1
|
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. [PMID: 39370660 DOI: 10.1002/mus.28267] [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: 03/21/2024] [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, Missouri, USA
| | - Amanda Rogers
- Department of Pediatrics, Norton Children's Medical Group, University of Louisville, Louisville, Kentucky, USA
| | - Mark Fisher
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Kapil Arya
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, Arkansas, USA
| | - John F Brandsema
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hiba Farah
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Michelle A Farrar
- Sydney Children's Hospital Network and Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Marcia V Felker
- Division of Pediatric Neurology, Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Melissa Gibbons
- Department of Pediatrics Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Omer Abdul Hamid
- Department of Pediatrics, Nemours Children's Health, Orlando, Florida, USA
| | - Matthew Harmelink
- Department of Neurology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Karen Herbert
- Sydney Children's Hospital Network and Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Elizabeth Kichula
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kiana King
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Arpita Lakhotia
- Department of Pediatrics, Norton Children's Medical Group, University of Louisville, Louisville, Kentucky, USA
| | - Bo Hoon Lee
- Department of Neurology, University of Rochester, Rochester, New York, USA
| | - Nancy L Kuntz
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Julie Parsons
- Department of Pediatrics Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Rebecca Rehborg
- Department of Neurology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Aravindhan Veerapaniyan
- Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, Arkansas, USA
| | - Craig M Zaidman
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Miletić M, Stević Z, Vujović S, Rakočević J, Tomić A, Tančić Gajić M, Stojanović M, Palibrk A, Žarković M. Glucose and Lipid Metabolism Disorders in Adults with Spinal Muscular Atrophy Type 3. Diagnostics (Basel) 2024; 14:2078. [PMID: 39335757 PMCID: PMC11431033 DOI: 10.3390/diagnostics14182078] [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: 07/11/2024] [Revised: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Spinal muscular atrophy type 3 (juvenile SMA, Kugelberg-Welander disease) is a genetic disease caused by changes in the survival motor neuron 1 (SMN) gene. However, there is increasing evidence of metabolic abnormalities in SMA patients, such as altered fatty acid metabolism, impaired glucose tolerance, and defects in the functioning of muscle mitochondria. Given that data in the literature are scarce regarding this subject, the purpose of this study was to estimate the prevalence of glucose and lipid metabolism disorders in adult patients with SMA type 3. METHODS We conducted a cross-sectional study of 23 adult patients with SMA type 3 who underwent a comprehensive evaluation, including a physical examination, biochemical analysis, and an oral glucose tolerance test during 2020-2023. RESULTS At least one lipid abnormality was observed in 60.8% of patients. All four lipid parameters were atypical in 4.3% of patients, three lipid parameters were abnormal in 21.7% of patients, and two lipid parameters were altered in 8.7% patients. A total of 91.3% of SMA3 patients met the HOMA-IR criteria for insulin resistance, with 30.43% having impaired glucose tolerance. None of the patients met the criteria for a diagnosis of overt DM2. CONCLUSIONS The prevalence of dyslipidemia and altered glucose metabolism in our study sets apart the adult population with SMA3 from the general population, confirming a significant interplay between muscle, liver, and adipose tissue. Ensuring metabolic care for aging patients with SMA 3 is crucial, as they are vulnerable to metabolic derangements and cardiovascular risks.
Collapse
Affiliation(s)
- Marija Miletić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.V.); (M.T.G.); (M.S.); (M.Ž.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.S.); (A.P.)
| | - Zorica Stević
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.S.); (A.P.)
- Clinic of Neurology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Svetlana Vujović
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.V.); (M.T.G.); (M.S.); (M.Ž.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.S.); (A.P.)
| | - Jelena Rakočević
- Institute of Histology and Embryology “Aleksandar Đ. Kostić”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ana Tomić
- Center for Radiology Imaging-Magnetic Resonance, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | - Milina Tančić Gajić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.V.); (M.T.G.); (M.S.); (M.Ž.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.S.); (A.P.)
| | - Miloš Stojanović
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.V.); (M.T.G.); (M.S.); (M.Ž.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.S.); (A.P.)
| | - Aleksa Palibrk
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.S.); (A.P.)
- Clinic of Neurology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Miloš Žarković
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (S.V.); (M.T.G.); (M.S.); (M.Ž.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.S.); (A.P.)
| |
Collapse
|
5
|
Liu F, Li R, Zhu Z, Yang Y, Lu F. Current developments of gene therapy in human diseases. MedComm (Beijing) 2024; 5:e645. [PMID: 39156766 PMCID: PMC11329757 DOI: 10.1002/mco2.645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 08/20/2024] Open
Abstract
Gene therapy has witnessed substantial advancements in recent years, becoming a constructive tactic for treating various human diseases. This review presents a comprehensive overview of these developments, with a focus on their diverse applications in different disease contexts. It explores the evolution of gene delivery systems, encompassing viral (like adeno-associated virus; AAV) and nonviral approaches, and evaluates their inherent strengths and limitations. Moreover, the review delves into the progress made in targeting specific tissues and cell types, spanning the eye, liver, muscles, and central nervous system, among others, using these gene technologies. This targeted approach is crucial in addressing a broad spectrum of genetic disorders, such as inherited lysosomal storage diseases, neurodegenerative disorders, and cardiovascular diseases. Recent clinical trials and successful outcomes in gene therapy, particularly those involving AAV and the clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated proteins, are highlighted, illuminating the transformative potentials of this approach in disease treatment. The review summarizes the current status of gene therapy, its prospects, and its capacity to significantly ameliorate patient outcomes and quality of life. By offering comprehensive analysis, this review provides invaluable insights for researchers, clinicians, and stakeholders, enriching the ongoing discourse on the trajectory of disease treatment.
Collapse
Affiliation(s)
- Fanfei Liu
- Department of OphthalmologyWest China HospitalChengduSichuanChina
| | - Ruiting Li
- State Key Laboratory of BiotherapyWest China HospitalChengduSichuanChina
| | - Zilin Zhu
- College of Life SciencesSichuan UniversityChengduSichuanChina
| | - Yang Yang
- Department of OphthalmologyWest China HospitalChengduSichuanChina
- State Key Laboratory of BiotherapyWest China HospitalChengduSichuanChina
| | - Fang Lu
- Department of OphthalmologyWest China HospitalChengduSichuanChina
| |
Collapse
|
6
|
Xue H, Han L, Sun H, Piao Z, Cao W, Qian H, Zhao Z, Lang MF, Gu C. Metastasis-associated 1 localizes to the sarcomeric Z-disc and is implicated in skeletal muscle pathology. Cytoskeleton (Hoboken) 2024; 81:427-435. [PMID: 38391059 DOI: 10.1002/cm.21841] [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: 07/19/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024]
Abstract
Metastasis-associated 1 (MTA1), a subunit of the nucleosome remodeling and histone deacetylation (NuRD) corepressor complex, was reported to be expressed in the cytoplasm of skeletal muscles. However, the exact subcellular localization and the functional implications of MTA1 in skeletal muscles have not been examined. This study aims to demonstrate the subcellular localization of MTA1 in skeletal muscles and reveal its possible roles in skeletal muscle pathogenesis. Striated muscles (skeletal and cardiac) from C57BL/6 mice of 4-5 weeks were collected to examine the expression of MTA1 by Western blotting and immunohistochemistry. Immunofluorescence and immunoelectron microscopy were performed for MTA1, α-actinin (a Z-disc marker protein), and SMN (survival of motor neuron) proteins. Gene Expression Omnibus (GEO) data sets were analyzed using the GEO2R online tool to explore the functional implications of MTA1 in skeletal muscles. MTA1 expression was detected by Western blotting and immunohistochemistry in skeletal and cardiac muscles. Subcellular localization of MTA1 was found in the Z-disc of sarcomeres, where α-actinin and SMN were expressed. Data mining of GEO profiles suggested that MTA1 dysregulation is associated with multiple skeletal muscle defects, such as Duchenne muscular dystrophy, Emery-Dreifuss muscular dystrophy, nemaline myopathy, and dermatomyositis. The GEO analysis also showed that MTA1 expression gradually decreased with age in mouse skeletal muscle precursor cells. The subcellular localization of MTA1 in sarcomeres of skeletal muscles implies its biological roles in sarcomere structures and its possible contribution to skeletal muscle pathology.
