequently confirmed genetically. In this prospective pilot study, no infants with SMA or STBCL were detected; however, the technique applied here was shown to be reliable and fast, further supporting the benefits and need to include SMA and SCID in national newborn screening (NBS) programs, as it will allow early supportive and curative therapy.
Collapse
Affiliation(s)
- Beatriz De Felipe
- Pediatrics Infectious Diseases, Rheumatology and Immunology Unit, Institute of Biomedicine of Seville, University Hospital Vírgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.D.F.); (P.O.); (P.B.-L.)
| | - Carmen Delgado-Pecellin
- Pediatrics Infectious Diseases, Rheumatology and Immunology Unit, Institute of Biomedicine of Seville, University Hospital Vírgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.D.F.); (P.O.); (P.B.-L.)
- Clinical Biochemistry Department, University Hospital Vírgen del Rocío, 41013 Seville, Spain
| | | | - Peter Olbrich
- Pediatrics Infectious Diseases, Rheumatology and Immunology Unit, Institute of Biomedicine of Seville, University Hospital Vírgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.D.F.); (P.O.); (P.B.-L.)
- Department of Pharmacology, Pediatrics and Radiology, University of Seville, 41008 Seville, Spain
| | - Pilar Blanco-Lobo
- Pediatrics Infectious Diseases, Rheumatology and Immunology Unit, Institute of Biomedicine of Seville, University Hospital Vírgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.D.F.); (P.O.); (P.B.-L.)
- Department of Pharmacology, Pediatrics and Radiology, University of Seville, 41008 Seville, Spain
| | | | - Carmen Salamanca
- Neonatology Unit, Hospital Universitario Virgen de Macarena, 41008 Seville, Spain
| | - Beatriz Mendoza
- Neonatology Service, Juan Ramón Jiménez Hospital, 21005 Huelva, Spain (M.R.-C.)
| | - Rocio Castro-Serrano
- Clinical Biochemistry Department, University Hospital Vírgen del Rocío, 41013 Seville, Spain
| | - Cristina Duque
- Neonatology Unit, University Hospital Vírgen del Rocío, 41013 Seville, Spain
| | - Mariana Moreno-Prieto
- Hospital Viamed Santa Angela de la Cruz, Sevilla and Neurolinkia, 41018 Seville, Spain (M.M.-G.)
| | - Marcos Madruga-Garrido
- Hospital Viamed Santa Angela de la Cruz, Sevilla and Neurolinkia, 41018 Seville, Spain (M.M.-G.)
| | - Jose M. Lucena
- Unidad de Inmunología, University Hospital Vírgen del Rocío, 41013 Seville, Spain
| | - Raquel M. Fernandez
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
| | - Maria Ruiz-Camacho
- Neonatology Service, Juan Ramón Jiménez Hospital, 21005 Huelva, Spain (M.R.-C.)
| | - Alberto Varona
- Paediatrics Service of Riotinto Hospital, 21660 Huelva, Spain
| | - Olaf Neth
- Pediatrics Infectious Diseases, Rheumatology and Immunology Unit, Institute of Biomedicine of Seville, University Hospital Vírgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain; (B.D.F.); (P.O.); (P.B.-L.)
| |
Collapse
|
10
|
Zhang W, Yin Y, Yang D, Liu M, Ye C, Yan R, Li R. Comprehensive analysis of adverse events associated with onasemnogene abeparvovec (Zolgensma) in spinal muscular atrophy patients: insights from FAERS database. Front Pharmacol 2025; 15:1475884. [PMID: 39840097 PMCID: PMC11747325 DOI: 10.3389/fphar.2024.1475884] [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: 08/04/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
Onasemnogene Abeparvovec (Zolgensma) is a gene therapy for the treatment of Spinal Muscular Atrophy (SMA) with improved motor neuron function and the potential for a singular treatment. Information on its adverse drug reactions is mainly from clinical trials and real-world studies with extensive sample sizes are lacking. In this study, we analyzed the U.S. Food and Drug Administration's Adverse Event Reporting System (FAERS) database to assess the drug safety profile of Zolgensma. A total of 1951 adverse event reports associated with onasemnogene abeparvovec (Zolgensma), containing 778 import important medical event (IME) signals, were identified from the FAERS database, and multiple disproportionate analysis algorithms were used to determine the significance of these adverse events. This study identified 281 onasemnogene abeparvovec-related adverse events (AEs), including some significant adverse events not mentioned in the product labelling. Elevated liver enzymes, fever, vomiting, and thrombocytopenia were the most common adverse reactions. Most adverse events manifested within the initial month of onasemnogene abeparvovec use, especially the first 8 days, but some may still occur after 1 year of treatment. Sex-specific scrutiny revealed differing risk levels for adverse events among women and men. Thrombocytopenia and thrombotic microangiopathy are more common in patients weighing ≥8.5 kg, and changes in renal function need to be closely monitored if thrombotic microangiopathy occurs. The above findings provide valuable insights into optimizing the utilization of onasemnogene abeparvovec, improving its effectiveness, and minimizing potential side effects, thereby greatly facilitating its practical application in clinical settings.
Collapse
Affiliation(s)
| | | | | | | | | | - Ruiling Yan
- Department of Fetal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ruiman Li
- Department of Fetal Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
11
|
Rodriguez-Torres R, Kanner CH, Gay EL, Uher D, Corbeil T, Coratti G, Dunaway Young S, Rohwer A, Muni Lofra R, McDermott MP, De Vivo DC, Wall MM, Glynn NW, Montes J. Development of the SMA EFFORT: A new approach to characterize perceived physical fatigability in spinal muscular atrophy. J Neuromuscul Dis 2025; 12:22143602241313326. [PMID: 39973458 DOI: 10.1177/22143602241313326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BACKGROUND Fatigue and fatigability are commonly reported in spinal muscular atrophy (SMA). Physical fatigability, proposed to be the most relevant to SMA pathophysiology, encompasses performance-based and perceived physical fatigability (PPF) assessments. While performance-based measures have highlighted physical fatigability as an SMA hallmark, PPF is not well characterized due to the lack of disease- and construct-specific scales. OBJECTIVE Our aim was to create a patient-reported outcome measure tailored for SMA, named the SMA EFFORT, to improve PPF assessment. Here, we describe the scale development process, assess scale properties, and discuss future research and application. METHODS SMA and scale development experts organized a comprehensive physical activity item bank, relevant across the SMA phenotypic spectrum. Activities were systematically categorized by varying intensities and durations. The SMA EFFORT was completed by an international cohort of individuals with all types of SMA. To compare PPF across demographic and clinical variables, SMA EFFORT PPF percent (PPF%) composite scoring was established. RESULTS One hundred eighteen participants completed the SMA EFFORT. Total PPF% scores were broadly distributed within functional groups, with differences between non-sitters (35.1 ± 21.0) and sitters (24.9 ± 15.1) (p = 0.006), and those with (34.4 ± 18.1) and without respiratory support (26.4 ± 17.8) (p = 0.02). Participants treated with disease modifying therapy (DMT) showed similar scores to those without treatment (p = 0.70). Further, no differences in scores were observed in participants with scoliosis surgery and those without (p = 0.71). Subscale analyses revealed differences in mean PPF% subscale scores by functional group. CONCLUSIONS The novel SMA EFFORT standardizes PPF ratings by anchoring activity to intensity and duration. Item and scale data insights will inform the next iteration, which will undergo additional investigation. The SMA EFFORT aims to improve upon current measures to better assess treatment impact on physical well-being across the SMA spectrum.
Collapse
Affiliation(s)
- Rafael Rodriguez-Torres
- Departments of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biobehavioral Sciences, Teacher's College, Columbia University, New York, NY, USA
| | - Cara H Kanner
- Departments of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biobehavioral Sciences, Teacher's College, Columbia University, New York, NY, USA
| | - Emma L Gay
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - David Uher
- Departments of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biobehavioral Sciences, Teacher's College, Columbia University, New York, NY, USA
| | - Thomas Corbeil
- Department of Psychiatry, Area Mental Health Data Science, Columbia University Irving Medical Center, New York, NY, USA
| | - Giorgia Coratti
- Centro Clinico Nemo, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sally Dunaway Young
- Department of Neurology and Clinical Neurosciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Annemarie Rohwer
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Robert Muni Lofra
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Michael P McDermott
- Departments of Biostatistics and Computational Biology, and Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Darryl C De Vivo
- Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Melanie M Wall
- Department of Psychiatry, Area Mental Health Data Science, Columbia University Irving Medical Center, New York, NY, USA
| | - Nancy W Glynn
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Jacqueline Montes
- Departments of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
12
|
Darras BT, Volpe JJ. Levels Above Lower Motor Neuron to Neuromuscular Junction. VOLPE'S NEUROLOGY OF THE NEWBORN 2025:1039-1073.e12. [DOI: 10.1016/b978-0-443-10513-5.00036-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
13
|
Horigome A, Takasago S, Arakawa R, Shimozawa K, Kaneshige M, Goishi K, Uryu H, Yamanaka J, Shichino H, Mizukami A. Evaluation of cardiac function in patients with SMA after treatment with onasemnogene abeparvovec. Pediatr Int 2025; 67:e70019. [PMID: 40365794 DOI: 10.1111/ped.70019] [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: 02/14/2023] [Revised: 07/31/2024] [Accepted: 09/16/2024] [Indexed: 05/15/2025]
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is a recessively inherited neurological disease resulting in motor neuron disorder. Onasemnogene abeparvovec is a gene replacement therapy used to treat patients with SMA. Cardiac toxicity was observed in animal studies on this therapy, and elevated cardiac troponin I levels were observed in clinical trials; however, the clinical importance of these findings is unknown. Therefore, this study aimed to reveal the cardiac toxicity of onasemnogene abeparvovec through careful investigation of cardiac function using cardiac strain analysis, which can detect early subtle abnormalities. METHODS This study included patients with SMA treated with onasemnogene abeparvovec between June 2020 and November 2020. Echocardiography, including peak global longitudinal strain (GLS), and other laboratory test results were evaluated. RESULTS Case 1 showed a relative GLS decrease of 14.5% compared with that at the baseline (GLS reduction from -22.1% to -18.9%), elevation of N-terminal prohormone B-type natriuretic peptide levels from 227 pg/mL to 494 pg/mL, and elevated liver enzyme concentrations after gene replacement therapy without reduction of the left ventricular ejection fraction. CONCLUSIONS Subclinical myocardial dysfunction after infusion of onasemnogene abeparvovec was suggested by careful investigation of cardiac function. Cardiac toxicity may correlate with liver impairment following gene replacement therapy. Long-term studies that allow for a higher number of patients and more extended observation periods should be conducted to confirm the cardiac toxicity of onasemnogene abeparvovec.
Collapse
Affiliation(s)
- Akihisa Horigome
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Satoshi Takasago
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Reiko Arakawa
- Department of Genomic Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Katsuyoshi Shimozawa
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Masao Kaneshige
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
- Department of Genomic Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Keiji Goishi
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Hideko Uryu
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Junko Yamanaka
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroyuki Shichino
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Ayumi Mizukami
- Department of Pediatrics, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Shi T, Zhou Z, Xiang T, Suo Y, Shi X, Li Y, Zhang P, Dai J, Sheng L. Cytoskeleton dysfunction of motor neuron in spinal muscular atrophy. J Neurol 2024; 272:19. [PMID: 39666039 PMCID: PMC11638312 DOI: 10.1007/s00415-024-12724-3] [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: 09/12/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024]
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by deletions or mutations of survival of motor neuron 1 (SMN1) gene. To date, the mechanism of selective cell death of motor neurons as a hallmark of SMA is still unclear. The severity of SMA is dependent on the amount of survival motor neuron (SMN) protein, which is an essential and ubiquitously expressed protein involved in various cellular processes including regulation of cytoskeletal dynamics. In this review, we discuss the effect of SMN ablation on cytoskeleton organization including actin dynamics, growth cone formation, axonal stability, neurite outgrowth, microtubule stability, synaptic vesicle dynamics and neurofilament protein release in SMA. We also summarized a list of critical proteins such as profilin-2 (PFN2), plastin-3 (PLS3), stathmin-1 (STMN1), microtubule-associated protein 1B (MAP1B) and neurofilament which play an important role in modulating cytoskeleton in SMA. Our aim is to highlight how cytoskeletal defects contribute to motor neuron degeneration in SMA disease progression and concentrating on cytoskeleton dynamics may be a promising approach to develop new therapy or biomarker.
Collapse
Affiliation(s)
- Tianyu Shi
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Zijie Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Taiyang Xiang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Yinxuan Suo
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Xiaoyan Shi
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
| | - Yaoyao Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Peng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Jun Dai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China.
| | - Lei Sheng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
15
|
Khanbazi A, Beheshtian M, Azad M, Akbari Kelishomi M, Afroozan F, Fatehi F, Noudehi K, Zamanian Najafabadi S, Omrani M, Habibi H, Taghdiri M, Abdi Rad I, Nafissi S, Jankhah A, Yazdan H, Daneshmand P, Saberi SH, Kahrizi K, Kariminejad A, Najmabadi H. Comprehensive copy number analysis of spinal muscular atrophy among the Iranian population. Sci Rep 2024; 14:29880. [PMID: 39622884 PMCID: PMC11612469 DOI: 10.1038/s41598-024-76815-x] [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: 05/16/2024] [Accepted: 10/16/2024] [Indexed: 12/06/2024] Open
Abstract
Copy number variations in the SMN1 gene on chromosome 5 are the primary cause of Spinal Muscular Atrophy (SMA) disease, characterized by muscle weakness and degeneration due to impaired alpha motor neurons in the spinal cord. To obtain a comprehensive molecular understanding of the SMA, including carriers, silent carriers, and patients in the Iranian population, we analyzed data from 5224 individuals referred to Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran, between 2006 and 2023 using MLPA and quantitative RT-PCR methods. The carrier frequency of SMA was estimated to be 5.55%. Furthermore, 3.06% of SMA parents (n = 24) had two copies of the SMN1 gene. Among 725 patients, those with an earlier onset of SMA were more likely to have two copies of the SMN2 gene (46.45%) and no copies of the NAIP gene (49.36%). Among the 654 fetal samples screened for SMA, 22.33% were found to be affected, while 3.46% of their parents tested normal. These findings are valuable for genetic counseling, carrier screening, and prenatal diagnosis of SMA in Iran. Furthermore, they underscore the importance of CNV analysis of SMN1, SMN2, and NAIP genes for accurate diagnosis and prognosis of SMA.