Collapse
Affiliation(s)
- Hongsheng Xue
- Department of Thoracic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Thoracic Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Li Han
- Department of Thoracic Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Haidi Sun
- Department of Physiology and Medical Biology, Medical College, Dalian University, Dalian, China
| | - Zhe Piao
- Department of Thoracic Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Wenjun Cao
- Department of Thoracic Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhilong Zhao
- Department of Thoracic Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ming-Fei Lang
- Department of Physiology and Medical Biology, Medical College, Dalian University, Dalian, China
| | - Chundong Gu
- Department of Thoracic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Polverini E, Squeri P, Gherardi V. Effect of E134K pathogenic mutation of SMN protein on SMN-SmD1 interaction, with implication in spinal muscular atrophy: A molecular dynamics study. Int J Biol Macromol 2024; 275:133663. [PMID: 38969036 DOI: 10.1016/j.ijbiomac.2024.133663] [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: 03/14/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Spinal muscular atrophy (SMA) is a disease that results from mutations in the Survival of Motor Neuron (SMN) gene 1, leading to muscle atrophy due to motor neurons degeneration. SMN plays a crucial role in the assembly of spliceosomal small nuclear ribonucleoprotein complexes via binding to the arginine-glycine rich C-terminal tails of Sm proteins recognized by SMN Tudor domain. E134K Tudor mutation, cause of the more severe type I SMA, compromises the SMN-Sm interaction without a perturbation of the domain fold. By molecular dynamics simulations, we investigated the mechanism of Tudor-SmD1 interaction, and the effects on it of E134K mutation. It was observed that E134 is crucial to catch the positive dimethylated arginines (DMRs) of the SmD1 tail that, wrapping around the acidic Tudor surface, enters a central DMR into an aromatic cage. The flexible cage residue Y130 must be blocked from the wrapped tail to assure a stable binding. The charge inversion in E134K mutation causes the loss of a critical anchor point, disfavoring the tail wrapping and leaving Y130 free to swing, leading to DMR detachments and exposition of the C-terminal region of the tail. This could suggest new hypotheses regarding a possible autoimmune response by anti-Sm autoantibodies.
Collapse
Affiliation(s)
- Eugenia Polverini
- Department of Mathematical, Physical and Computer Sciences, University of Parma, Parco Area delle Scienze 7/A, 43124 Parma, Italy.
| | - Pietro Squeri
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| | - Valeria Gherardi
- Department of Chemistry, Life Science and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy
| |
Collapse
|
8
|
Rhee J, Kang J, Jo Y, Yoo K, Kim YL, Hann S, Kim Y, Kim H, Kim J, Kong Y. Improved therapeutic approach for spinal muscular atrophy via ubiquitination-resistant survival motor neuron variant. J Cachexia Sarcopenia Muscle 2024; 15:1404-1417. [PMID: 38650097 PMCID: PMC11294043 DOI: 10.1002/jcsm.13486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/06/2024] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Zolgensma is a gene-replacement therapy that has led to a promising treatment for spinal muscular atrophy (SMA). However, clinical trials of Zolgensma have raised two major concerns: insufficient therapeutic effects and adverse events. In a recent clinical trial, 30% of patients failed to achieve motor milestones despite pre-symptomatic treatment. In addition, more than 20% of patients showed hepatotoxicity due to excessive virus dosage, even after the administration of an immunosuppressant. Here, we aimed to test whether a ubiquitination-resistant variant of survival motor neuron (SMN), SMNK186R, has improved therapeutic effects for SMA compared with wild-type SMN (SMNWT). METHODS A severe SMA mouse model, SMA type 1.5 (Smn-/-; SMN2+/+; SMN∆7+/-) mice, was used to compare the differences in therapeutic efficacy between AAV9-SMNWT and AAV9-SMNK186R. All animals were injected within Postnatal Day (P) 1 through a facial vein or cerebral ventricle. RESULTS AAV9-SMNK186R-treated mice showed increased lifespan, body weight, motor neuron number, muscle weight and functional improvement in motor functions as compared with AAV9-SMNWT-treated mice. Lifespan increased by more than 10-fold in AAV9-SMNK186R-treated mice (144.8 ± 26.11 days) as compared with AAV9-SMNWT-treated mice (26.8 ± 1.41 days). AAV9-SMNK186R-treated mice showed an ascending weight pattern, unlike AAV9-SMNWT-treated mice, which only gained weight until P20 up to 5 g on average. Several motor function tests showed the improved therapeutic efficacy of SMNK186R. In the negative geotaxis test, AAV9-SMNK186R-treated mice turned their bodies in an upward direction successfully, unlike AAV9-SMNWT-treated mice, which failed to turn upwards from around P23. Hind limb clasping phenotype was rarely observed in AAV9-SMNK186R-treated mice, unlike AAV9-SMNWT-treated mice that showed clasping phenotype for more than 20 out of 30 s. At this point, the number of motor neurons (1.5-fold) and the size of myofibers (2.1-fold) were significantly increased in AAV9-SMNK186R-treated mice compared with AAV9-SMNWT-treated mice without prominent neurotoxicity. AAV9-SMNK186R had fewer liver defects compared with AAV9-SMNWT, as judged by increased proliferation of hepatocytes (P < 0.0001) and insulin-like growth factor-1 production (P < 0.0001). Especially, low-dose AAV9-SMNK186R (nine-fold) also reduced clasping time compared with SMNWT. CONCLUSIONS SMNK186R will provide improved therapeutic efficacy in patients with severe SMA with insufficient therapeutic efficacy. Low-dose treatment of SMA patients with AAV9-SMNK186R can reduce the adverse events of Zolgensma. Collectively, SMNK186R has value as a new treatment for SMA that improves treatment effectiveness and reduces adverse events simultaneously.
Collapse
Affiliation(s)
- Joonwoo Rhee
- School of Biological SciencesSeoul National UniversitySeoulSouth Korea
| | - Jong‐Seol Kang
- School of Biological SciencesSeoul National UniversitySeoulSouth Korea
| | - Young‐Woo Jo
- School of Biological SciencesSeoul National UniversitySeoulSouth Korea
| | - Kyusang Yoo
- School of Biological SciencesSeoul National UniversitySeoulSouth Korea
| | - Ye Lynne Kim
- School of Biological SciencesSeoul National UniversitySeoulSouth Korea
| | - Sang‐Hyeon Hann
- School of Biological SciencesSeoul National UniversitySeoulSouth Korea
| | - Yea‐Eun Kim
- School of Biological SciencesSeoul National UniversitySeoulSouth Korea
| | - Hyun Kim
- School of Biological SciencesSeoul National UniversitySeoulSouth Korea
| | - Ji‐Hoon Kim
- Molecular Recognition Research CenterKorea Institute of Science and TechnologySeoulSouth Korea
| | - Young‐Yun Kong
- School of Biological SciencesSeoul National UniversitySeoulSouth Korea
| |
Collapse
|
9
|
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
|
10
|
Haque US, Yokota T. Recent Progress in Gene-Targeting Therapies for Spinal Muscular Atrophy: Promises and Challenges. Genes (Basel) 2024; 15:999. [PMID: 39202360 PMCID: PMC11353366 DOI: 10.3390/genes15080999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a severe genetic disorder characterized by the loss of motor neurons, leading to progressive muscle weakness, loss of mobility, and respiratory complications. In its most severe forms, SMA can result in death within the first two years of life if untreated. The condition arises from mutations in the SMN1 (survival of motor neuron 1) gene, causing a deficiency in the survival motor neuron (SMN) protein. Humans possess a near-identical gene, SMN2, which modifies disease severity and is a primary target for therapies. Recent therapeutic advancements include antisense oligonucleotides (ASOs), small molecules targeting SMN2, and virus-mediated gene replacement therapy delivering a functional copy of SMN1. Additionally, recognizing SMA's broader phenotype involving multiple organs has led to the development of SMN-independent therapies. Evidence now indicates that SMA affects multiple organ systems, suggesting the need for SMN-independent treatments along with SMN-targeting therapies. No single therapy can cure SMA; thus, combination therapies may be essential for comprehensive treatment. This review addresses the SMA etiology, the role of SMN, and provides an overview of the rapidly evolving therapeutic landscape, highlighting current achievements and future directions.
Collapse
Affiliation(s)
- Umme Sabrina Haque
- Department of Neuroscience, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada;
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Toshifumi Yokota
- Department of Neuroscience, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada;
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada HM Toupin Neurological Science Research, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
11
|
Torri F, Mancuso M, Siciliano G, Ricci G. Beyond Motor Neurons in Spinal Muscular Atrophy: A Focus on Neuromuscular Junction. Int J Mol Sci 2024; 25:7311. [PMID: 39000416 PMCID: PMC11242411 DOI: 10.3390/ijms25137311] [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] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
5q-Spinal muscular atrophy (5q-SMA) is one of the most common neuromuscular diseases due to homozygous mutations in the SMN1 gene. This leads to a loss of function of the SMN1 gene, which in the end determines lower motor neuron degeneration. Since the generation of the first mouse models of SMA neuropathology, a complex degenerative involvement of the neuromuscular junction and peripheral axons of motor nerves, alongside lower motor neurons, has been described. The involvement of the neuromuscular junction in determining disease symptoms offers a possible parallel therapeutic target. This narrative review aims at providing an overview of the current knowledge about the pathogenesis and significance of neuromuscular junction dysfunction in SMA, circulating biomarkers, outcome measures and available or developing therapeutic approaches.
Collapse
Affiliation(s)
- Francesca Torri
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Michelangelo Mancuso
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Giulia Ricci
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
12
|
Garg V, Geurten BRH. Diving deep: zebrafish models in motor neuron degeneration research. Front Neurosci 2024; 18:1424025. [PMID: 38966756 PMCID: PMC11222423 DOI: 10.3389/fnins.2024.1424025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 05/30/2024] [Indexed: 07/06/2024] Open
Abstract
In the dynamic landscape of biomedical science, the pursuit of effective treatments for motor neuron disorders like hereditary spastic paraplegia (HSP), amyotrophic lateral sclerosis (ALS), and spinal muscular atrophy (SMA) remains a key priority. Central to this endeavor is the development of robust animal models, with the zebrafish emerging as a prime candidate. Exhibiting embryonic transparency, a swift life cycle, and significant genetic and neuroanatomical congruencies with humans, zebrafish offer substantial potential for research. Despite the difference in locomotion-zebrafish undulate while humans use limbs, the zebrafish presents relevant phenotypic parallels to human motor control disorders, providing valuable insights into neurodegenerative diseases. This review explores the zebrafish's inherent traits and how they facilitate profound insights into the complex behavioral and cellular phenotypes associated with these disorders. Furthermore, we examine recent advancements in high-throughput drug screening using the zebrafish model, a promising avenue for identifying therapeutically potent compounds.
Collapse
Affiliation(s)
- Vranda Garg
- Department of Cellular Neurobiology, Georg-August-University Göttingen, Göttingen, Lower Saxony, Germany
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montreal, QC, Canada
| | | |
Collapse
|
13
|
Lilien C, Vrscaj E, Thapaliya G, Deconinck N, De Waele L, Duong T, Haberlová J, Kumhera M, Peirens G, Szabo L, Tahon V, Tang WJ, Benmhammed N, Médard L, Servais L. Patients' Perceptions of Nusinersen Effects According to Their Responder Status. J Clin Med 2024; 13:3418. [PMID: 38929947 PMCID: PMC11205004 DOI: 10.3390/jcm13123418] [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: 05/09/2024] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Background and Objective: Patients with spinal muscular atrophy (SMA) treated with a disease-modifying therapy (DMT) are often classified as responders or non-responders based on the attainment of a specific improvement threshold on validated functional scales. This categorization may significantly impact treatment reimbursement in some countries. The aim of this research is to evaluate the perception of treatments and their benefit by patients considered as responders or non-responders. Methods: In this non-commercial multicenter study, 99 post-symptomatically treated SMA type I-III patients with a median age of 11.2 (0.39-57.4) years at treatment initiation were stratified into three groups based on their treatment outcomes, i.e., those exhibiting clinically significant improvement (N = 41), those with non-clinically significant improvement (N = 18), or those showing no improvement (N = 40). Fifteen months after treatment, the initiation patients or patients' caregivers were assessed using a patient-rated scoring system based on the Patient Global Impression of Change (PGIC) scale, comprising 22 questions targeting important aspects and tasks in the daily life of patients with SMA. Results: We found no statistical difference in the patient perception of treatment benefits in 17 out of 22 domains across patient groups. Conclusions: Our results suggest that functional motor scales do not recapitulate patients' and patients' caregivers' experience of the effect of nusinersen treatment in SMA.
Collapse
Affiliation(s)
- Charlotte Lilien
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK; (C.L.); (G.T.)
| | - Eva Vrscaj
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Paediatrics, University Hospital Liege & University of Liege, 4000 Liege, Belgium; (E.V.); (N.B.); (L.M.)
- Department for Pediatric Neurology, University Children’s Hospital, University Medical Centre Ljubljana, Bohoričeva 20, 1525 Ljubljana, Slovenia
| | - Gita Thapaliya
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK; (C.L.); (G.T.)
| | - Nicolas Deconinck
- NMRC UZ Gent, Ghent University Hospital, 9000 Ghent, Belgium; (N.D.); (V.T.)
- Neuromuscular Reference Center and Department of Paediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, HUB, Université Libre de Bruxelles, 1020 Brussels, Belgium
| | - Liesbeth De Waele
- Department of Paediatrics, University Hospitals Leuven, 3000 Leuven, Belgium; (L.D.W.); (G.P.)
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Tina Duong
- John W. Day Lab, Department of Neurology and Neurological Sciences, University of Stanford, Stanford, CA 94305, USA; (T.D.); (W.J.T.)
| | - Jana Haberlová
- Department of Paediatric Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, 150 06 Praha, Czech Republic; (J.H.); (M.K.)
| | - Markéta Kumhera
- Department of Paediatric Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, 150 06 Praha, Czech Republic; (J.H.); (M.K.)
| | - Geertrui Peirens
- Department of Paediatrics, University Hospitals Leuven, 3000 Leuven, Belgium; (L.D.W.); (G.P.)
| | - Lena Szabo
- Paediatric Center, Semmelweis University, 1083 Budapest, Hungary;
| | - Valentine Tahon
- NMRC UZ Gent, Ghent University Hospital, 9000 Ghent, Belgium; (N.D.); (V.T.)
| | - Whitney J. Tang
- John W. Day Lab, Department of Neurology and Neurological Sciences, University of Stanford, Stanford, CA 94305, USA; (T.D.); (W.J.T.)
| | - Noor Benmhammed
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Paediatrics, University Hospital Liege & University of Liege, 4000 Liege, Belgium; (E.V.); (N.B.); (L.M.)
| | - Laurie Médard
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Paediatrics, University Hospital Liege & University of Liege, 4000 Liege, Belgium; (E.V.); (N.B.); (L.M.)
| | - Laurent Servais
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK; (C.L.); (G.T.)