Collapse
Affiliation(s)
- Ali Khanbazi
- Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran
- Genetics Research Center, University of Social Welfare & Rehabilitation Sciences, Tehran, Iran
| | - Maryam Beheshtian
- Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran
- Genetics Research Center, University of Social Welfare & Rehabilitation Sciences, Tehran, Iran
| | - Maryam Azad
- Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran
| | | | - Fariba Afroozan
- Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran
| | - Fatemeh Fatehi
- Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran
| | - Khadijeh Noudehi
- Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran
| | | | - Mohammadamin Omrani
- Urology and Nephrology Research Center (UNRC), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Haleh Habibi
- Dr Habibi genetic counseling center, Hamedan, Iran
| | - Maryam Taghdiri
- Shiraz Genetic Counseling Center, Welfare Office, Shiraz, Iran
| | - Isa Abdi Rad
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Shahriar Nafissi
- Neuromuscular Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hilda Yazdan
- Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran
| | | | | | - Kimia Kahrizi
- Genetics Research Center, University of Social Welfare & Rehabilitation Sciences, Tehran, Iran
| | | | - Hossein Najmabadi
- Kariminejad - Najmabadi Pathology & Genetics Center, Tehran, Iran.
- Genetics Research Center, University of Social Welfare & Rehabilitation Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Goedeker NL, Rogers A, Fisher M, Arya K, Brandsema JF, Farah H, Farrar MA, Felker MV, Gibbons M, Hamid OA, Harmelink M, Herbert K, Kichula E, King K, Lakhotia A, Lee BH, Kuntz NL, Parsons J, Rehborg R, Veerapaniyan A, Zaidman CM. Outcomes of early-treated infants with spinal muscular atrophy: A multicenter, retrospective cohort study. Muscle Nerve 2024; 70:1247-1256. [PMID: 39370660 PMCID: PMC11560615 DOI: 10.1002/mus.28267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 10/08/2024]
Abstract
INTRODUCTION/AIMS While prompt identification and treatment of infants with spinal muscular atrophy (SMA) can ameliorate outcomes, variability persists. This study assessed management and outcomes of early-treated infants with SMA. METHODS We analyzed retrospective data at 12 centers on infants with SMA treated at age ≤6 weeks from August 2018 to December 2023. RESULTS Sixty-six patients, 35 with two SMN2 copies and 31 with ≥3 SMN2 copies, were included. Twenty-five (38%, 22 with two SMN2 copies), had SMA findings before initial treatment which was onasemnogene abeparvovec in 47 (71%) and nusinersen in 19 (29%). Thirty-two received sequential or combination treatments, including 16 adding nusinersen or risdiplam due to SMA findings following onasemnogene abeparvovec. All sat independently. Compared to children with ≥3 SMN2 copies, those with two SMN2 copies were less likely to walk (23/34 [68%] vs. 31/31 [100%], p < .001) and less likely to walk on time (9/34 [26%] vs. 29/31 [94%], p < .001); one non-ambulatory child was <18 months old and was excluded from this analysis. No patients required permanent ventilation or exclusively enteral nutrition; six required nocturnal non-invasive ventilation and four utilized supplemental enteral nutrition, all with two SMN2 copies. DISCUSSION Early treatment of infants with SMA can improve outcomes as indicated by our cohort, all of whom sat independently and are without permanent ventilation. However, our study demonstrates ongoing disability in most children with two SMN2 copies despite early monotherapy and emphasizes the need for additional research, including earlier monotherapy, initial combination therapy, prenatal treatment, and non-SMN modifying treatments.
Collapse
Affiliation(s)
- Natalie L. Goedeker
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO
| | - Amanda Rogers
- Department of Pediatrics, Norton Children’s Medical Group, University of Louisville, Louisville, KY
| | - Mark Fisher
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO
| | - Kapil Arya
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children’s Hospital, Little Rock, AR
| | - John F. Brandsema
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Hiba Farah
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Michelle A. Farrar
- Sydney Children’s Hospital Network and Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine, UNSW Sydney, Sydney, AUS
| | - Marcia V. Felker
- Division of Pediatric Neurology, Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Melissa Gibbons
- Department of Pediatrics Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado
| | - Omer Abdul Hamid
- Department of Pediatrics, Nemours Children’s Health, Orlando, FL
| | - Matthew Harmelink
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI
| | - Karen Herbert
- Sydney Children’s Hospital Network and Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine, UNSW Sydney, Sydney, AUS
| | - Elizabeth Kichula
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Kiana King
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Arpita Lakhotia
- Department of Pediatrics, Norton Children’s Medical Group, University of Louisville, Louisville, KY
| | - Bo Hoon Lee
- Department of Neurology, University of Rochester, Rochester, NY
| | - Nancy L. Kuntz
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Julie Parsons
- Department of Pediatrics Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado
| | - Rebecca Rehborg
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI
| | - Aravindhan Veerapaniyan
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children’s Hospital, Little Rock, AR
| | - Craig M. Zaidman
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO
| |
Collapse
|
17
|
Bzdok J, Czibere L, Burggraf S, Pauly N, Maier EM, Röschinger W, Becker M, Durner J. A Modular Genetic Approach to Newborn Screening from Spinal Muscular Atrophy to Sickle Cell Disease-Results from Six Years of Genetic Newborn Screening. Genes (Basel) 2024; 15:1467. [PMID: 39596667 PMCID: PMC11593867 DOI: 10.3390/genes15111467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Genetic newborn screening (NBS) has already entered the phase of common practice in many countries. In Germany, spinal muscular atrophy (SMA), severe combined immunodeficiency (SCID) and sickle cell disease (SCD) are currently a mandatory part of NBS. Here, we describe the experience of six years of genetic NBS including the prevalence of those three diseases in Germany. METHODS Samples and nucleic acids were extracted from dried blood spot cards, commonly used for NBS. A qPCR assay was used to detect disease-causing variants for SMA and SCD, and the detection of T-cell receptor excision circles (TRECs) was performed for SCID screening. RESULTS The results of the NBS of over 1 million newborns for SMA, approximately 770,000 for SCID and over 410,000 for SCD are discussed in detail. In these newborns, we have identified 121 cases of SMA, 15 cases of SCID and syndrome-based immunodeficiencies and 77 cases of SCD or β-thalassemia. CONCLUSIONS The flexibility of multiplex qPCR is assessed as an effective tool for incorporating different molecular genetic markers for screening. The processing of dried blood spot (DBS) filter cards for molecular genetic assays and the assays are described in detail; turn-around times and cost estimations are included to give an insight into the processes and discuss further options for optimization. The identified cases are in the range expected for the total number of screened newborns, but present a more exact view on the actual prevalences for Germany.
Collapse
Affiliation(s)
- Jessica Bzdok
- Department of Operative/Restorative Dentistry, Periodontology and Pedodontics, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
- Laboratory Labor Becker MVZ eGbR, 81671 Munich, Germany
| | | | | | - Natalie Pauly
- TIB Molbiol Syntheselabor GmbH, 12103 Berlin, Germany
| | | | | | - Marc Becker
- Department of Operative/Restorative Dentistry, Periodontology and Pedodontics, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
- Laboratory Labor Becker MVZ eGbR, 81671 Munich, Germany
| | - Jürgen Durner
- Department of Operative/Restorative Dentistry, Periodontology and Pedodontics, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
- Laboratory Labor Becker MVZ eGbR, 81671 Munich, Germany
| |
Collapse
|
18
|
Tachibana Y, Sato R, Makioka H, Hoshino M, Jin M. Safety and effectiveness of nusinersen, a treatment for spinal muscular atrophy, in 524 patients: results from an interim analysis of post-marketing surveillance in Japan. Int J Neurosci 2024; 134:1185-1197. [PMID: 37649429 DOI: 10.1080/00207454.2023.2251662] [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: 05/12/2023] [Revised: 07/31/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023]
Abstract
PURPOSE Nusinersen is an antisense oligonucleotide approved for the treatment of spinal muscular atrophy (SMA). A post-marketing surveillance (PMS) has been ongoing (August 2017-August 2025) in all patients in Japan who were administered nusinersen intrathecally in real-world clinical settings. We report the interim analysis results for safety and effectiveness. METHODS This interim analysis was conducted using data collected from 524 patients whose case report forms were obtained at least once by May 30, 2022. Collected data included patient demographics and adverse events (AEs) for safety, and motor function assessments and Clinical Global Impressions of Improvement (CGI-I) for effectiveness. RESULTS Of the 524 patients in the safety analysis set, 522 patients who were diagnosed with SMA were included in the effectiveness analysis (infantile-onset SMA [n = 153, 29.3%], later-onset SMA [n = 369, 70.7%]). The median duration of treatment was 785.0 (range 1-1549) days. AEs occurred in 35.9% of patients (49.0% in infantile-onset SMA and 30.6% in later-onset SMA). Nusinersen treatment significantly improved Hammersmith Infant Neurological Examination scores in patients with infantile-onset SMA and Hammersmith Functional Motor Scale-Expanded scores in patients with later-onset SMA for up to nearly 3 years. Based on CGI-I assessments, 98.5-100% of patients receiving nusinersen 'improved' or remain 'unchanged'. CONCLUSIONS This interim analysis of the large-scale, all-case PMS in patients who were administered nusinersen in Japan supports the safety and effectiveness of nusinersen. The benefit-risk balance of nusinersen treatment remains favorable.
Collapse
|
19
|
Tizzano EF, Quijano-Roy S, Servais L, Parsons JA, Aharoni S, Lakhotia A, Finkel RS. Outcomes for patients in the RESTORE registry with spinal muscular atrophy and four or more SMN2 gene copies treated with onasemnogene abeparvovec. Eur J Paediatr Neurol 2024; 53:18-24. [PMID: 39260228 DOI: 10.1016/j.ejpn.2024.08.006] [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: 04/25/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024]
Abstract
OBJECTIVE We describe outcomes following onasemnogene abeparvovec monotherapy for patients with ≥four survival motor neuron 2 (SMN2) gene copies in RESTORE, a noninterventional spinal muscular atrophy patient registry. METHODS We evaluated baseline characteristics, motor milestone achievement, post-treatment motor function, use of ventilatory/nutritional support, and adverse events as of December 22, 2022. RESULTS At data cutoff, 19 patients in RESTORE had ≥four SMN2 copies and were treated with onasemnogene abeparvovec monotherapy (n=12 [63.2%] four copies; n=7 [36.8%] >four copies). All patients were identified by newborn screening and were reported as asymptomatic at diagnosis. Median age at onasemnogene abeparvovec administration was 3.0 months. Median time from treatment to last recorded visit was 15.4 months, with a range of post-treatment follow-up of 0.03-39.4 months. All 12 children who were assessed for motor development achieved new milestones, including standing alone (n=2) and walking alone (n=5). Five children reported one or more treatment-emergent adverse events (one Grade 3 or greater). No deaths or use of ventilatory/nutritional support were reported. CONCLUSIONS Real-world findings from the RESTORE registry indicate that patients with ≥four SMN2 gene copies treated with onasemnogene abeparvovec monotherapy demonstrated improvements in motor function. Adverse events experienced by these patients were consistent with previously reported findings.
Collapse
Affiliation(s)
- Eduardo F Tizzano
- Department of Clinical and Molecular Genetics, Hospital Vall d'Hebron, Passeig de la Vall d'Hebron, 119-129, Horta-Guinardó, 08035, Barcelona, Spain.
| | - Susana Quijano-Roy
- Garches Neuromuscular Reference Center, APHP Raymond Poincaré University Hospital (UVSQ Paris Saclay), 104 Bd Raymond Poincaré, 92380, Garches, France
| | - Laurent Servais
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre, & NIHR Oxford Biomedical Research, University of Oxford, Headly Way, Headington, OX3 9DU, Oxford, UK; Department of Pediatrics, Neuromuscular Reference Center, University and University Hospital of Liège, Bât. B35 Département des Sciences Cliniques, Quartier Hôpital, Avenue de l'Hôpital 13, 4000, Liège, Belgium
| | - Julie A Parsons
- Children's Hospital Colorado, University of Colorado School of Medicine, 13001 East 17th Place, Aurora, CO, 80045, USA
| | - Sharon Aharoni
- Institute of Pediatric Neurology, Schneider Children's Medical Center of Israel, Kaplan St 14, Petah Tikva, Israel; Faculty of Medical and Health Sciences, Tel-Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Arpita Lakhotia
- University of Louisville, Norton Children's Medical Group, 411 East Chestnut Street, Floor 6, Louisville, KY, 40202, USA
| | - Richard S Finkel
- Center for Experimental Neurotherapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| |
Collapse
|
20
|
Reilly A, Beauvais A, Al-Aarg M, Yaworski R, Sutton ER, Thebault S, Kothary R. Peripheral defects precede neuromuscular pathology in the Smn2B/- mouse model of spinal muscular atrophy. J Neuromuscul Dis 2024; 11:1200-1210. [PMID: 39973467 DOI: 10.1177/22143602241288036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BACKGROUND Spinal Muscular Atrophy (SMA) is an inherited neurodegenerative disease caused by the loss or mutation of the survival motor neuron 1 (SMN1) gene. Though classically regarded as a motor neuron disorder, reports are increasingly describing the involvement of non-neuronal organs in SMA. The Smn2B/- mouse is a model of SMA that displays a peripheral phenotype including metabolic defects. OBJECTIVE Here, we characterized several neuronal and non-neuronal defects in the Smn2B/- mouse throughout development to better understand the progression of the disease and the relationship between tissue defects. METHODS We collected tissues from mutant Smn2B/- mice and Smn2B/+ littermate controls at several timepoints and evaluated spinal cord motor neuron loss, neuromuscular junction pathology, muscle fiber size, liver steatosis, and pancreatic islet cell composition. Blood glucose and plasma neurofilament light chain (NfL) were also measured. RESULTS Smn2B/- mice displayed several peripheral defects prior to motor neuron loss and showed early elevations in neurofilament light chain (NfL) protein. CONCLUSIONS This work provides an important framework for guiding future research with this mouse model and demonstrates that the liver may be an early target in the development of SMA.