- Division of Child Neurology, Reference Center for Neuromuscular Diseases, Department of Paediatrics, University Hospital Liege & University of Liege, 4000 Liege, Belgium; (E.V.); (N.B.); (L.M.)
| |
Collapse
|
14
|
Kubo S, Tanaka Y. Evolution of diagnostic criteria and new insights into clinical testing in mixed connective tissue disease; anti-survival motor neuron complex antibody as a novel marker of severity of the disease. Immunol Med 2024; 47:52-57. [PMID: 38597289 DOI: 10.1080/25785826.2024.2338593] [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: 03/08/2024] [Accepted: 03/31/2024] [Indexed: 04/11/2024] Open
Abstract
Mixed connective tissue disease (MCTD) is an autoimmune disorder characterized by a combination of clinical features from systemic lupus erythematosus, systemic sclerosis, and inflammatory muscle disease, along with the presence of positive anti-U1-ribonucleoprotein (U1-RNP) antibodies. The exact etiology of the disease remains unclear, but it is believed to involve vascular damage within the context of heightened autoimmune responses. Consequently, Raynaud's phenomenon and pulmonary arterial hypertension are observed in patients with MCTD. While specific biomarkers for MCTD have not yet been identified, the recent study of the utility of anti-survival motor neuron complex (SMN) antibodies in MCTD suggests a promising avenue for further research and the accumulation of additional evidence.
Collapse
Affiliation(s)
- Satoshi Kubo
- Department of Molecular Targeted Therapies, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| |
Collapse
|
15
|
Vill K, Tacke M, König A, Baumann M, Baumgartner M, Steinbach M, Bernert G, Blaschek A, Deschauer M, Flotats-Bastardas M, Friese J, Goldbach S, Gross M, Günther R, Hahn A, Hagenacker T, Hauser E, Horber V, Illsinger S, Johannsen J, Kamm C, Koch JC, Koelbel H, Koehler C, Kolzter K, Lochmüller H, Ludolph A, Mensch A, Meyer Zu Hoerste G, Mueller M, Mueller-Felber W, Neuwirth C, Petri S, Probst-Schendzielorz K, Pühringer M, Steinbach R, Schara-Schmidt U, Schimmel M, Schrank B, Schwartz O, Schlachter K, Schwerin-Nagel A, Schreiber G, Smitka M, Topakian R, Trollmann R, Tuerk M, Theophil M, Rauscher C, Vorgerd M, Walter MC, Weiler M, Weiss C, Wilichowski E, Wurster CD, Wunderlich G, Zeller D, Ziegler A, Kirschner J, Pechmann A. 5qSMA: standardised retrospective natural history assessment in 268 patients with four copies of SMN2. J Neurol 2024; 271:2787-2797. [PMID: 38409538 PMCID: PMC11055798 DOI: 10.1007/s00415-024-12188-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 02/28/2024]
Abstract
Newborn screening for 5qSMA offers the potential for early, ideally pre-symptomatic, therapeutic intervention. However, limited data exist on the outcomes of individuals with 4 copies of SMN2, and there is no consensus within the SMA treatment community regarding early treatment initiation in this subgroup. To provide evidence-based insights into disease progression, we performed a retrospective analysis of 268 patients with 4 copies of SMN2 from the SMArtCARE registry in Germany, Austria and Switzerland. Inclusion criteria required comprehensive baseline data and diagnosis outside of newborn screening. Only data prior to initiation of disease-modifying treatment were included. The median age at disease onset was 3.0 years, with a mean of 6.4 years. Significantly, 55% of patients experienced symptoms before the age of 36 months. 3% never learned to sit unaided, a further 13% never gained the ability to walk independently and 33% of ambulatory patients lost this ability during the course of the disease. 43% developed scoliosis, 6.3% required non-invasive ventilation and 1.1% required tube feeding. In conclusion, our study, in line with previous observations, highlights the substantial phenotypic heterogeneity in SMA. Importantly, this study provides novel insights: the median age of disease onset in patients with 4 SMN2 copies typically occurs before school age, and in half of the patients even before the age of three years. These findings support a proactive approach, particularly early treatment initiation, in this subset of SMA patients diagnosed pre-symptomatically. However, it is important to recognize that the register will not include asymptomatic individuals.
Collapse
Affiliation(s)
- Katharina Vill
- Department of Pediatric Neurology and Developmental Medicine and LMU Center for Children With Medical Complexity, Dr. Von Hauner Children's Hospital, LMU Hospital, Ludwig-Maximilians-University, 80337, Munich, Germany.
- School of Medicine, Klinikum Rechts Der Isar, Department of Human Genetics, Technical University of Munich, Munich, Germany.
| | - Moritz Tacke
- Department of Pediatric Neurology and Developmental Medicine and LMU Center for Children With Medical Complexity, Dr. Von Hauner Children's Hospital, LMU Hospital, Ludwig-Maximilians-University, 80337, Munich, Germany
| | - Anna König
- Department of Pediatric Neurology and Developmental Medicine and LMU Center for Children With Medical Complexity, Dr. Von Hauner Children's Hospital, LMU Hospital, Ludwig-Maximilians-University, 80337, Munich, Germany
| | - Matthias Baumann
- Division of Pediatric Neurology, Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Manuela Baumgartner
- Department of Children and Adolescents, Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Meike Steinbach
- Department of Neurology, University Medical Center Schleswig-Holstein, Kiel, Germany
| | | | - Astrid Blaschek
- Department of Pediatric Neurology and Developmental Medicine and LMU Center for Children With Medical Complexity, Dr. Von Hauner Children's Hospital, LMU Hospital, Ludwig-Maximilians-University, 80337, Munich, Germany
| | - Marcus Deschauer
- School of Medicine, Klinikum Rechts Der Isar, Department of Neurology, Technical University of Munich, Munich, Germany
| | | | - Johannes Friese
- Department of Neuropediatrics, University Hospital Bonn, Center for Pediatrics, Bonn, Germany
| | | | - Martin Gross
- Department of Neurological Intensive Care and Rehabilitation, Evangelisches Krankenhaus Oldenburg, Oldenburg, Germany
| | - René Günther
- University Hospital Carl Gustav Carus Dresden at Technische Universität Dresden, Dresden, Germany
| | - Andreas Hahn
- Department of Child Neurology, Justus-Liebig-University Gießen, Gießen, Germany
| | - Tim Hagenacker
- Department of Neurology, and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, Essen, Germany
| | - Erwin Hauser
- Department for Neuropädiatrie, Landeskrankenhaus Mödling, Mödling, Austria
| | - Veronka Horber
- Department of Paediatric Neurology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Sabine Illsinger
- Hannover Medical School, Clinic for Pediatric Kidney-, Liver- and Metabolic Diseases, Hannover, Germany
| | - Jessika Johannsen
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Kamm
- Department of Neurology, University of Rostock, Rostock, Germany
| | - Jan C Koch
- Klinik Für Neurologie Universitätsmedizin Göttingen, Göttingen, Germany
| | - Heike Koelbel
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Cornelia Koehler
- Klinik Für Kinder-Und Jugendmedizin der Ruhr-Universität Bochum Im St. Josef-Hospital, Bochum, Germany
| | - Kirsten Kolzter
- Kliniken Köln, Sozialpädiatrisches Zentrum, Cologne, Germany
| | - Hanns Lochmüller
- Division of Neurology, Department of Medicine, Children's Hospital of Eastern Ontario Research Institute, The Ottawa Hospital and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
- Department of Neuropediatrics and Muscle Disorders, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Albert Ludolph
- Department for Neurology, University of Ulm, Ulm, Germany
- Department of Neurology, Ulm University, Ulm, Germany
| | - Alexander Mensch
- Department of Neurology, University Medicine Halle, Halle, Saale, Germany
| | | | - Monika Mueller
- Department for Neuropediatrics, University of Wuerzburg, Würzburg, Germany
| | - Wolfgang Mueller-Felber
- Department of Pediatric Neurology and Developmental Medicine and LMU Center for Children With Medical Complexity, Dr. Von Hauner Children's Hospital, LMU Hospital, Ludwig-Maximilians-University, 80337, Munich, Germany
| | - Christoph Neuwirth
- Neuromuscular Diseases Unit/ALS Clinic, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | - Manuel Pühringer
- Department of Pediatrics and Adolescent Medicine, Kepler University Hospital Linz, Linz, Austria
| | - Robert Steinbach
- Department of Neurology, University Hospital Jena, Jena, Germany
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Mareike Schimmel
- Pediatric Neurology, Pediatrics and Adolescent Medicine, University Medical Center Augsburg, Augsburg, Germany
| | - Bertold Schrank
- Department of Neurology, DKD Helios Klinik Wiesbaden, Wiesbaden, Germany
| | - Oliver Schwartz
- Universitätsklinikum Münster Klinik Für Kinder- Und Jugendpädiatrie- Neuropädiatrie, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Kurt Schlachter
- Department of Neuropediatrics, Landeskrankenhaus Bregenz, Bregenz, Austria
| | | | | | - Martin Smitka
- Department of Neuropediatrics, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Raffi Topakian
- Department of Neurology, Academic Teaching Hospital Wels-Grieskirchen, Wels, Austria
| | - Regina Trollmann
- Department of Pediatrics, Friedrich-Alexander Universität Erlangen-Nürnberg Pediatric Neurology, Erlangen, Germany
| | - Matthias Tuerk
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Centre for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | | | - Christian Rauscher
- Department for Neuropediatrics, University of Salzburg, Salzburg, Austria
| | - Mathias Vorgerd
- Department of Neurology, BG-University Hospital Bergmannsheil gGmbH, Heimer Institute for Muscle Research, Ruhr-University Bochum, Bochum, Germany
| | - Maggie C Walter
- Friedrich Baur Institute at the Department of Neurology, LMU University Hospital, Ludwig Maximilians University, Munich, Germany
| | - Markus Weiler
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Claudia Weiss
- Charité - University Medicine Berlin, Center for Chronically Sick Children, Berlin, Germany
| | | | | | - Gilbert Wunderlich
- German Center for Neurodegenerative Diseases, DZNE, Site Ulm, Ulm, Germany
- Faculty of Medicine and University Hospital, Department of Neurology and Center for Rare Diseases, University of Cologne, Cologne, Germany
| | - Daniel Zeller
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Ziegler
- Center for Childhood and Adolescent Medicine, Department of Metabolic Medicine and Pediatric Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Janbernd Kirschner
- Klinik Für Kinder-Und Jugendmedizin der Ruhr-Universität Bochum Im St. Josef-Hospital, Bochum, Germany
| | - Astrid Pechmann
- Klinik Für Kinder-Und Jugendmedizin der Ruhr-Universität Bochum Im St. Josef-Hospital, Bochum, Germany
| |
Collapse
|
16
|
Cottam NC, Harrington MA, Schork PM, Sun J. No significant sex differences in incidence or phenotype for the SMNΔ7 mouse model of spinal muscular atrophy. Neuromuscul Disord 2024; 37:13-22. [PMID: 38493520 PMCID: PMC11031329 DOI: 10.1016/j.nmd.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive disease that affects 1 out of every 6,000-10,000 individuals at birth, making it the leading genetic cause of infant mortality. In recent years, reports of sex differences in SMA patients have become noticeable. The SMNΔ7 mouse model is commonly used to investigate pathologies and treatments in SMA. However, studies on sex as a contributing biological variable are few and dated. Here, we rigorously investigated the effect of sex on a series of characteristics in SMA mice of the SMNΔ7 model. Incidence and lifespan of 23 mouse litters were tracked and phenotypic assessments were performed at 2-day intervals starting at postnatal day 6 for every pup until the death of the SMA pup(s) in each litter. Brain weights were also collected post-mortem. We found that male and female SMA incidence does not differ significantly, survival periods are the same across sexes, and there was no phenotypic difference between male and female SMA pups, other than for females exhibiting lesser body weights at early ages. Overall, this study ensures that sex is not a biological variable that contributes to the incidence ratio or disease severity in the SMNΔ7 mouse model.
Collapse
Affiliation(s)
- Nicholas C Cottam
- Delaware State University, Department of Biological Sciences, 1200 N Dupont Highway, Dover, DE, USA
| | - Melissa A Harrington
- Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| | - Pamela M Schork
- Delaware State University, Department of Biological Sciences, 1200 N Dupont Highway, Dover, DE, USA
| | - Jianli Sun
- Delaware State University, Department of Biological Sciences, 1200 N Dupont Highway, Dover, DE, USA; Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA.
| |
Collapse
|
17
|
Sharma G, Paganin M, Lauria F, Perenthaler E, Viero G. The SMN-ribosome interplay: a new opportunity for Spinal Muscular Atrophy therapies. Biochem Soc Trans 2024; 52:465-479. [PMID: 38391004 PMCID: PMC10903476 DOI: 10.1042/bst20231116] [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: 12/22/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
The underlying cause of Spinal Muscular Atrophy (SMA) is in the reduction of survival motor neuron (SMN) protein levels due to mutations in the SMN1 gene. The specific effects of SMN protein loss and the resulting pathological alterations are not fully understood. Given the crucial roles of the SMN protein in snRNP biogenesis and its interactions with ribosomes and translation-related proteins and mRNAs, a decrease in SMN levels below a specific threshold in SMA is expected to affect translational control of gene expression. This review covers both direct and indirect SMN interactions across various translation-related cellular compartments and processes, spanning from ribosome biogenesis to local translation and beyond. Additionally, it aims to outline deficiencies and alterations in translation observed in SMA models and patients, while also discussing the implications of the relationship between SMN protein and the translation machinery within the context of current and future therapies.
Collapse
|
18
|
Gonzalez D, Vásquez-Doorman C, Luna A, Allende ML. Modeling Spinal Muscular Atrophy in Zebrafish: Current Advances and Future Perspectives. Int J Mol Sci 2024; 25:1962. [PMID: 38396640 PMCID: PMC10888324 DOI: 10.3390/ijms25041962] [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: 11/15/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 02/25/2024] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive neurodegenerative disease characterized by degeneration of lower motor neurons (LMNs), causing muscle weakness, atrophy, and paralysis. SMA is caused by mutations in the Survival Motor Neuron 1 (SMN1) gene and can be classified into four subgroups, depending on its severity. Even though the genetic component of SMA is well known, the precise mechanisms underlying its pathophysiology remain elusive. Thus far, there are three FDA-approved drugs for treating SMA. While these treatments have shown promising results, their costs are extremely high and unaffordable for most patients. Thus, more efforts are needed in order to identify novel therapeutic targets. In this context, zebrafish (Danio rerio) stands out as an ideal animal model for investigating neurodegenerative diseases like SMA. Its well-defined motor neuron circuits and straightforward neuromuscular structure offer distinct advantages. The zebrafish's suitability arises from its low-cost genetic manipulation and optical transparency exhibited during larval stages, which facilitates in vivo microscopy. This review explores advancements in SMA research over the past two decades, beginning with the creation of the first zebrafish model. Our review focuses on the findings using different SMA zebrafish models generated to date, including potential therapeutic targets such as U snRNPs, Etv5b, PLS3, CORO1C, Pgrn, Cpg15, Uba1, Necdin, and Pgk1, among others. Lastly, we conclude our review by emphasizing the future perspectives in the field, namely exploiting zebrafish capacity for high-throughput screening. Zebrafish, with its unique attributes, proves to be an ideal model for studying motor neuron diseases and unraveling the complexity of neuromuscular defects.