Collapse
Affiliation(s)
- Aoife Reilly
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Canada
| | - Ariane Beauvais
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Majd Al-Aarg
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Faculty of Science, University of Ottawa, Ottawa, Canada
| | - Rebecca Yaworski
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Canada
| | - Emma R Sutton
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Canada
| | - Simon Thebault
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Canada
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
21
|
Maretina M, Koroleva V, Shchugareva L, Glotov A, Kiselev A. The Relevance of Spinal Muscular Atrophy Biomarkers in the Treatment Era. Biomedicines 2024; 12:2486. [PMID: 39595052 PMCID: PMC11591959 DOI: 10.3390/biomedicines12112486] [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: 10/01/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a severe neuromuscular disorder that currently has an approved treatment for all forms of the disease. Previously, biomarkers were primarily used for diagnostic purposes, such as detecting the presence of the disease or determining a specific clinical type of SMA. Currently, with the availability of therapy, biomarkers have become more valuable due to their potential for prognostic, predictive, and pharmacodynamic applications. This review describes the most promising physiological, functional, imaging and molecular biomarkers for SMA, derived from different patients' tissues. The review summarizes information about classical biomarkers that are already used in clinical practice as well as fresh findings on promising biomarkers that have been recently disclosed. It highlights the usefulness, limitations, and strengths of each potential biomarker, indicating the purposes for which each is best suited and when combining them may be most beneficial.
Collapse
Affiliation(s)
- Marianna Maretina
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia; (M.M.); (A.G.)
| | - Valeria Koroleva
- Municipal Hospital for Children No. 1, 198205 Saint-Petersburg, Russia; (V.K.); (L.S.)
| | - Lyudmila Shchugareva
- Municipal Hospital for Children No. 1, 198205 Saint-Petersburg, Russia; (V.K.); (L.S.)
- Department of Pediatric Neuropathology and Neurosurgery, North-Western State Medical University Named After I.I. Mechnikov, 191015 Saint-Petersburg, Russia
| | - Andrey Glotov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia; (M.M.); (A.G.)
| | - Anton Kiselev
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia; (M.M.); (A.G.)
| |
Collapse
|
22
|
Barbo M, Koritnik B, Leonardis L, Blagus T, Dolžan V, Ravnik-Glavač M. Genetic Variability in Oxidative Stress, Inflammatory, and Neurodevelopmental Pathways: Impact on the Susceptibility and Course of Spinal Muscular Atrophy. Cell Mol Neurobiol 2024; 44:71. [PMID: 39463208 PMCID: PMC11513727 DOI: 10.1007/s10571-024-01508-y] [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: 07/12/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024]
Abstract
The spinal muscular atrophy (SMA) phenotype strongly correlates with the SMN2 gene copy number. However, the severity and progression of the disease vary widely even among affected individuals with identical copy numbers. This study aimed to investigate the impact of genetic variability in oxidative stress, inflammatory, and neurodevelopmental pathways on SMA susceptibility and clinical progression. Genotyping for 31 genetic variants across 20 genes was conducted in 54 SMA patients and 163 healthy controls. Our results revealed associations between specific polymorphisms and SMA susceptibility, disease type, age at symptom onset, and motor and respiratory function. Notably, the TNF rs1800629 and BDNF rs6265 polymorphisms demonstrated a protective effect against SMA susceptibility, whereas the IL6 rs1800795 was associated with an increased risk. The polymorphisms CARD8 rs2043211 and BDNF rs6265 were associated with SMA type, while SOD2 rs4880, CAT rs1001179, and MIR146A rs2910164 were associated with age at onset of symptoms after adjustment for clinical parameters. In addition, GPX1 rs1050450 and HMOX1 rs2071747 were associated with motor function scores and lung function scores, while MIR146A rs2910164, NOTCH rs367398 SNPs, and GSTM1 deletion were associated with motor and upper limb function scores, and BDNF rs6265 was associated with lung function scores after adjustment. These findings emphasize the potential of genetic variability in oxidative stress, inflammatory processes, and neurodevelopmental pathways to elucidate the complex course of SMA. Further exploration of these pathways offers a promising avenue for developing personalized therapeutic strategies for SMA patients.
Collapse
Affiliation(s)
- Maruša Barbo
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Blaž Koritnik
- Institute of Clinical Neurophysiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Department of Neurology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Lea Leonardis
- Institute of Clinical Neurophysiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Department of Neurology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Blagus
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vita Dolžan
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Metka Ravnik-Glavač
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
- , Ljubljana, Slovenia.
| |
Collapse
|
23
|
Barbo M, Glavač D, Jezernik G, Ravnik-Glavač M. MicroRNAs as Biomarkers in Spinal Muscular Atrophy. Biomedicines 2024; 12:2428. [PMID: 39594995 PMCID: PMC11592373 DOI: 10.3390/biomedicines12112428] [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: 07/31/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a severe neurodegenerative disease caused by the loss of the survival motor neuron (SMN) protein, leading to degeneration of anterior motor neurons and resulting in progressive muscle weakness and atrophy. Given that SMA has a single, well-defined genetic cause, gene-targeted therapies have been developed, aiming to increase SMN production in SMA patients. The SMN protein is likely involved in the synthesis of microRNAs (miRNAs), and dysregulated miRNA expression is increasingly associated with the pathophysiology of SMA. Currently, there is a lack of reliable biomarkers to monitor SMA; therefore, the search for novel SMA biomarkers, including miRNAs, is crucial as reliable tools are needed to track disease progression, predict the response to therapy and understand the different clinical outcomes of available treatments. In this review, we compile data on miRNAs associated with SMA pathogenesis and their potential use as biomarkers. Based on current knowledge, the most frequently deregulated miRNAs between SMA patients and controls, as well as pre- and post-treatment in SMA patients, include miR-1-3p, miR-133a-3p, miR-133b, and miR-206. These findings offer promising possibilities for improving patient classification and monitoring disease progression and response to treatment. Additionally, these findings provide insights into the broader molecular mechanisms and networks of SMA that could inform the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Maruša Barbo
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
| | - Damjan Glavač
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (D.G.); (G.J.)
- Department of Molecular Genetics, Institute of Pathology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Gregor Jezernik
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (D.G.); (G.J.)
| | - Metka Ravnik-Glavač
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
| |
Collapse
|
24
|
Aldukain M, Aldukain A, Hobani A, Barakat A, Alsayyed L, Alomar M, Zain Al-Abeden MS, Alzoum N, Asseri AA. The Impact of Nusinersen Treatment on Respiratory Function in Patients with Spinal Muscular Atrophy: A Systematic Review. J Clin Med 2024; 13:6306. [PMID: 39518446 PMCID: PMC11546051 DOI: 10.3390/jcm13216306] [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: 08/31/2024] [Revised: 10/02/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: This systematic review evaluated the impact of nusinersen therapy on respiratory health and function in individuals with spinal muscular atrophy (SMA) and determined whether nusinersen improves pulmonary function, focusing on differences based on patient age and the timing of treatment initiation. Methods: A systematic search of PubMed, Ovid Medline, ScienceDirect, and Web of Science databases was conducted up to January 2024 in accordance with the PRISMA guidelines. Thirteen studies were included, comprising clinical trials, observational studies, and case series that focused on respiratory outcomes in SMA patients treated with nusinersen. The data on study design, participant characteristics, nusinersen intervention, respiratory outcomes, and adverse events were extracted. The Joanna Briggs Institute Critical Appraisal Tool was used to assess study quality. A narrative synthesis was conducted to address the heterogeneity of the studies. Results: This review found a general trend of improvement in pulmonary function, specifically in forced vital capacity (FVC), although the extent and duration of improvement varied across the studies. Peak cough flow (PCF) and peak expiratory flow (PEF) showed positive trends in some studies, although the results were not consistently significant. Respiratory function improvements were frequently observed, particularly in younger patients and those treated earlier. Conclusions: Nusinersen appears to enhance respiratory function and improve motor outcomes in SMA patients, especially with early treatment. However, further research is needed to fully understand its mechanisms and long-term effects on respiratory health in SMA.
Collapse
Affiliation(s)
- Mona Aldukain
- Faculty of Medicine, King Khaled University, Abha 62521, Saudi Arabia; (M.A.); (A.A.); (L.A.)
| | - Ali Aldukain
- Faculty of Medicine, King Khaled University, Abha 62521, Saudi Arabia; (M.A.); (A.A.); (L.A.)
| | - Assal Hobani
- College of Medicine, Ibn Sina National College, Jeddah 22421, Saudi Arabia;
| | - Abdulmalik Barakat
- Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Lujain Alsayyed
- Faculty of Medicine, King Khaled University, Abha 62521, Saudi Arabia; (M.A.); (A.A.); (L.A.)
| | - Maher Alomar
- College of Medicine, Sulaiman Al Rajhi University, Al-Bukairiyah 52726, Saudi Arabia; (M.A.); (M.S.Z.A.-A.)
| | - Maha Saad Zain Al-Abeden
- College of Medicine, Sulaiman Al Rajhi University, Al-Bukairiyah 52726, Saudi Arabia; (M.A.); (M.S.Z.A.-A.)
| | - Nora Alzoum
- College of Medicine, Princess Nourah bint Abdulrahaman University, Riyadh 11671, Saudi Arabia;
| | - Ali Alsuheel Asseri
- Department of Child Health, College of Medicine, King Khalid University, Abha 62529, Saudi Arabia
| |
Collapse
|
25
|
Barrois R, Tervil B, Cacioppo M, Barnerias C, Deladrière E, Leloup-Germa V, Hervé A, Oudre L, Ricard D, Vidal PP, Vayatis N, Roy SQ, Brochard S, Gitiaux C, Desguerre I. Acceptability, validity and responsiveness of inertial measurement units for assessing motor recovery after gene therapy in infants with early onset spinal muscular atrophy: a prospective cohort study. J Neuroeng Rehabil 2024; 21:183. [PMID: 39415296 PMCID: PMC11483959 DOI: 10.1186/s12984-024-01477-9] [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: 03/26/2024] [Accepted: 09/24/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Onasemnogene abeparvovec gene replacement therapy (GT) has changed the prognosis of patients with spinal muscular atrophy (SMA) with variable outcome regarding motor development in symptomatic patients. This pilot study evaluates acceptability, validity and clinical relevance of Inertial Measurement Units (IMU) to monitor spontaneous movement recovery in early onset SMA patients after GT. METHODS Clinical assessments including CHOPINTEND score (the gold standard motor score for infants with SMA) and IMU measurements were performed before (M0) and repeatedly after GT. Inertial data was recorded during a 25-min spontaneous movement task, the child lying on the back, without (10 min) and with a playset (15 min) wearing IMUs. Two commonly used parameters, norm acceleration 95th centile (||A||_95) and counts per minute (||A||_CPM) were computed for each wrist, elbow and foot sensors. RESULTS 23 SMA-patients were included (mean age at diagnosis 8 months [min 2, max 20], 19 SMA type 1, three type 2 and one presymptomatic) and 104 IMU-measurements were performed, all well accepted by families and 84/104 with a good child participation (evaluated with Brazelton scale). ||A||_95 and ||A||_CPM showed high internal consistency (without versus with a playset) with interclass correlation coefficient for the wrist sensors of 0.88 and 0.85 respectively and for the foot sensors of 0.93 and 0.91 respectively. ||A||_95 and ||A||_CPM were strongly correlated with CHOPINTEND (r for wrist sensors 0.74 and 0.67 respectively and for foot sensors 0.61 and 0.68 respectively, p-values < 0.001). ||A||_95 for the foot, the wrist, the elbow sensors and ||A||_CPM for the foot, the wrist, the elbow sensors increased significantly between baseline and the 12 months follow-up visit (respective p-values: 0.004, < 0.001, < 0.001, 0.006, < 0.001, < 0.001). CONCLUSION IMUs were well accepted, consistent, concurrently valid, responsive and associated with unaided sitting acquisition especially for the elbow sensors. This study is the first reporting a large set of inertial sensor derived data after GT in SMA patients and paves the way for IMU-based follow-up of SMA patients after treatment.
Collapse
Affiliation(s)
- R Barrois
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, AP-HP, Hôpital Necker-Enfants Malades, 149 Rue de Sèvres, 75015, Paris, France.
- Clinical Neurophysiology Department, AP-HP, Hôpital Necker Enfants Malades, Paris, France.
- Université Paris Saclay, Université Paris Cité, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, 91190, Gif-Sur-Yvette, France.