Collapse
Affiliation(s)
- David Gonzalez
- Millennium Institute Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, RM, Chile
- Departamento de Ciencias Químicas y Biológicas, Facultad de Ciencias de la Salud, Universidad Bernardo O'Higgins, Santiago 8370854, RM, Chile
| | - Constanza Vásquez-Doorman
- Millennium Institute Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, RM, Chile
- Departamento de Ciencias Químicas y Biológicas, Facultad de Ciencias de la Salud, Universidad Bernardo O'Higgins, Santiago 8370854, RM, Chile
| | - Adolfo Luna
- Departamento de Ciencias Químicas y Biológicas, Facultad de Ciencias de la Salud, Universidad Bernardo O'Higgins, Santiago 8370854, RM, Chile
| | - Miguel L Allende
- Millennium Institute Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago 7800003, RM, Chile
| |
Collapse
|
19
|
Hann SH, Kim SY, Kim YL, Jo YW, Kang JS, Park H, Choi SY, Kong YY. Depletion of SMN protein in mesenchymal progenitors impairs the development of bone and neuromuscular junction in spinal muscular atrophy. eLife 2024; 12:RP92731. [PMID: 38318851 PMCID: PMC10945524 DOI: 10.7554/elife.92731] [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: 02/07/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder characterized by the deficiency of the survival motor neuron (SMN) protein, which leads to motor neuron dysfunction and muscle atrophy. In addition to the requirement for SMN in motor neurons, recent studies suggest that SMN deficiency in peripheral tissues plays a key role in the pathogenesis of SMA. Using limb mesenchymal progenitor cell (MPC)-specific SMN-depleted mouse models, we reveal that SMN reduction in limb MPCs causes defects in the development of bone and neuromuscular junction (NMJ). Specifically, these mice exhibited impaired growth plate homeostasis and reduced insulin-like growth factor (IGF) signaling from chondrocytes, rather than from the liver. Furthermore, the reduction of SMN in fibro-adipogenic progenitors (FAPs) resulted in abnormal NMJ maturation, altered release of neurotransmitters, and NMJ morphological defects. Transplantation of healthy FAPs rescued the morphological deterioration. Our findings highlight the significance of mesenchymal SMN in neuromusculoskeletal pathogenesis of SMA and provide insights into potential therapeutic strategies targeting mesenchymal cells for the treatment of SMA.
Collapse
Affiliation(s)
- Sang-Hyeon Hann
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seon-Yong Kim
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Ye Lynne Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Woo Jo
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jong-Seol Kang
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyerim Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Young-Yun Kong
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
20
|
Rashid S, Dimitriadi M. Autophagy in spinal muscular atrophy: from pathogenic mechanisms to therapeutic approaches. Front Cell Neurosci 2024; 17:1307636. [PMID: 38259504 PMCID: PMC10801191 DOI: 10.3389/fncel.2023.1307636] [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: 10/04/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a devastating neuromuscular disorder caused by the depletion of the ubiquitously expressed survival motor neuron (SMN) protein. While the genetic cause of SMA has been well documented, the exact mechanism(s) by which SMN depletion results in disease progression remain elusive. A wide body of evidence has highlighted the involvement and dysregulation of autophagy in SMA. Autophagy is a highly conserved lysosomal degradation process which is necessary for cellular homeostasis; defects in the autophagic machinery have been linked with a wide range of neurodegenerative disorders, including amyotrophic lateral sclerosis, Alzheimer's disease and Parkinson's disease. The pathway is particularly known to prevent neurodegeneration and has been suggested to act as a neuroprotective factor, thus presenting an attractive target for novel therapies for SMA patients. In this review, (a) we provide for the first time a comprehensive summary of the perturbations in the autophagic networks that characterize SMA development, (b) highlight the autophagic regulators which may play a key role in SMA pathogenesis and (c) propose decreased autophagic flux as the causative agent underlying the autophagic dysregulation observed in these patients.
Collapse
Affiliation(s)
| | - Maria Dimitriadi
- School of Life and Medical Science, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
21
|
Deng C, Chen H. Brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling in spinal muscular atrophy and amyotrophic lateral sclerosis. Neurobiol Dis 2024; 190:106377. [PMID: 38092270 DOI: 10.1016/j.nbd.2023.106377] [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: 09/17/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023] Open
Abstract
Tropomyosin receptor kinase B (TrkB) and its primary ligand brain-derived neurotrophic factor (BDNF) are expressed in the neuromuscular system, where they affect neuronal survival, differentiation, and functions. Changes in BDNF levels and full-length TrkB (TrkB-FL) signaling have been revealed in spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS), two common forms of motor neuron diseases that are characterized by defective neuromuscular junctions in early disease stages and subsequently progressive muscle weakness. This review summarizes the current understanding of BDNF/TrkB-FL-related research in SMA and ALS, with an emphasis on their alterations in the neuromuscular system and possible BDNF/TrkB-FL-targeting therapeutic strategies. The limitations of current studies and future directions are also discussed, giving the hope of discovering novel and effective treatments.
Collapse
Affiliation(s)
- Chunchu Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
22
|
Novikova ES. [Experience with the use of risdiplam in a familial case of spinal muscular atrophy 5q in patients with a homozygous deletion of the SMN1 gene and the same copy number of the SMN2 gene]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:138-141. [PMID: 38884441 DOI: 10.17116/jnevro2024124051138] [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: 06/18/2024]
Abstract
Advances in the treatment of spinal muscular atrophy (SMA) have revolutionized the field. SMA is a rare autosomal recessive neurodegenerative motor neuron disease in which wide phenotypic variability has been described. The rate of increase in neurological deficit and the severity of the disease is mainly determined by the amount of functional SMN (Survival of Motor Neuron) protein. However, the clinical picture may differ significantly in patients carrying homozygous deletions of the SMN1 gene (Survival of Motor Neuron 1) and an identical number of copies of the SMN2 gene (Survival of Motor Neuron 2). A family clinical case of adult patients with spinal muscular atrophy 5q with a homozygous deletion of the SMN1 gene and the same number of copies of the SMN2 gene, having a different clinical picture of the disease, is presented, and the dynamics of the condition against the background of oral pathogenetic therapy is presented.