- Service d'explorations Fonctionnelles, Unité de Neurophysiologie Clinique, AP-HP Hôpital Necker, 149 Rue de Sèvres, 75015, Paris, France.
| | - B Tervil
- Université Paris Saclay, Université Paris Cité, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, 91190, Gif-Sur-Yvette, France
| | - M Cacioppo
- Pediatric Neurology Unit, Children's Hospital, Geneva University Hospitals, 1205, Geneva, Switzerland
- LaTIM UMR 1101 Laboratory, Inserm, Brest, France
| | - C Barnerias
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, AP-HP, Hôpital Necker-Enfants Malades, 149 Rue de Sèvres, 75015, Paris, France
| | - E Deladrière
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, AP-HP, Hôpital Necker-Enfants Malades, 149 Rue de Sèvres, 75015, Paris, France
| | - V Leloup-Germa
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, AP-HP, Hôpital Necker-Enfants Malades, 149 Rue de Sèvres, 75015, Paris, France
| | - A Hervé
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, AP-HP, Hôpital Necker-Enfants Malades, 149 Rue de Sèvres, 75015, Paris, France
| | - L Oudre
- Université Paris Saclay, Université Paris Cité, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, 91190, Gif-Sur-Yvette, France
| | - D Ricard
- Université Paris Saclay, Université Paris Cité, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, 91190, Gif-Sur-Yvette, France
- Service de Neurologie, HIA Percy, Service de Santé des Armées, Clamart, France
| | - P P Vidal
- Université Paris Saclay, Université Paris Cité, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, 91190, Gif-Sur-Yvette, France
| | - N Vayatis
- Université Paris Saclay, Université Paris Cité, ENS Paris Saclay, CNRS, SSA, INSERM, Centre Borelli, 91190, Gif-Sur-Yvette, France
| | - S Quijano Roy
- Pediatric Neurology and ICU Department, Garches Reference Center for Neuromuscular Diseases, AP-HP Paris-Saclay Université, Hôpital Raymond Poincaré (UVSQ), 104 Boulevard Raymond Poincaré, 92380, Garches, France
| | - S Brochard
- LaTIM UMR 1101 Laboratory, Inserm, Brest, France
- University Hospital of Brest, Brest, France
| | - C Gitiaux
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, AP-HP, Hôpital Necker-Enfants Malades, 149 Rue de Sèvres, 75015, Paris, France
- Clinical Neurophysiology Department, AP-HP, Hôpital Necker Enfants Malades, Paris, France
| | - I Desguerre
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, AP-HP, Hôpital Necker-Enfants Malades, 149 Rue de Sèvres, 75015, Paris, France
- Paris Cité University, IHU Imagine, 75015, Paris, France
| |
Collapse
|
26
|
Kruse T, Leflerovà D, Cap A, Portegys S, Wirth B, Heller R, Brakemeier S, Hagenacker T, Braumann B, Wunderlich G. Oral functions in adult persons with spinal muscular atrophy compared to a healthy control group: a prospective cross-sectional study with a multimodal approach. Orphanet J Rare Dis 2024; 19:382. [PMID: 39407277 PMCID: PMC11481369 DOI: 10.1186/s13023-024-03405-5] [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: 07/18/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Oral function tests have been shown to reliably detect impaired bulbar function in adults with spinal muscular atrophy (SMA). Although not routinely recorded, it is known that persons with SMA are affected to varying degrees. Detecting differences in bite and tongue force, endurance, and maximum mouth opening has become particularly promising since the introduction of causal therapy for SMA. This study aimed to compare oral function among adult persons with SMA with different SMA types, walking abilities, and treatment status to a healthy control group. METHODS Data from oral function tests conducted on 58 persons with SMA and 45 healthy individuals were analyzed. Differences in oral function between SMA subgroups were pairwise tested and compared to the healthy control group using Wilcoxon rank sum tests. RESULTS In an overall comparison, three out of five oral function tests revealed lower values for the SMA group compared to the control group. Subgroup analyses indicated lower scores for most oral function tests in non-ambulatory, untreated patients with SMA type 2 compared to controls. Ambulatory, treated patients with SMA type 3 achieved strength and endurance values comparable to those of healthy individuals. CONCLUSIONS The impairment of oral function varies across persons with SMA. Routine measurement of oral function is warranted to determine individual bulbar involvement stages. Further evaluation should be scheduled if indicators such as restricted maximum mouth opening arise. Trial registration DRKS, DRKS00015842. Registered 30 July 2019, https://drks.de/register/de/trial/DRKS00015842/preview .
Collapse
Affiliation(s)
- Teresa Kruse
- Department of Orthodontics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 32, 50931, Cologne, Germany.
- Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany.
| | - Diana Leflerovà
- Department of Orthodontics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 32, 50931, Cologne, Germany
| | - Annette Cap
- Department of Orthodontics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 32, 50931, Cologne, Germany
| | - Sara Portegys
- Department of Orthodontics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 32, 50931, Cologne, Germany
| | - Brunhilde Wirth
- Department of Orthodontics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 32, 50931, Cologne, Germany
- Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Genetics, University of Cologne, Cologne, Germany
| | - Raoul Heller
- Department of Orthodontics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 32, 50931, Cologne, Germany
- Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Genetics, University of Cologne, Cologne, Germany
- Genetic Health Service NZ - Northern Hub, Auckland District Health Board, Auckland City Hospital, 90-102 Grafton Rd, Grafton, Auckland, 1010, New Zealand
| | - Svenja Brakemeier
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Tim Hagenacker
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Bert Braumann
- Department of Orthodontics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 32, 50931, Cologne, Germany
- Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | - Gilbert Wunderlich
- Department of Orthodontics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 32, 50931, Cologne, Germany
- Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
27
|
Zhang Q, Hong Y, Brusa C, Scoto M, Cornell N, Patel P, Baranello G, Muntoni F, Zhou H. Profiling neuroinflammatory markers and response to nusinersen in paediatric spinal muscular atrophy. Sci Rep 2024; 14:23491. [PMID: 39379509 PMCID: PMC11461652 DOI: 10.1038/s41598-024-74338-z] [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: 07/25/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024] Open
Abstract
Neuroinflammation is an emerging clinical feature in spinal muscular atrophy (SMA). Characterizing neuroinflammatory cytokines in cerebrospinal fluid (CSF) in SMA and their response to nusinersen is important for identifying new biomarkers and understanding the pathophysiology of SMA. We measured twenty-seven neuroinflammatory markers in CSF from twenty SMA children at different time points, and correlated the findings with motor function improvement. At baseline, MCP-1, IL-7 and IL-8 were significantly increased in SMA1 patients compared to SMA2, and were significantly correlated with disease severity. After six months of nusinersen treatment, CSF levels of eotaxin and MIP-1β were markedly reduced, while IL-2, IL-4 and VEGF-A were increased. The decreases in eotaxin and MIP-1β were associated with changes in motor scores in SMA1. We also detected a transient increase in MCP-1, MDC, MIP-1α, IL-12/IL-23p40 and IL-8 after the first or second injection of nusinersen, followed by a steady return to baseline levels within six months. Our study provides a detailed profile of neuroinflammatory markers in SMA CSF. Our data confirms the potential of MCP-1, eotaxin and MIP-1β as new neuroinflammatory biomarkers in SMA1 and indicates the presence of a subtle inflammatory response to nusinersen during the early phase of treatment.
Collapse
Affiliation(s)
- Qiang Zhang
- Genetics and Genomic Medicine Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
- School of Physical Education, Huangshan University, Huangshan, China
| | - Ying Hong
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Chiara Brusa
- Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, The Dubowitz Neuromuscular Centre, University College London, London, UK
| | - Mariacristina Scoto
- Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, The Dubowitz Neuromuscular Centre, University College London, London, UK
| | - Nikki Cornell
- Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, The Dubowitz Neuromuscular Centre, University College London, London, UK
| | - Parth Patel
- Genetics and Genomic Medicine Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Giovanni Baranello
- Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, The Dubowitz Neuromuscular Centre, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Francesco Muntoni
- Developmental Neurosciences Research and Teaching Department, Great Ormond Street Institute of Child Health, The Dubowitz Neuromuscular Centre, University College London, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Haiyan Zhou
- Genetics and Genomic Medicine Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK.
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
28
|
Gandhi G, Kodiappan R, Abdullah S, Teoh HK, Tai L, Cheong SK, Yeo WWY. Revealing the potential role of hsa-miR-663a in modulating the PI3K-Akt signaling pathway via miRNA microarray in spinal muscular atrophy patient fibroblast-derived iPSCs. J Neuropathol Exp Neurol 2024; 83:822-832. [PMID: 38894621 DOI: 10.1093/jnen/nlae065] [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] [Indexed: 06/21/2024] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder due to deletion or mutation of survival motor neuron 1 (SMN1) gene. Although survival motor neuron 2 (SMN2) gene is still present in SMA patients, the production of full-length survival motor neuron (SMN) protein is insufficient owing to missing or mutated SMN1. No current disease-modifying therapies can cure SMA. The aim of this study was to explore microRNA (miRNA)-based therapies that may serve as a potential target for therapeutic intervention in delaying SMA progression or as treatment. The study screened for potentially dysregulated miRNAs in SMA fibroblast-derived iPSCs using miRNA microarray. Results from the miRNA microarray were validated using quantitative reverse transcription polymerase chain reaction. Bioinformatics analysis using various databases was performed to predict the potential putative gene targeted by hsa-miR-663a. The findings showed differential expression of hsa-miR-663a in SMA patients in relation to a healthy control. Bioinformatics analysis identified GNG7, IGF2, and TNN genes that were targeted by hsa-miR-663a to be involved in the PI3K-AKT pathway, which may be associated with disease progression in SMA. Thus, this study suggests the potential role of hsa-miR-663a as therapeutic target for the treatment of SMA patients in the near future.
Collapse
Affiliation(s)
- Gayatri Gandhi
- Perdana University Graduate School of Medicine, Perdana University, Kuala Lumpur, Malaysia
| | - Radha Kodiappan
- Department of Research and Training, MAHSA Specialist Hospital, Selangor, Malaysia
| | - Syahril Abdullah
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Genetics & Regenerative Medicine Research Group, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Malaysia Genome and Vaccine Institute, National Institutes of Biotechnology Malaysia, Selangor, Malaysia
| | - Hoon Koon Teoh
- Centre for Stem Cell Research, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Lihui Tai
- Centre for Stem Cell Research, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
- Cytopeutics Sdn. Bhd, Selangor, Malaysia
| | - Soon Keng Cheong
- Centre for Stem Cell Research, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Wendy Wai Yeng Yeo
- Perdana University Graduate School of Medicine, Perdana University, Kuala Lumpur, Malaysia
- School of Pharmacy, Monash University Malaysia, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
29
|
Schüning T, Zeug A, Strienke K, Franz P, Tsiavaliaris G, Hensel N, Viero G, Ponimaskin E, Claus P. The spinal muscular atrophy gene product regulates actin dynamics. FASEB J 2024; 38:e70055. [PMID: 39305126 DOI: 10.1096/fj.202300183r] [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: 02/01/2023] [Revised: 07/31/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024]
Abstract
Spinal Muscular Atrophy (SMA) is a neuromuscular disease caused by low levels of the Survival of Motoneuron (SMN) protein. SMN interacts with and regulates the actin-binding protein profilin2a, thereby influencing actin dynamics. Dysfunctional actin dynamics caused by SMN loss disrupts neurite outgrowth, axonal pathfinding, and formation of functional synapses in neurons. Whether the SMN protein directly interacts with and regulates filamentous (F-) and monomeric globular (G-) actin is still elusive. In a quantitative single cell approach, we show that SMN loss leads to dysregulated F-/G-actin fractions. Furthermore, quantitative assessment of cell morphology suggests an F-actin organizational defect. Interestingly, this is mediated by an interaction of SMN with G- and F-actin. In co-immunoprecipitation, in-vitro pulldown and co-localization assays, we elucidated that this interaction is independent of the SMN-profilin2a interaction. Therefore, we suggest two populations being relevant for functional actin dynamics in healthy neurons: SMN-profilin2a-actin and SMN-actin. Additionally, those two populations may influence each other and therefore regulate binding of SMN to actin. In SMA, we showed a dysregulated co-localization pattern of SMN-actin which could only partially rescued by SMN restoration. However, dysregulation of F-/G-actin fractions was reduced by SMN restoration. Taken together, our results suggest a novel molecular function of SMN in binding to actin independent from SMN-profilin2a interaction.
Collapse
Affiliation(s)
- Tobias Schüning
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Hannover, Germany
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Andre Zeug
- Institute of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Katharina Strienke
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Hannover, Germany
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Peter Franz
- Cellular Biophysics, Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Georgios Tsiavaliaris
- Cellular Biophysics, Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Niko Hensel
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Gabriella Viero
- Institute of Biophysics (IBF), CNR Unit at Trento, Trento, Italy
| | - Evgeni Ponimaskin
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Peter Claus
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
30
|
Nishio H, Niba ETE, Saito T, Okamoto K, Lee T, Takeshima Y, Awano H, Lai PS. Clinical and Genetic Profiles of 5q- and Non-5q-Spinal Muscular Atrophy Diseases in Pediatric Patients. Genes (Basel) 2024; 15:1294. [PMID: 39457418 PMCID: PMC11506990 DOI: 10.3390/genes15101294] [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: 08/07/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is a genetic disease characterized by loss of motor neurons in the spinal cord and lower brainstem. The term "SMA" usually refers to the most common form, 5q-SMA, which is caused by biallelic mutations in SMN1 (located on chromosome 5q13). However, long before the discovery of SMN1, it was known that other forms of SMA existed. Therefore, SMA is currently divided into two groups: 5q-SMA and non-5q-SMA. This is a simple and practical classification, and therapeutic drugs have only been developed for 5q-SMA (nusinersen, onasemnogene abeparvovec, risdiplam) and not for non-5q-SMA disease. METHODS We conducted a non-systematic critical review to identify the characteristics of each SMA disease. RESULTS Many of the non-5q-SMA diseases have similar symptoms, making DNA analysis of patients essential for accurate diagnosis. Currently, genetic analysis technology using next-generation sequencers is rapidly advancing, opening up the possibility of elucidating the pathology and treating non-5q-SMA. CONCLUSION Based on accurate diagnosis and a deeper understanding of the pathology of each disease, treatments for non-5q-SMA diseases may be developed in the near future.
Collapse
Affiliation(s)
- Hisahide Nishio
- Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani-cho, Nishi-ku, Kobe 651-2180, Japan
| | - Emma Tabe Eko Niba
- Laboratory of Molecular and Biochemical Research, Biomedical Research Core Facilities, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan;
| | - Toshio Saito
- Department of Neurology, National Hospital Organization Osaka Toneyama Medical Center, 5-1-1 Toneyama, Toyonaka 560-8552, Japan;
| | - Kentaro Okamoto
- Department of Pediatrics, Ehime Prefectural Imabari Hospital, 4-5-5 Ishi-cho, Imabari 794-0006, Japan;
| | - Tomoko Lee
- Department of Pediatrics, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (T.L.); (Y.T.)
| | - Yasuhiro Takeshima
- Department of Pediatrics, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (T.L.); (Y.T.)
| | - Hiroyuki Awano
- Organization for Research Initiative and Promotion, Research Initiative Center, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan;
| | - Poh-San Lai
- Department of Pediatrics, National University of Singapore, 1E Lower Kent Ridge Road, Singapore 119228, Singapore;
| |
Collapse
|
31
|
Long J, Cui D, Yu C, Meng W. Evaluating the clinical efficacy of a long-read sequencing-based approach for carrier screening of spinal muscular atrophy. Hum Genomics 2024; 18:110. [PMID: 39343938 PMCID: PMC11440943 DOI: 10.1186/s40246-024-00676-8] [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: 07/10/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Spinal muscular atrophy (SMA) is the second most common fatal genetic disease in infancy. It is caused by deletion or intragenic pathogenic variants of the causative gene SMN1, which degenerates anterior horn motor neurons and leads to progressive myasthenia and muscle atrophy. Early treatment improves motor function and prognosis in patients with SMA, but drugs are expensive and do not cure the disease. Therefore, carrier screening seems to be the most effective way to prevent SMA birth defects. In this study, we genetically analyzed 1400 samples using multiplex ligation-dependent probe amplification (MLPA) and quantitative polymerase chain reaction (qPCR), and compared the consistency of the results. We randomly selected 44 samples with consistent MLPA and qPCR results for comprehensive SMA analysis (CASMA) using a long-read sequencing (LRS)-based approach. CASMA results showed 100% consistency, visually and intuitively explained the inconsistency between exons 7 and 8 copy numbers detected by MLPA in 13 samples. A total of 16 samples showed inconsistent MLPA and qPCR results for SMN1 exon 7. CASMA was performed on all samples and the results were consistent with those of resampling for MLPA and qPCR detection. CASMA also detected an additional intragenic variant c.-39A>G in a sample with two copies of SMN1 (RT02). Finally, we detected 23 SMA carriers, with an estimated carrier rate of 1/61 in this cohort. In addition, CASMA identified the "2 + 0" carrier status of SMN1 and SMN2 in a family by analyzing the genotypes of only three samples (parents and one sibling). CASMA has great advantages over MLPA and qPCR assays, and could become a powerful technical support for large-scale screening of SMA.
Collapse
Affiliation(s)
- Ju Long
- Laboratory of Medical Genetics, Qinzhou Maternal and Child Health Care Hospital, Qinzhou, Guangxi Province, 535099, China.
| | - Di Cui
- Berry Genomics Corporation, Beijing, 102200, China
| | - Chunhui Yu
- Laboratory of Medical Genetics, Qinzhou Maternal and Child Health Care Hospital, Qinzhou, Guangxi Province, 535099, China
| | - Wanli Meng
- Berry Genomics Corporation, Beijing, 102200, China
| |
Collapse
|
32
|
Yang H, Yang J, Xue Y, Liao L, Cai Q, Luo R. Cognitive impairment in children with 5q-associated spinal muscular atrophy type 1: two case reports and the review of the literature. Front Pediatr 2024; 12:1407341. [PMID: 39398417 PMCID: PMC11466754 DOI: 10.3389/fped.2024.1407341] [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: 03/26/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive disease caused by mutations in the survival motor neuron 1 (SMN1) gene on chromosome 5, leading to the degeneration of lower motor neurons. There are few studies on cognitive impairment comorbid with SMA. Here, we report two cases of severe cognitive impairment in Chinese children with SMA type 1, marking the first such reports in this demographic. We propose that severe cognitive dysfunction may be a comorbidity of SMA. Clinicians should consider SMA in patients presenting with severe muscle weakness and atrophy accompanied by cognitive impairments, to avoid misdiagnosis and oversight.
Collapse
Affiliation(s)
- Hua Yang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Jie Yang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Yawen Xue
- Department of Pediatric Neurology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lihui Liao
- Department of Pediatric Neurology Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qianyun Cai
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Rong Luo
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Guo D, Liu Z, Zhou J, Ke C, Li D. Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:9947. [PMID: 39337436 PMCID: PMC11432010 DOI: 10.3390/ijms25189947] [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: 07/27/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Programmed cell death (PCD) is a form of cell death distinct from accidental cell death (ACD) and is also referred to as regulated cell death (RCD). Typically, PCD signaling events are precisely regulated by various biomolecules in both spatial and temporal contexts to promote neuronal development, establish neural architecture, and shape the central nervous system (CNS), although the role of PCD extends beyond the CNS. Abnormalities in PCD signaling cascades contribute to the irreversible loss of neuronal cells and function, leading to the onset and progression of neurodegenerative diseases. In this review, we summarize the molecular processes and features of different modalities of PCD, including apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, and other novel forms of PCD, and their effects on the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), multiple sclerosis (MS), traumatic brain injury (TBI), and stroke. Additionally, we examine the key factors involved in these PCD signaling pathways and discuss the potential for their development as therapeutic targets and strategies. Therefore, therapeutic strategies targeting the inhibition or facilitation of PCD signaling pathways offer a promising approach for clinical applications in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Dong Guo
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Zhihao Liu
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Jinglin Zhou
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Chongrong Ke
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Daliang Li
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| |
Collapse
|
34
|
Desguerre I, Barrois R, Audic F, Barnerias C, Chabrol B, Davion JB, Durigneux J, Espil-Taris C, Gomez-Garcia de la Banda M, Guichard M, Isapof A, Nougues MC, Laugel V, Le Goff L, Mercier S, Pervillé A, Richelme C, Thibaud M, Sarret C, Schweitzer C, Testard H, Trommsdorff V, Vanhulle C, Walther-Louvier U, Altuzarra C, Chouchane M, Ropars J, Quijano-Roy S, Cances C. Real-world multidisciplinary outcomes of onasemnogene abeparvovec monotherapy in patients with spinal muscular atrophy type 1: experience of the French cohort in the first three years of treatment. Orphanet J Rare Dis 2024; 19:344. [PMID: 39272200 PMCID: PMC11401247 DOI: 10.1186/s13023-024-03326-3] [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: 05/09/2023] [Accepted: 08/18/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Spinal muscular atrophy type 1 (SMA1) is the most severe and early form of SMA, a genetic disease with motor neuron degeneration. Onasemnogene abeparvovec gene transfer therapy (GT) has changed the natural history of SMA1, but real-world data are scarce. METHODS A French national expert committee identified 95 newly diagnosed treatment-naive SMA1 patients between June 2019 and June 2022. We prospectively report on children treated with GT as the first and only therapy who had more than one-year of follow-up. RESULTS Forty-six SMA1 patients received GT. Twelve patients received other treatments. Patients with respiratory insufficiency were oriented toward palliative care after discussion with families. Twenty-nine of the treated patients with more than 12 months of follow-up were included in the follow-up analysis. Among them, 17 had 24 months of follow-up. The mean age at treatment was 7.5 (2.1-12.5) months. Twenty-two patients had two SMN2 copies, and seven had three copies. One infant died in the month following GT due to severe thrombotic microangiopathy, and another died due to respiratory distress. Among the 17 patients with 24 months of follow-up, 90% required spinal bracing (15/17), three patients required nocturnal noninvasive ventilation, and two needed gastrostomy. Concerning motor milestones at the 24-month follow-up, all patients held their head, 15/17 sat for 30 s unassisted, and 12/17 stood with aid. Motor scores (CHOPINTEND and HINE-2) and thoracic circumference significantly improved in all patients. CONCLUSIONS Our study shows favorable motor outcomes and preserved respiratory and feeding functions in treatment-naive SMA1 infants treated by GT as the first and only therapy before respiratory and bulbar dysfunctions occurred. Nevertheless, almost all patients developed spinal deformities.
Collapse
Affiliation(s)
- Isabelle Desguerre
- IHU Imagine, Paris University, 24, Boulevard du Montparnasse, 75015, Paris, France
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, AP-HP, Hôpital Necker-Enfants Malades, 149 rue de sèvres, 75015, Paris, France
| | - Rémi Barrois
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, AP-HP, Hôpital Necker-Enfants Malades, 149 rue de sèvres, 75015, Paris, France.
| | - Frédérique Audic
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, Hôpital Timone Enfants, 264 rue Saint-Pierre, 13385, Marseille, France
| | - Christine Barnerias
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, AP-HP, Hôpital Necker-Enfants Malades, 149 rue de sèvres, 75015, Paris, France
| | - Brigitte Chabrol
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, Hôpital Timone Enfants, 264 rue Saint-Pierre, 13385, Marseille, France
| | - Jean Baptiste Davion
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, Lille University Hospital Center, 2 avenue Oscar Lambret, 59000, Lille, France
| | - Julien Durigneux
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, Angers University Hospital Center, 4 rue Larrey, 49933, Angers, France
| | - Caroline Espil-Taris
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, Pellegrin University Hospital Center, Hôpital des Enfants, place Amélie-Raba-Léon, 33086, Bordeaux, France
| | - Marta Gomez-Garcia de la Banda
- Pediatric Neurology and ICU Department, Garches Reference Center for Neuromuscular Diseases (NEIF for FILNEMUS; RPC for Euro-NMD ERN), AP-HP Paris-Saclay Université, Hôpital Raymond Poincaré (UVSQ), 104 boulevard Raymond Poincaré, 92380, Garches, France
| | - Marine Guichard
- Department of Pediatric Neurology and Handicaps, French Competence Center for Neuromuscular Diseases, Boulevard Tonnellé, Hôpital Clocheville, 2 Boulevard Tonnellé, 37000, Tours, France
| | - Arnaud Isapof
- Department of Pediatric Neurology, AP-HP, French Reference Center for Neuromuscular Diseases, Hôpital Armand Trousseau, 26 avenue du Docteur Arnold-Netter, 75012, Paris, France
| | - Marie Christine Nougues
- Department of Pediatric Neurology, AP-HP, French Reference Center for Neuromuscular Diseases, Hôpital Armand Trousseau, 26 avenue du Docteur Arnold-Netter, 75012, Paris, France
| | - Vincent Laugel
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, Strasbourg University Hospital Center, Hôpital de Hautepierre, 1 avenue Molière, 67098, Strasbourg, France
| | - Laure Le Goff
- Department of Neuromuscular Pathology, French Reference Center for Neuromuscular Diseases, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, -Bron, 59 boulevard Pinel, 69677, Lyon-Bron, France
| | - Sandra Mercier
- Department of Medical Genetics, French Reference Center for Neuromuscular Diseases, Nantes University Hospital Center, 1 Place Alexis-Ricordeau, 44093, Nantes, France
| | - Anne Pervillé
- Department of Pediatrics, French Competence Center for Neuromuscular Diseases, Hôpital d'Enfants ASFA, CS 81010, 97404, Saint Denis Cedex, Réunion, France
| | - Christian Richelme
- Department of Pediatric Neurology, French Reference Center for Neuromuscular Diseases, Nice University Hospital Center, Hôpital Lenval, 57 Avenue de la Californie, 06200, Nice, France
| | - Marie Thibaud
- Department of Pediatrics, French Reference Center for Neuromuscular Diseases, American Memorial Hospital, Reims University Hospital Center, 49 Rue Cognacq Jay, 51092, Reims, France
| | - Catherine Sarret
- CMR Neuromusculaire, French Reference Center for Neuromuscular Diseases, Clermont-Ferrand University Hospital Center, Clermont-Ferrand, France
| | - Cyril Schweitzer
- Department of Infant Medicine, French Reference Center for Neuromuscular Diseases, Nancy University Hospital Center, Rue du Morvan, 54511, Vandoeuvre lès Nancy, France
| | - Hervé Testard
- Department of Pediatric Neurology, French Competence Center for Neuromuscular Diseases, Grenoble University Hospital Center, Hôpital Couple Enfant, Quai Yermolof, 38700, Grenoble, France
| | - Valérie Trommsdorff
- Department of Pediatrics, French Reference Center for Neuromuscular Diseases, University Hospital Center, Avenue François Mitterrand, BP 350, 97448, Saint Pierre Cedex, Réunion, France
| | - Catherine Vanhulle
- Department of Pediatrics, French Competence Center for Neuromuscular Diseases, Rouen University Hospital Center, Charles Nicolle, 1 Rue de Germont, 76031, Rouen, France
| | - Ulrike Walther-Louvier
- Department of Pediatric Neurology, French Greater South‒West Reference Center for Neuromuscular Diseases, Hôpital Gui de Chauliac, University Hospital Center Montpellier, 80 Avenue Augustin Fliche, 34295, Montpellier, France
| | - Cécilia Altuzarra
- Department of Pediatrics, French Reference Center for Neuromuscular Diseases, Besançon University Hospital Center - Hôpital Jean Minjoz, 3 boulevard A. Fleming, 25030, Besançon, France
| | - Mondher Chouchane
- Department of Pediatric Neurology, French Competence Center for Neuromuscular Diseases, Dijon University Hospital Center, Hôpital d'Enfants, 14 rue Paul Gaffarel, 21079, Dijon, France
| | - Juliette Ropars
- LaTIM INSERM UMR 1101, French Reference Center for Neuromuscular Diseases Brest University Hospital Center, Hôpital Morvan, Boulevard Tanguy Prigent, 29609, Brest, France
| | - Susana Quijano-Roy
- Pediatric Neurology and ICU Department, Garches Reference Center for Neuromuscular Diseases (NEIF for FILNEMUS; RPC for Euro-NMD ERN), AP-HP Paris-Saclay Université, Hôpital Raymond Poincaré (UVSQ), 104 boulevard Raymond Poincaré, 92380, Garches, France
| | - Claude Cances
- Department of Pediatric Neurology, French Greater South‒West Reference Center for Neuromuscular Diseases, Hôpital des Enfants, University Hospital Center Toulouse, 330 av de Grande Bretagne-TSA, 31059, Toulouse, France
| |
Collapse
|
35
|
Grandi FC, Astord S, Pezet S, Gidaja E, Mazzucchi S, Chapart M, Vasseur S, Mamchaoui K, Smeriglio P. Characterization of SMA type II skeletal muscle from treated patients shows OXPHOS deficiency and denervation. JCI Insight 2024; 9:e180992. [PMID: 39264856 PMCID: PMC11530132 DOI: 10.1172/jci.insight.180992] [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: 03/12/2024] [Accepted: 09/10/2024] [Indexed: 09/14/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a recessive developmental disorder caused by the genetic loss or mutation of the gene SMN1 (survival of motor neuron 1). SMA is characterized by neuromuscular symptoms and muscle weakness. Several years ago, SMA treatment underwent a radical transformation, with the approval of 3 different SMN-dependent disease-modifying therapies. This includes 2 SMN2 splicing therapies - risdiplam and nusinersen. One main challenge for type II SMA patients treated with these drugs is ongoing muscle fatigue, limited mobility, and other skeletal problems. To date, few molecular studies have been conducted on SMA patient-derived tissues after treatment, limiting our understanding of what targets remain unchanged after the spinal cord-targeted therapies are applied. Therefore, we collected paravertebral muscle from 8 type II patients undergoing spinal surgery for scoliosis and 7 controls. We used RNA-seq to characterize their transcriptional profiles and correlate these molecular changes with muscle histology. Despite the limited cohort size and heterogeneity, we observed a consistent loss of oxidative phosphorylation (OXPHOS) machinery of the mitochondria, a decrease in mitochondrial DNA copy number, and a correlation between signals of cellular stress, denervation, and increased fibrosis. This work provides new putative targets for combination therapies for type II SMA.