Collapse
Affiliation(s)
- E S Novikova
- Vladimirsky Moscow Regional Research and Clinical Institute, Moscow, Russia
| |
Collapse
|
23
|
Bhat MA, Dhaneshwar S. Neurodegenerative Diseases: New Hopes and Perspectives. Curr Mol Med 2024; 24:1004-1032. [PMID: 37691199 DOI: 10.2174/1566524023666230907093451] [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: 05/29/2023] [Revised: 07/10/2023] [Accepted: 07/27/2023] [Indexed: 09/12/2023]
Abstract
Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, Huntington's disease, and Friedrich ataxia are all incurable neurodegenerative diseases defined by the continuous progressive loss of distinct neuronal subtypes. Despite their rising prevalence among the world's ageing population, fewer advances have been made in the concurrent massive efforts to develop newer drugs. Recently, there has been a shift in research focus towards the discovery of new therapeutic agents for neurodegenerative diseases. In this review, we have summarized the recently developed therapies and their status in the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mohammad Aadil Bhat
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, Noida, UP, India
| | - Suneela Dhaneshwar
- Amity Institute of Pharmacy, Amity University Maharashtra, Mumbai, Maharashtra, India
| |
Collapse
|
24
|
Kong L, Li S, Zhao Z, Feng J, Fu X, Li H, Zhu J, Wang Y, Tang W, Yuan C, Li F, Han X, Wu D, Kong X, Sun L. Exploring factors impacting haplotype-based noninvasive prenatal diagnosis for single-gene recessive disorders. Clin Genet 2024; 105:52-61. [PMID: 37822034 DOI: 10.1111/cge.14434] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Haplotype-based noninvasive prenatal diagnosis (NIPD) is applicable for various recessive single-gene disorders in proband families. However, a comprehensive exploration of critical factors influencing the assay performance, such as fetal fraction, informative single nucleotide polymorphism (SNP) count, and recombination events, has yet to be performed. It is critical to identify key factors affecting NIPD performance, including its accuracy and success rate, and their impact on clinical diagnostics to guide clinical practice. We conducted a prospective study, recruiting 219 proband families with singleton pregnancies at risk for eight recessive single-gene disorders (Duchenne muscular dystrophy, spinal muscular atrophy, phenylketonuria, methylmalonic acidemia, hemophilia A, hemophilia B, non-syndromic hearing loss, and congenital adrenal hyperplasia) at 7-14 weeks of gestation. Haplotype-based NIPD was performed by evaluating the relative haplotype dosage (RHDO) in maternal circulation, and the results were validated via invasive prenatal diagnosis or newborn follow-ups. Among the 219 families, the median gestational age at first blood draw was 8+5 weeks. Initial testing succeeded for 190 families and failed for 29 due to low fetal fraction (16), insufficient informative SNPs (9), and homologous recombination near pathogenic variation (4). Among low fetal fraction families, successful testing was achieved for 11 cases after a redraw, while 5 remained inconclusive. Test failures linked to insufficient informative SNPs correlated with linkage disequilibrium near the genes, with F8 and MMUT exhibiting the highest associated failure rates (14.3% and 25%, respectively). Homologous recombination was relatively frequent around the DMD and SMN1 genes (8.8% and 4.8%, respectively) but led to detection failure in only 44.4% (4/9) of such cases. All NIPD results from the 201 successful families were consistent with invasive diagnostic findings or newborn follow-up. Fetal fraction, informative SNPs count, and homologous recombination are pivotal to NIPD performance. Redrawing blood effectively improves the success rate for low fetal fraction samples. However, informative SNPs count and homologous recombination rates vary significantly across genes, necessitating careful consideration in clinical practice. We have designed an in silico method based on linkage disequilibrium data to predict the number of informative SNPs. This can identify genomic regions where there might be an insufficient number of SNPs, thereby guiding panel design. With these factors properly accounted for, NIPD is highly accurate and reliable.
Collapse
Affiliation(s)
- Lingrong Kong
- Department of Fetal Medicine & Prenatal Diagnosis Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shaojun Li
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenhua Zhao
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Feng
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Xinyu Fu
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huanyun Li
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingqi Zhu
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanan Wang
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weiqin Tang
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Chao Yuan
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Feifei Li
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Xiujuan Han
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Di Wu
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Xiangdong Kong
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Luming Sun
- Department of Fetal Medicine & Prenatal Diagnosis Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
25
|
Kelly KM, Mizell J, Bigdeli L, Paul S, Tellez MA, Bartlett A, Heintzman S, Reynolds JE, Sterling GB, Rajneesh KF, Kolb SJ, Elsheikh B, Arnold WD. Differential impact on motor unit characteristics across severities of adult spinal muscular atrophy. Ann Clin Transl Neurol 2023; 10:2208-2222. [PMID: 37735861 PMCID: PMC10723249 DOI: 10.1002/acn3.51906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 08/22/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023] Open
Abstract
OBJECTIVE To test the hypotheses that decomposition electromyography (dEMG) motor unit action potential (MUAP) amplitude and firing rate are altered in SMA; dEMG parameters are associated with strength and function; dEMG parameters are correlated with traditional electrophysiological assessments. METHODS Ambulatory and non-ambulatory adults with SMA on nusinersen and healthy controls were enrolled. MUAPs were decomposed from multielectrode surface recordings during 30-s maximum contraction of the abductor digiti minimi (ADM). Isometric strength, upper limb function, patient-reported function, and standard electrophysiologic measures of the ADM (compound muscle action potential [CMAP], single motor unit potential [SMUP], motor unit number estimation [MUNE]) were collected. RESULTS dEMG MUAP amplitudes were higher in ambulatory versus control and non-ambulatory groups and were higher in controls versus non-ambulatory SMA. In contrast, dEMG firing rates were higher in ambulatory versus non-ambulatory and control groups but similar between non-ambulatory and control. dEMG parameters showed moderate to strong positive correlation with strength and function whereas CMAP and MUNE better correlated with function than strength. SMUP did not correlate with strength, function, or dEMG MUAP amplitude. dEMG parameters show overall good test-retest reliability. INTERPRETATION dEMG provided reliable, noninvasive measure of MUAP amplitude size and firing rate and revealed divergent patterns across disease severity in adults with SMA. Firing rate enhancement, as seen in milder SMA, may provide a therapeutic avenue for improving function in more severe SMA, where firing rates appear preserved. MUAP amplitude size and firing rate, quantified with dEMG, may be promising monitoring biomarker candidates for noninvasive assessment of SMA.
Collapse
Affiliation(s)
- Kristina Marie Kelly
- Department of Physical Medicine & RehabilitationUniversity of MissouriColumbiaMOUSA
- NextGen Precision HealthUniversity of MissouriColumbiaMOUSA
| | - Jordan Mizell
- College of MedicineThe Ohio State UniversityColumbusOHUSA
| | - Ladan Bigdeli
- College of MedicineThe Ohio State UniversityColumbusOHUSA
| | - Samuel Paul
- College of MedicineThe Ohio State UniversityColumbusOHUSA
| | - Marco Antonio Tellez
- Department of NeurologyThe Ohio State University Wexner Medical CenterColumbusOHUSA
| | - Amy Bartlett
- Center for Clinical and Translational ScienceThe Ohio State University Wexner Medical CenterColumbusOHUSA
| | - Sarah Heintzman
- Department of NeurologyThe Ohio State University Wexner Medical CenterColumbusOHUSA
| | | | - Gary Brent Sterling
- Department of NeurologyThe Ohio State University Wexner Medical CenterColumbusOHUSA
| | | | - Stephen James Kolb
- Department of NeurologyThe Ohio State University Wexner Medical CenterColumbusOHUSA
| | - Bakri Elsheikh
- Department of NeurologyThe Ohio State University Wexner Medical CenterColumbusOHUSA
| | - William David Arnold
- Department of Physical Medicine & RehabilitationUniversity of MissouriColumbiaMOUSA
- NextGen Precision HealthUniversity of MissouriColumbiaMOUSA
| |
Collapse
|
26
|
Miller N, Xu Z, Quinlan KA, Ji A, McGivern JV, Feng Z, Shi H, Ko CP, Tsai LH, Heckman CJ, Ebert AD, Ma YC. Mitigating aberrant Cdk5 activation alleviates mitochondrial defects and motor neuron disease symptoms in spinal muscular atrophy. Proc Natl Acad Sci U S A 2023; 120:e2300308120. [PMID: 37976261 PMCID: PMC10666147 DOI: 10.1073/pnas.2300308120] [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: 01/06/2023] [Accepted: 07/31/2023] [Indexed: 11/19/2023] Open
Abstract