Collapse
Affiliation(s)
- Fiorella Carla Grandi
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Stéphanie Astord
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Sonia Pezet
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Elèna Gidaja
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Sabrina Mazzucchi
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Maud Chapart
- Centre de Ressources Biologiques - Myobank-AFM de l’Institut de Myologie, Hôpital de la Pitié-Salpêtrière F - 75013 Paris, France
| | - Stéphane Vasseur
- Centre de Ressources Biologiques - Myobank-AFM de l’Institut de Myologie, Hôpital de la Pitié-Salpêtrière F - 75013 Paris, France
| | - Kamel Mamchaoui
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Piera Smeriglio
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| |
Collapse
|
36
|
Panicucci C, Sahin E, Bartolucci M, Casalini S, Brolatti N, Pedemonte M, Baratto S, Pintus S, Principi E, D'Amico A, Pane M, Sframeli M, Messina S, Albamonte E, Sansone VA, Mercuri E, Bertini E, Sezerman U, Petretto A, Bruno C. Proteomics profiling and machine learning in nusinersen-treated patients with spinal muscular atrophy. Cell Mol Life Sci 2024; 81:393. [PMID: 39254732 PMCID: PMC11387582 DOI: 10.1007/s00018-024-05426-6] [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: 02/25/2024] [Revised: 08/11/2024] [Accepted: 08/25/2024] [Indexed: 09/11/2024]
Abstract
AIM The availability of disease-modifying therapies and newborn screening programs for spinal muscular atrophy (SMA) has generated an urgent need for reliable prognostic biomarkers to classify patients according to disease severity. We aim to identify cerebrospinal fluid (CSF) prognostic protein biomarkers in CSF samples of SMA patients collected at baseline (T0), and to describe proteomic profile changes and biological pathways influenced by nusinersen before the sixth nusinersen infusion (T302). METHODS In this multicenter retrospective longitudinal study, we employed an untargeted liquid chromatography mass spectrometry (LC-MS)-based proteomic approach on CSF samples collected from 61 SMA patients treated with nusinersen (SMA1 n=19, SMA2 n=19, SMA3 n=23) at T0 at T302. The Random Forest (RF) machine learning algorithm and pathway enrichment analysis were applied for analysis. RESULTS The RF algorithm, applied to the protein expression profile of naïve patients, revealed several proteins that could classify the different types of SMA according to their differential abundance at T0. Analysis of changes in proteomic profiles identified a total of 147 differentially expressed proteins after nusinersen treatment in SMA1, 135 in SMA2, and 289 in SMA3. Overall, nusinersen-induced changes on proteomic profile were consistent with i) common effects observed in allSMA types (i.e. regulation of axonogenesis), and ii) disease severity-specific changes, namely regulation of glucose metabolism in SMA1, of coagulation processes in SMA2, and of complement cascade in SMA3. CONCLUSIONS This untargeted LC-MS proteomic profiling in the CSF of SMA patients revealed differences in protein expression in naïve patients and showed nusinersen-related modulation in several biological processes after 10 months of treatment. Further confirmatory studies are needed to validate these results in larger number of patients and over abroader timeframe.
Collapse
Affiliation(s)
- Chiara Panicucci
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini, 5, I-16147, Genova, Italy
| | - Eray Sahin
- Department of Biostatistics and Bioinformatics, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Martina Bartolucci
- Core Facilities-Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Sara Casalini
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini, 5, I-16147, Genova, Italy
| | - Noemi Brolatti
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini, 5, I-16147, Genova, Italy
| | - Marina Pedemonte
- Pediatric Neurology Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Serena Baratto
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini, 5, I-16147, Genova, Italy
| | - Sara Pintus
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini, 5, I-16147, Genova, Italy
| | - Elisa Principi
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini, 5, I-16147, Genova, Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Marika Pane
- Centro Clinico Nemo, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Marina Sframeli
- Department of Neurosciences, University of Messina, Messina, Italy
| | - Sonia Messina
- Department of Neurosciences, University of Messina, Messina, Italy
| | - Emilio Albamonte
- Neurorehabilitation Unit, Centro Clinico NeMO, University of Milan, Milan, Italy
| | - Valeria A Sansone
- Neurorehabilitation Unit, Centro Clinico NeMO, University of Milan, Milan, Italy
| | - Eugenio Mercuri
- Centro Clinico Nemo, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Ugur Sezerman
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Andrea Petretto
- Core Facilities-Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Claudio Bruno
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini, 5, I-16147, Genova, Italy.
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health- DINOGMI, University of Genova, Genova, Italy.
| |
Collapse
|
37
|
Price TR, Hodgkinson V, Westbury G, Korngut L, Innes MA, Marshall CR, Nelson TN, Huang L, Parboosingh J, Mah JK. A Study on the Incidence and Prevalence of 5q Spinal Muscular Atrophy in Canada Using Multiple Data Sources. Can J Neurol Sci 2024; 51:660-671. [PMID: 38178730 DOI: 10.1017/cjn.2024.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
OBJECTIVES Spinal muscular atrophy (SMA) is a leading genetic cause of infant death and represents a significant burden of care. An improved understanding of the epidemiology of SMA in Canada may help inform strategies to improve the standard of care for individuals living with SMA. METHODS We employed a multisource approach to estimate the minimal incidence and prevalence of 5q SMA and to gain greater insight into recent clinical practices and treatment trends for the Canadian SMA population. Data sources included the Canadian Paediatric Surveillance Program (CPSP), Canadian Neuromuscular Disease Registry (CNDR), and molecular genetics laboratories in Canada. RESULTS The estimated annual minimum incidence of 5q SMA was 4.38, 3.44, and 7.99 cases per 100,000 live births in 2020 and 2021, based on CPSP, CNDR, and molecular genetics laboratories data, respectively, representing approximately 1 in 21,472 births (range 12,516-29,070) in Canada. SMA prevalence was estimated to be 0.85 per 100,000 persons aged 0-79 years. Delay in diagnosis exists across all SMA subtypes. Most common presenting symptoms were delayed milestones, hypotonia, and muscle weakness. Nusinersen was the most common disease-modifying treatment received. Most patients utilized multidisciplinary clinics for management of SMA. CONCLUSION This study provides data on the annual minimum incidence of pediatric 5q SMA in Canada. Recent therapeutic advances and newborn screening have the potential to drastically alter the natural history of SMA. Findings underline the importance of ongoing surveillance of the epidemiology and long-term health outcomes of SMA in the Canadian population.
Collapse
Affiliation(s)
- Tiffany R Price
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Victoria Hodgkinson
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Grace Westbury
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lawrence Korngut
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Micheil A Innes
- Departments of Pediatrics and Medical Genetics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Christian R Marshall
- Division of Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tanya N Nelson
- Division of Genome Diagnostics, Department of Pathology and Laboratory Medicine, BC Children's Hospital, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lijia Huang
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Jillian Parboosingh
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jean K Mah
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
38
|
Zhang W, Feng Y, Yan Y, Yao M, Gao F, Lin W, Mao S. Health information literacy among children with spinal muscular atrophy and their caregivers. Ital J Pediatr 2024; 50:157. [PMID: 39183350 PMCID: PMC11346139 DOI: 10.1186/s13052-024-01723-9] [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: 05/21/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disease that leads to multiple organ dysfunction. The advent of disease-modifying treatments makes the early diagnosis of SMA critical. Health information literacy is vital for obtaining, understanding, screening, and using health information. Considering the importance of early diagnosis and the challenges in obtaining accurate information on patients with SMA, this cross-sectional study assessed health information literacy among children with SMA and their caregivers in China. METHODS Interviews with the caregivers of 10 patients with SMA were conducted by neurologists specializing in SMA. A questionnaire for evaluating the level of health information literacy was further developed among 145 children with SMA aged 10.0-120.0 months, with the average age of 81.9 months, and their caregivers. Parameters, such as the age at the onset of the first symptom and time from recognition of the first symptom to diagnosis, were examined. Health information literacy was measured using four dimensions: cognition, search, evaluation, and application. RESULTS The average time from the first symptom to first medical consultation was 4.8 months, and that from the first symptom to diagnosis was 10.8 months. There is a significant delay from the onset of the initial symptoms to a definitive diagnosis. Thirty-five (24%) patients had poor while 26 (18%) had high health information literacy. The overall score for health information literacy was 69; the scores for health information cognition and application were 90 and 84, respectively. The scores for evaluation (61) and search (57) were low. Medical personnel were considered the most professional and credible sources of information. Additionally, search engines and patient organizations were the other two most important sources of health literacy. CONCLUSION Patients with SMA and their caregivers had low levels of health information literacy. SMA information visibility and standardization need to be improved. Medical personnel with experience in the diagnosis and treatment of SMA and media should aim to share knowledge and increase the quality of life of those with SMA.
Collapse
Affiliation(s)
- Weiran Zhang
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| | - Yijie Feng
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| | - Yue Yan
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| | - Mei Yao
- Department of Infection, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| | - Feng Gao
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China
| | - Wei Lin
- Film and New Media Studies, College of Media and International Culture, Institute of Leisure Studies and Philosophy of Art, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
| | - Shanshan Mao
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, China.
| |
Collapse
|
39
|
Grass T, Dokuzluoglu Z, Buchner F, Rosignol I, Thomas J, Caldarelli A, Dalinskaya A, Becker J, Rost F, Marass M, Wirth B, Beyer M, Bonaguro L, Rodriguez-Muela N. Isogenic patient-derived organoids reveal early neurodevelopmental defects in spinal muscular atrophy initiation. Cell Rep Med 2024; 5:101659. [PMID: 39067446 PMCID: PMC11384962 DOI: 10.1016/j.xcrm.2024.101659] [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: 07/25/2023] [Revised: 02/26/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024]
Abstract
Whether neurodevelopmental defects underlie postnatal neuronal death in neurodegeneration is an intriguing hypothesis only recently explored. Here, we focus on spinal muscular atrophy (SMA), a neuromuscular disorder caused by reduced survival of motor neuron (SMN) protein levels leading to spinal motor neuron (MN) loss and muscle wasting. Using the first isogenic patient-derived induced pluripotent stem cell (iPSC) model and a spinal cord organoid (SCO) system, we show that SMA SCOs exhibit abnormal morphological development, reduced expression of early neural progenitor markers, and accelerated expression of MN progenitor and MN markers. Longitudinal single-cell RNA sequencing reveals marked defects in neural stem cell specification and fewer MNs, favoring mesodermal progenitors and muscle cells, a bias also seen in early SMA mouse embryos. Surprisingly, SMN2-to-SMN1 conversion does not fully reverse these developmental abnormalities. These suggest that early neurodevelopmental defects may underlie later MN degeneration, indicating that postnatal SMN-increasing interventions might not completely amend SMA pathology in all patients.
Collapse
Affiliation(s)
- Tobias Grass
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany.
| | - Zeynep Dokuzluoglu
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Felix Buchner
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Ines Rosignol
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany; Technische Universität Dresden (TUD), Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Joshua Thomas
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Antonio Caldarelli
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Anna Dalinskaya
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Jutta Becker
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany
| | - Fabian Rost
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering, TUD, Dresden, Germany
| | - Michele Marass
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Center for Rare Diseases, University Hospital of Cologne, Cologne, Germany
| | - Marc Beyer
- Systems Medicine, DZNE, Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE & University of Bonn and West German Genome Center, Bonn, Germany; Immunogenomics & Neurodegeneration, DZNE, Bonn, Germany
| | - Lorenzo Bonaguro
- Systems Medicine, DZNE, Bonn, Germany; Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Natalia Rodriguez-Muela
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany; Technische Universität Dresden (TUD), Center for Regenerative Therapies Dresden, Dresden, Germany; Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
40
|
Brkušanin M, Garai N, Karanović J, Šljivančanin Jakovljević T, Dimitrijević A, Jovanović K, Mitrović TL, Miković Ž, Brajušković G, Nikolić DM, Savić-Pavićević D. Our Journey from Individual Efforts to Nationwide Support: Implementing Newborn Screening for Spinal Muscular Atrophy in Serbia. Int J Neonatal Screen 2024; 10:57. [PMID: 39189229 PMCID: PMC11348157 DOI: 10.3390/ijns10030057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Abstract
Innovative treatments for spinal muscular atrophy (SMA) yield the utmost advantages only within the presymptomatic phase, underlining the significance of newborn screening (NBS). We aimed to establish statewide NBS for SMA in Serbia. Our stepwise implementation process involved technical validation of a screening assay, collaboration with patient organizations and medical professionals, a feasibility study, and negotiation with public health representatives. Over 12,000 newborns were tested during the 17-month feasibility study, revealing two unrelated SMA infants and one older sibling. All three children received therapeutic interventions during the presymptomatic phase and have shown no signs of SMA. No false-negative results were found among the negative test results. As frontrunners in this field in Serbia, we established screening and diagnostic algorithms and follow-up protocols and raised awareness among stakeholders about the importance of early disease detection, leading to the incorporation of NBS for SMA into the national program on 15 September 2023. Since then, 54,393 newborns have been tested, identifying six SMA cases and enabling timely treatment. Our study demonstrates that effective collaborations between academia, non-profit organizations, and industry are crucial in bringing innovative healthcare initiatives to fruition, and highlights the potential of NBS to revolutionize healthcare outcomes for presymptomatic SMA infants and their families.
Collapse
Affiliation(s)
- Miloš Brkušanin
- Centre for Human Molecular Genetics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (N.G.); (J.K.); (G.B.)
| | - Nemanja Garai
- Centre for Human Molecular Genetics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (N.G.); (J.K.); (G.B.)
| | - Jelena Karanović
- Centre for Human Molecular Genetics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (N.G.); (J.K.); (G.B.)
| | | | - Aleksandra Dimitrijević
- Department of Gynecology and Obstetrics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
- Gynaecology and Obstetrics Clinic, Clinical Centre of Kragujevac, 34000 Kragujevac, Serbia
| | - Kristina Jovanović
- Neurology Department, University Children’s Hospital, 11000 Belgrade, Serbia; (K.J.); (D.M.N.)
| | - Tanja Lazić Mitrović
- Department of Neonatology, Obstetrics and Gynaecology Clinic Narodni Front, 11000 Belgrade, Serbia; (T.Š.J.); (T.L.M.)
| | - Željko Miković
- High-Risk Pregnancy Unit, Obstetrics and Gynaecology Clinic Narodni Front, 11000 Belgrade, Serbia;
| | - Goran Brajušković
- Centre for Human Molecular Genetics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (N.G.); (J.K.); (G.B.)
| | - Dimitrije Mihailo Nikolić
- Neurology Department, University Children’s Hospital, 11000 Belgrade, Serbia; (K.J.); (D.M.N.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Dušanka Savić-Pavićević
- Centre for Human Molecular Genetics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia; (N.G.); (J.K.); (G.B.)
| |
Collapse
|
41
|
McPheron MA, Felker MV. Clinical perspectives: Treating spinal muscular atrophy. Mol Ther 2024; 32:2489-2504. [PMID: 38894541 PMCID: PMC11405177 DOI: 10.1016/j.ymthe.2024.06.020] [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: 12/05/2023] [Revised: 04/26/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024] Open
Abstract
Spinal muscular atrophy is a rare and progressive neuromuscular disease that, without treatment, leads to progressive weakness and often death. A plethora of studies have led to the approval of three high-cost and effective treatments since 2016. These treatments, nusinersen, onasemnogene abeparvovec, and risdiplam, have not been directly compared and have varying challenges in administration. In this review, we discuss the evidence supporting the use of these medications, the process of treatment selection, monitoring after treatment, the limited data comparing treatments, as well as future directions for investigation and therapy.
Collapse
Affiliation(s)
- Molly A McPheron
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
| | - Marcia V Felker
- Department of Neurology, Indiana University, Indianapolis, IN 46202, USA.
| |
Collapse
|
42
|
Schroth M, Deans J, Arya K, Castro D, De Vivo DC, Gibbons MA, Ionita C, Kuntz NL, Lakhotia A, Neil Knierbein E, Scoto M, Sejersen T, Servais L, Tian C, Waldrop MA, Vázquez-Costa JF. Spinal Muscular Atrophy Update in Best Practices: Recommendations for Diagnosis Considerations. Neurol Clin Pract 2024; 14:e200310. [PMID: 38915908 PMCID: PMC11195435 DOI: 10.1212/cpj.0000000000200310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 02/21/2024] [Indexed: 06/26/2024]
Abstract
Background and Objectives Spinal muscular atrophy (SMA) is an autosomal recessive progressive neurodegenerative primary motor neuron disorder caused by biallelic variants of the survival motor neuron 1 (SMN1) gene. The most recent SMA best practice recommendations were published in 2018 shortly after the approval of the first SMN-enhancing treatment. The availability of disease-modifying therapies for 5q SMA and implementation of SMA newborn screening (NBS) has led to urgency to update the SMA best practice recommendations for diagnosis and to reevaluate the current classification of SMA. In addition, the availability of disease-modifying therapies has opened the door to explore improved diagnosis of adult-onset SMA. Methods A systematic literature review was conducted on SMA NBS. An SMA working group of American and European health care providers developed recommendations through a modified Delphi technique with serial surveys and virtual meeting feedback on SMA diagnosis to fill information gaps for topics with limited evidence. A community working group of an individual with SMA and caregivers provided insight and perspective on SMA diagnosis and support through a virtual meeting to guide recommendations. Results The health care provider working group achieved consensus that SMA NBS is essential to include in the updated best practice for SMA diagnosis (100%). Recommendations for the following are described: characterizing NBS-identified infants before treatment; minimum recommendations for starting or offering SMA NBS in a state or country; recommendations for activities and services to be provided by an SMA specialty care center accepting SMA NBS referrals; and recommendations for partnership with individuals with SMA and caregivers to support NBS-identified infants and their caregivers. Limited data are available to advance efficient diagnosis of adult-onset SMA. Discussion Updating best practice recommendations for SMA diagnosis to include SMA NBS implementation is essential to advancing care for individuals with SMA. In addition to testing, processes for the efficient management of positive newborn screen with access to knowledgeable and skilled health care providers and access to treatment options is critical to successful early diagnosis. Additional evidence is required to improve adult-onset SMA diagnosis.
Collapse
Affiliation(s)
- Mary Schroth
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Jennifer Deans
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Kapil Arya
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Diana Castro
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Darryl C De Vivo
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Melissa A Gibbons
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Cristian Ionita
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Nancy L Kuntz
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Arpita Lakhotia
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Erin Neil Knierbein
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Mariacristina Scoto
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Thomas Sejersen
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Laurent Servais
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Cuixia Tian
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Megan A Waldrop
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| | - Juan F Vázquez-Costa
- Cure SMA (M. Schroth, JD), Elk Grove Village, IL; Department of Pediatrics (KA), Division of Neurology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock; Neurology and Neuromuscular Care Center (DC), Denton, TX; Departments of Neurology and Pediatrics (DCDV), Columbia University Irving Medical Center, New York; Department of Pediatrics (MAG), University of Colorado School of Medicine, Aurora; Department of Pediatrics (Neurology) (CI), Yale University School of Medicine, New Haven, CT; Department of Pediatrics and Neurology (NLK), Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern Feinberg School of Medicine, IL; Department of Neurology (AL), University of Louisville, Norton Children's Medical Group, KY; Department of Pediatrics (ENK), University of Michigan Health, Ann Arbor; The Dubowitz Neuromuscular Centre (M. Scoto), Great Ormond Street Hospital Trust, London, UK & Great Ormond Street Institute of Child Health, University College London, United Kingdom; Department of Women's and Children's Health (TS), Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden, and Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong; MDUK Oxford Neuromuscular Center & NIHR Oxford Biomedical Research Centre (LS), University of Oxford, United Kingdom, and Neuromuscular Center, Department of Paediatrics, University of Liege and University Hospital of Liege, Belgium; Division of Neurology (CT), Cincinnati Children's Hospital Medical Center & Department of Pediatrics, University of Cincinnati Medical College, OH; Center for Gene Therapy (MAW), The Abigail Wexner Research Institute, Nationwide Children's Hospital, Departments of Pediatric and Neurology, The Ohio State University Wexner Medical Center, Columbus; and Motor Neuron Disease Unit (JFV-C), Hospital la Fe, IIS La Fe, CIBERER, University of Valencia, Spain
| |
Collapse
|
43
|
Cook SL, Stout C, Kirkeby L, Vidal-Folch N, Oglesbee D, Hasadsri L, Selcen D, Milone M, Anderson D, Staff NP. SMN1 c.5C>G (p.Ala2Gly) missense variant, a challenging molecular SMA diagnosis associated with mild disease, preserves SMN nuclear gems in patient-specific fibroblasts. Front Genet 2024; 15:1406819. [PMID: 39139818 PMCID: PMC11319185 DOI: 10.3389/fgene.2024.1406819] [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: 03/26/2024] [Accepted: 06/25/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction Spinal muscular atrophy (SMA) is caused by homozygous loss of the SMN1 gene with SMN2 gene copy number correlating with disease severity. Rarely SMA is caused by a deletion on one allele and a pathogenic variant on the other. The pathogenic missense variant c.5C>G (p.Ala2Gly) correlates with a mild disease phenotype that does not correlate with SMN2 copy number. In a mouse model the c.5C>G transgene produces SMN that is thought to form partially functional SMN complexes, but levels in humans have not yet been investigated. Methods We identified two patients with mild SMA caused by a heterozygous deletion of SMN1 and the heterozygous variant, c.5C>G. Molecular findings were confirmed with deletion/duplication analysis and Sanger sequencing. Skin fibroblasts were collected and cultured, and SMN expression was analyzed using immunofluorescence. Results Two patients with slowly progressing mild weakness were confirmed to have heterozygous pathogenic missense variant c.5C>G and a heterozygous deletion of SMN1. Their clinical presentation revealed much milder disease progression than patients with matched SMN2 copy number. Analysis of the patients' fibroblasts revealed much higher numbers of SMN nuclear complexes than a patient with a homozygous SMN1 deletion and matched SMN2 copy number. Conclusions These case reports reinforce that the rare c.5C>G variant causes mild disease. Furthermore, the analysis of SMA nuclear gems in patient samples supports the theory that the p.Ala2Gly SMN can form partially functional SMN complexes that may carry out essential cellular functions and result in mild disease.
Collapse
Affiliation(s)
- Sara L. Cook
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Christian Stout
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Lindsey Kirkeby
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Noemi Vidal-Folch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Devin Oglesbee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Linda Hasadsri
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Duygu Selcen
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | | | - Daniel Anderson
- Department of Neurology, Mayo Clinic Health System, La Crosse, WI, United States
| | - Nathan P. Staff
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
44
|
AlTawari A, Zakaria M, Kamel W, Shaalan N, Elghazawi GAI, Ali MEA, Salota D, Attia A, Elanay EEA, Shalaby O, Alqallaf F, Mitic V, Bastaki L. Nusinersen Treatment for Spinal Muscular Atrophy: Retrospective Multicenter Study of Pediatric and Adult Patients in Kuwait. Neurol Int 2024; 16:631-642. [PMID: 38921951 PMCID: PMC11206794 DOI: 10.3390/neurolint16030047] [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: 04/16/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
Spinal muscular atrophy is a neuromuscular genetic condition associated with progressive muscle weakness and atrophy. Nusinersen is an antisense oligonucleotide therapy approved for the treatment of 5q spinal muscular atrophy in pediatric and adult patients. The objective of this clinical case series is to describe the efficacy and safety of nusinersen in treating spinal muscular atrophy in 20 pediatric and 18 adult patients across six treatment centers in Kuwait. Functional motor assessments (Children's Hospital of Philadelphia Infant Test of Neuromuscular Disorders, Hammersmith Functional Motor Scale Expanded, and Revised Upper Limb Module) were used to assess changes in motor function following nusinersen treatment. The safety assessment involved clinical monitoring of adverse events. The results demonstrate clinically meaningful or considerable improvement in motor performance for nearly all patients, lasting over 4 years in some cases. A total of 70% of patients in the pediatric cohort and 72% of patients in the adult cohort achieved a clinically meaningful improvement in motor function following nusinersen treatment. Additionally, nusinersen was well-tolerated in both cohorts. These findings add to the growing body of evidence relating to the clinical efficacy and safety of nusinersen.
Collapse
Affiliation(s)
- Asma AlTawari
- Pediatric Department, Neurology Unit, Al Sabah Hospital, Shuwaikh Industrial 70050, Kuwait
| | | | - Walaa Kamel
- Neurology Department, Ibn Sina Hospital, Shuwaikh Industrial 70050, Kuwait
| | - Nayera Shaalan
- Neurology Department, Ibn Sina Hospital, Shuwaikh Industrial 70050, Kuwait
| | | | | | - Dalia Salota
- Pediatric Department, Neurology Unit, Al Sabah Hospital, Shuwaikh Industrial 70050, Kuwait
| | - Amr Attia
- Pediatric Department, Neurology Unit, Al Sabah Hospital, Shuwaikh Industrial 70050, Kuwait
| | | | - Osama Shalaby
- Pediatric Department, Al Jahra Hospital, Al Jahra 003200, Kuwait
| | - Fatema Alqallaf
- Pediatric Department, Neurology Unit, Mubarak Hospital, Jabriya 46300, Kuwait
| | - Vesna Mitic
- Pediatric Department, Al Farwaniya Hospital, Al Farwaniya 85000, Kuwait
| | - Laila Bastaki
- Kuwait Medical Genetics Center, Shuwaikh Industrial 70050, Kuwait
| |
Collapse
|
45
|
Kahraman A, Mutlu A, Livanelioğlu A. General movements in spinal muscular atrophy type 1. Physiother Theory Pract 2024; 40:1249-1255. [PMID: 36611288 DOI: 10.1080/09593985.2023.2164842] [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: 12/09/2021] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023]
Abstract
PURPOSE To investigate the motor repertoire of infants diagnosed with spinal muscular atrophy Type I (SMA Type I) without administration of any disease-modifying agent. METHODS Motor Optimality Score-Revised (MOS-R) was calculated from videos recorded between post-term weeks 9-17 for 22 infants with SMA Type I. The MOS-R of infants with SMA Type I was compared with those of 22 infants with cerebral palsy (CP) and 22 infants with typical development. RESULTS Of the infants with SMA Type I, 17 had absent fidgety movements (FMs) and 5 had sporadic FMs. Age adequate movement repertoire was absent, and the variety of movements in infants was very low. Movements were symmetrical but movements of four limbs remained on the surface level. Antigravity movements were very rare. Movement characterization was monotonous, slow speed, and small amplitude. The MOS-R of infants with SMA Type I was lower than those of infants with typical development but similar to those of infants with CP. CONCLUSIONS Infants with SMA Type I had a motor repertoire similar to infants with CP, while they had a poorer motor repertoire than infants with typical development in the fidgety period as evidenced by MOS-R. Central nervous system involvement in these infants with SMA Type I with absent FMs and reduced MOS-R is unknown. Further studies are needed to determine the role of problems in the afferent and efferent pathways of spinal cord and muscle atrophy in the observation of normal FMs.
Collapse
Affiliation(s)
- Aysu Kahraman
- Developmental and Early Physiotherapy Unit, Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Samanpazarı, Turkey
| | - Akmer Mutlu
- Developmental and Early Physiotherapy Unit, Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Samanpazarı, Turkey
| | - Ayşe Livanelioğlu
- Developmental and Early Physiotherapy Unit, Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Samanpazarı, Turkey
| |
Collapse
|
46
|
Brkušanin M, Kosać A, Branković-Srećković V, Jovanović K, Perić S, Karanović J, Matijašević Joković S, Garai N, Pešović J, Nikolić D, Stević Z, Brajušković G, Milić-Rašić V, Savić-Pavićević D. Phosphorylated neurofilament heavy chain in cerebrospinal fluid and plasma as a Nusinersen treatment response marker in childhood-onset SMA individuals from Serbia. Front Neurol 2024; 15:1394001. [PMID: 38756215 PMCID: PMC11097956 DOI: 10.3389/fneur.2024.1394001] [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: 02/29/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction Biomarkers capable of reflecting disease onset and short- and long-term therapeutic effects in individuals with spinal muscular atrophy (SMA) are still an unmet need and phosphorylated neurofilament heavy chain (pNF-H) holds significant promise. Methods We conducted a longitudinal prospective study to evaluate pNF-H levels in the cerebrospinal fluid (CSF) and plasma of 29 individuals with childhood-onset SMA treated with Nuinersen (SMA type 1: n = 6, 2: n = 17, 3: n = 6). pNF-H levels before and during treatment were compared with the levels of controls (n = 22), patients with Duchenne muscular dystrophy (n = 17), myotonic dystrophy type 1 (n = 11), untreated SMA individuals with chronic type 3 disease (n = 8), and children with presymptomatic SMA (n = 3). Results SMA type 1 showed the highest mean CSF pNF-H levels before treatment initiation. All Nusinersen-treated individuals (types 1, 2, and 3) showed significantly elevated mean baseline CSF pNF-H compared to controls, which inversely correlated with age at disease onset, age at first dose, disease duration and the initial CHOP INTEND result (SMA type 1 and 2). During 22 months of treatment, CSF pNF-H levels declined during loading doses, stabilizing at reduced levels from the initial maintenance dose in all individuals. Baseline plasma pNF-H levels in type 1 and 2 SMA were significantly increased compared to other cohorts and decreased notably in type 1 after 2 months of treatment and type 2 after 14 months. Conversely, SMA type 3, characterized by lower baseline pNF-H levels, did not show significant fluctuations in plasma pNF-H levels after 14 months of treatment. Conclusion Our findings suggest that CSF pNF-H levels in untreated SMA individuals are significantly higher than in controls and that monitoring of CSF pNF-H levels may serve as an indicator of rapid short-term treatment response in childhood-onset SMA individuals, irrespective of the subtype of the disease, while also suggesting its potential for assessing long-term suppression of neurodegeneration. Plasma pNF-H may serve as an appropriate outcome measure for disease progression and/or response to treatment in types 1 and 2 but not in type 3. Presymptomatic infants with SMA may show elevated pNF-H levels, confirming early neuronal degeneration.
Collapse
Affiliation(s)
- Miloš Brkušanin
- Faculty of Biology, Centre for Human Molecular Genetics, University of Belgrade, Belgrade, Serbia
| | - Ana Kosać
- Clinic for Neurology and Psychiatry for Children and Youth, Belgrade, Serbia
| | | | - Kristina Jovanović
- University Children's Hospital Tirsova, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Stojan Perić
- Neurology Clinic, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Jelena Karanović
- Faculty of Biology, Centre for Human Molecular Genetics, University of Belgrade, Belgrade, Serbia
| | | | - Nemanja Garai
- Faculty of Biology, Centre for Human Molecular Genetics, University of Belgrade, Belgrade, Serbia
| | - Jovan Pešović
- Faculty of Biology, Centre for Human Molecular Genetics, University of Belgrade, Belgrade, Serbia
| | - Dimitrije Nikolić
- University Children's Hospital Tirsova, University Clinical Centre of Serbia, Belgrade, Serbia
- School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Zorica Stević
- Neurology Clinic, University Clinical Centre of Serbia, Belgrade, Serbia
- School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Goran Brajušković
- Faculty of Biology, Centre for Human Molecular Genetics, University of Belgrade, Belgrade, Serbia
| | - Vedrana Milić-Rašić
- Clinic for Neurology and Psychiatry for Children and Youth, Belgrade, Serbia
- School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dušanka Savić-Pavićević
- Faculty of Biology, Centre for Human Molecular Genetics, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
47
|
Wang N, Jiao K, He J, Zhu B, Cheng N, Sun J, Chen L, Chen W, Gong L, Qiao K, Xi J, Wu Q, Zhao C, Zhu W. Diagnosis of Challenging Spinal Muscular Atrophy Cases with Long-Read Sequencing. J Mol Diagn 2024; 26:364-373. [PMID: 38490302 DOI: 10.1016/j.jmoldx.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/17/2024] [Accepted: 02/07/2024] [Indexed: 03/17/2024] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder primarily caused by the deletion or mutation of the survival motor neuron 1 (SMN1) gene. This study assesses the diagnostic potential of long-read sequencing (LRS) in three patients with SMA. For Patient 1, who has a heterozygous SMN1 deletion, LRS unveiled a missense mutation in SMN1 exon 5. In Patient 2, an Alu/Alu-mediated rearrangement covering the SMN1 promoter and exon 1 was identified through a blend of multiplex ligation-dependent probe amplification, LRS, and PCR across the breakpoint. The third patient, born to a consanguineous family, bore four copies of hybrid SMN genes. LRS determined the genomic structures, indicating two distinct hybrids of SMN2 exon 7 and SMN1 exon 8. However, a discrepancy was found between the SMN1/SMN2 ratio interpretations by LRS (0:2) and multiplex ligation-dependent probe amplification (0:4), which suggested a limitation of LRS in SMA diagnosis. In conclusion, this newly adapted long PCR-based third-generation sequencing introduces an additional avenue for SMA diagnosis.
Collapse
Affiliation(s)
- Ningning Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kexin Jiao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin He
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Bochen Zhu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Nachuan Cheng
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Sun
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Chen
- Department of Neurology, Nantong First People's Hospital, Nantong, China
| | - Wanjin Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Lingyun Gong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kai Qiao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianying Xi
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qihan Wu
- Shanghai Ministry of Science and Technology Key Laboratory of Health and Disease Genomics, National Health Commission Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenhua Zhu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Shanghai, China; Huashan Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
48
|
Shekhawat DS, Didel S, Dixit SG, Singh P, Singh K. Carrier Screening and Diagnosis for Spinal Muscular Atrophy Using Droplet Digital PCR Versus MLPA: Analytical Validation and Early Test Outcome. Genet Test Mol Biomarkers 2024; 28:207-212. [PMID: 38533877 DOI: 10.1089/gtmb.2023.0073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024] Open
Abstract
Background: Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular life-threatening disorder. Owing to high carrier frequency, population-wide SMA screening to quantify the copy number of SMN gene is recommended by American College of Medical Genetics and Genomics. An accurate, reliable, short runaround time and cost-effective method may be helpful in mass population screening for SMA. Methods: Multiplex ligation-dependent probe amplification (MLPA) is a gold standard to estimate the copy number variation (CNV) for SMN1 and SMN2 genes. In this study, we validated droplet digital polymerase chain reaction (ddPCR) for the determination of CNV for both SMN1 and SMN2 exon 7 for a diagnostic purpose. In total, 66 clinical samples were tested using ddPCR, and results were compared with the MLPA as a reference test. Results: For all samples, CNV for SMN1 and SMN2 exon 7 was consentaneous between ddPCR and MLPA test results (κ = 1.000, p < 0.0001). In addition, ddPCR also showed a significant acceptable degree of test repeatability, coefficient of variation < 4%. Conclusion: ddPCR is expected to be utilitarian for CNV detection for carrier screening and diagnosis of SMA. ddPCR test results for CNV detection for SMN1/SMN2 exon 7 are concordant with the gold standard. ddPCR is a more cost-effective and time-saving diagnostic test for SMA than MLPA. Furthermore, it can be used for population-wide carrier screening for SMA.
Collapse
Affiliation(s)
- Dolat Singh Shekhawat
- Department of Pediatrics, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
- Department of Medical Genetics, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
- NIDAN Kendra, Genetic Laboratory, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
| | - Siyaram Didel
- Department of Pediatrics, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
| | - Shilpi Gupta Dixit
- NIDAN Kendra, Genetic Laboratory, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
- Department of Anatomy, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
| | - Pratibha Singh
- NIDAN Kendra, Genetic Laboratory, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
- Department of Obstetrics & Gynaecology, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
| | - Kuldeep Singh
- Department of Pediatrics, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
- Department of Medical Genetics, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
- NIDAN Kendra, Genetic Laboratory, All India Institute of Medical Science Jodhpur, Jodhpur, Rajasthan, India
| |
Collapse
|
49
|
Pozzobon M, Bean C. Mitochondria replacement from transplanted amniotic fluid stem cells: a promising therapy for non-neuronal defects in spinal muscular atrophy. Neural Regen Res 2024; 19:971-972. [PMID: 37862193 PMCID: PMC10749600 DOI: 10.4103/1673-5374.385304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/20/2023] [Accepted: 08/01/2023] [Indexed: 10/22/2023] Open
Affiliation(s)
- Michela Pozzobon
- Women’s and Children’s Health Department, University of Padova; Foundation Institute of Pediatric Research Città della Speranza, Padova, Italy
| | - Camilla Bean
- Women’s and Children’s Health Department, University of Padova; Foundation Institute of Pediatric Research Città della Speranza, Padova, Italy
| |
Collapse
|
50
|
Harding ER, Kanner CH, Pasternak A, Glanzman AM, Dunaway Young S, Rao AK, McDermott MP, Zolkipli-Cunningham Z, Day JW, Finkel RS, Darras BT, De Vivo DC, Montes J. Beyond Contractures in Spinal Muscular Atrophy: Identifying Lower-Limb Joint Hypermobility. J Clin Med 2024; 13:2634. [PMID: 38731167 PMCID: PMC11084694 DOI: 10.3390/jcm13092634] [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: 03/06/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Background: The natural history of spinal muscular atrophy (SMA) is well understood, with progressive muscle weakness resulting in declines in function. The development of contractures is common and negatively impacts function. Clinically, joint hypermobility (JH) is observed but is poorly described, and its relationship with function is unknown. Methods: Lower-limb ROM (range of motion) assessments of extension and flexion at the hip, knee, and ankle were performed. ROMs exceeding the published norms were included in the analysis. The functional assessments performed included the six-minute walk test (6 MWT) and the Hammersmith Functional Motor Scale-Expanded (HFMSE). Results: Of the 143 participants, 86% (n = 123) had at least one ROM measure that was hypermobile, and 22% (n = 32) had three or more. The HFMSE scores were inversely correlated with hip extension JH (r = -0.60, p = 0.21; n = 6) and positively correlated with knee flexion JH (r = 0.24, p = 0.02, n = 89). There was a moderate, inverse relationship between the 6 MWT distance and ankle plantar flexion JH (r = -0.73, p = 0.002; n = 15). Conclusions: JH was identified in nearly all participants in at least one joint in this study. Hip extension, knee flexion and ankle plantar flexion JH was associated with function. A further understanding of the trajectory of lower-limb joint ROM is needed to improve future rehabilitation strategies.
Collapse
Affiliation(s)
- Elizabeth R. Harding
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA (A.K.R.); (J.M.)
| | - Cara H. Kanner
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA (A.K.R.); (J.M.)
| | - Amy Pasternak
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.P.); (B.T.D.)
- Department of Physical and Occupational Therapy Services, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Allan M. Glanzman
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Sally Dunaway Young
- Department of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA 94304, USA; (S.D.Y.); (J.W.D.)
| | - Ashwini K. Rao
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA (A.K.R.); (J.M.)
| | - Michael P. McDermott
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY 14642, USA;
| | | | - John W. Day
- Department of Neurology and Clinical Neurosciences, Stanford University, Palo Alto, CA 94304, USA; (S.D.Y.); (J.W.D.)
| | - Richard S. Finkel
- Center for Experimental Neurotherapeutics, Department of Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Basil T. Darras
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.P.); (B.T.D.)
| | - Darryl C. De Vivo
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA;
| | - Jacqueline Montes
- Department of Rehabilitation and Regenerative Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA (A.K.R.); (J.M.)
| |
Collapse
|