Spinal muscular atrophy (SMA), the top genetic cause of infant mortality, is characterized by motor neuron degeneration. Mechanisms underlying SMA pathogenesis remain largely unknown. Here, we report that the activity of cyclin-dependent kinase 5 (Cdk5) and the conversion of its activating subunit p35 to the more potent activator p25 are significantly up-regulated in mouse models and human induced pluripotent stem cell (iPSC) models of SMA. The increase of Cdk5 activity occurs before the onset of SMA phenotypes, suggesting that it may be an initiator of the disease. Importantly, aberrant Cdk5 activation causes mitochondrial defects and motor neuron degeneration, as the genetic knockout of p35 in an SMA mouse model rescues mitochondrial transport and fragmentation defects, and alleviates SMA phenotypes including motor neuron hyperexcitability, loss of excitatory synapses, neuromuscular junction denervation, and motor neuron degeneration. Inhibition of the Cdk5 signaling pathway reduces the degeneration of motor neurons derived from SMA mice and human SMA iPSCs. Altogether, our studies reveal a critical role for the aberrant activation of Cdk5 in SMA pathogenesis and suggest a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Nimrod Miller
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL60611
| | - Zhaofa Xu
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL60611
| | - Katharina A. Quinlan
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Physical Therapy and Human Movement Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI02881
| | - Amy Ji
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Jered V. McGivern
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI53226
| | - Zhihua Feng
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA90089
| | - Han Shi
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL60611
| | - Chien-Ping Ko
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA90089
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Charles J. Heckman
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Physical Therapy and Human Movement Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI53226
| | - Yongchao C. Ma
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Stanley Manne Children's Research Institute, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL60611
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| |
Collapse
|
27
|
Giorgia Q, Gomez Garcia de la Banda M, Smeriglio P. Role of circulating biomarkers in spinal muscular atrophy: insights from a new treatment era. Front Neurol 2023; 14:1226969. [PMID: 38020652 PMCID: PMC10679720 DOI: 10.3389/fneur.2023.1226969] [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: 05/23/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a lower motor neuron disease due to biallelic mutations in the SMN1 gene on chromosome 5. It is characterized by progressive muscle weakness of limbs, bulbar and respiratory muscles. The disease is usually classified in four different phenotypes (1-4) according to age at symptoms onset and maximal motor milestones achieved. Recently, three disease modifying treatments have received approval from the Food and Drug Administration (FDA) and the European Medicines Agency (EMA), while several other innovative drugs are under study. New therapies have been game changing, improving survival and life quality for SMA patients. However, they have also intensified the need for accurate biomarkers to monitor disease progression and treatment efficacy. While clinical and neurophysiological biomarkers are well established and helpful in describing disease progression, there is a great need to develop more robust and sensitive circulating biomarkers, such as proteins, nucleic acids, and other small molecules. Used alone or in combination with clinical biomarkers, they will play a critical role in enhancing patients' stratification for clinical trials and access to approved treatments, as well as in tracking response to therapy, paving the way to the development of individualized therapeutic approaches. In this comprehensive review, we describe the foremost circulating biomarkers of current significance, analyzing existing literature on non-treated and treated patients with a special focus on neurofilaments and circulating miRNA, aiming to identify and examine their role in the follow-up of patients treated with innovative treatments, including gene therapy.
Collapse
Affiliation(s)
- Querin Giorgia
- APHP, Service de Neuromyologie, Hôpital Pitié-Salpêtrière, Centre Référent pour les Maladies Neuromusculaires Nord/Est/Ile de France, Paris, France
- Institut de Myologie, I-Motion Clinical Trials Platform, Paris, France
- European Reference Center Network (Euro-NMD ERN), Paris, France
| | - Marta Gomez Garcia de la Banda
- Institut de Myologie, I-Motion Clinical Trials Platform, Paris, France
- APHP, Pediatric Neurology Department, Hôpital Armand Trousseau, Centre Référent pour les Maladies Neuromusculaires Nord/Est/Ile de France, Paris, France
- APHP, Pediatric Neurology and ICU Department, Université Paris Saclay, DMU Santé de l'Enfant et de l'Adolescent, Hôpital Raymond Poincaré, Garches, France
| | - Piera Smeriglio
- Centre of Research in Myology, Institute of Myology, Sorbonne Université, INSERM, Paris, France
| |
Collapse
|
28
|
Yang XC, Desotell A, Lin MH, Paige AS, Malinowska A, Sun Y, Aik WS, Dadlez M, Tong L, Dominski Z. In vitro methylation of the U7 snRNP subunits Lsm11 and SmE by the PRMT5/MEP50/pICln methylosome. RNA (NEW YORK, N.Y.) 2023; 29:1673-1690. [PMID: 37562960 PMCID: PMC10578488 DOI: 10.1261/rna.079709.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/08/2023] [Indexed: 08/12/2023]
Abstract
U7 snRNP is a multisubunit endonuclease required for 3' end processing of metazoan replication-dependent histone pre-mRNAs. In contrast to the spliceosomal snRNPs, U7 snRNP lacks the Sm subunits D1 and D2 and instead contains two related proteins, Lsm10 and Lsm11. The remaining five subunits of the U7 heptameric Sm ring, SmE, F, G, B, and D3, are shared with the spliceosomal snRNPs. The pathway that assembles the unique ring of U7 snRNP is unknown. Here, we show that a heterodimer of Lsm10 and Lsm11 tightly interacts with the methylosome, a complex of the arginine methyltransferase PRMT5, MEP50, and pICln known to methylate arginines in the carboxy-terminal regions of the Sm proteins B, D1, and D3 during the spliceosomal Sm ring assembly. Both biochemical and cryo-EM structural studies demonstrate that the interaction is mediated by PRMT5, which binds and methylates two arginine residues in the amino-terminal region of Lsm11. Surprisingly, PRMT5 also methylates an amino-terminal arginine in SmE, a subunit that does not undergo this type of modification during the biogenesis of the spliceosomal snRNPs. An intriguing possibility is that the unique methylation pattern of Lsm11 and SmE plays a vital role in the assembly of the U7 snRNP.
Collapse
Affiliation(s)
- Xiao-Cui Yang
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Anthony Desotell
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Min-Han Lin
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Andrew S Paige
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Agata Malinowska
- Department of Biophysics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Yadong Sun
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Wei Shen Aik
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Michał Dadlez
- Department of Biophysics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Institute of Genetics and Biotechnology, Warsaw University, 02-106 Warsaw, Poland
| | - Liang Tong
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Zbigniew Dominski
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
29
|
Ling Q, Herstine JA, Bradbury A, Gray SJ. AAV-based in vivo gene therapy for neurological disorders. Nat Rev Drug Discov 2023; 22:789-806. [PMID: 37658167 DOI: 10.1038/s41573-023-00766-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2023] [Indexed: 09/03/2023]
Abstract
Recent advancements in gene supplementation therapy are expanding the options for the treatment of neurological disorders. Among the available delivery vehicles, adeno-associated virus (AAV) is often the favoured vector. However, the results have been variable, with some trials dramatically altering the course of disease whereas others have shown negligible efficacy or even unforeseen toxicity. Unlike traditional drug development with small molecules, therapeutic profiles of AAV gene therapies are dependent on both the AAV capsid and the therapeutic transgene. In this rapidly evolving field, numerous clinical trials of gene supplementation for neurological disorders are ongoing. Knowledge is growing about factors that impact the translation of preclinical studies to humans, including the administration route, timing of treatment, immune responses and limitations of available model systems. The field is also developing potential solutions to mitigate adverse effects, including AAV capsid engineering and designs to regulate transgene expression. At the same time, preclinical research is addressing new frontiers of gene supplementation for neurological disorders, with a focus on mitochondrial and neurodevelopmental disorders. In this Review, we describe the current state of AAV-mediated neurological gene supplementation therapy, including critical factors for optimizing the safety and efficacy of treatments, as well as unmet needs in this field.
Collapse
Affiliation(s)
- Qinglan Ling
- Department of Paediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jessica A Herstine
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Paediatrics, The Ohio State University, Columbus, OH, USA
| | - Allison Bradbury
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Paediatrics, The Ohio State University, Columbus, OH, USA
| | - Steven J Gray
- Department of Paediatrics, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
30
|
Hu Y, Hou Y, Zhou S, Wang Y, Shen C, Mu L, Su D, Zhang R. Mechanism of assembly of snRNP cores assisted by ICln and the SMN complex in fission yeast. iScience 2023; 26:107604. [PMID: 37664592 PMCID: PMC10470402 DOI: 10.1016/j.isci.2023.107604] [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: 05/24/2023] [Revised: 07/11/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023] Open
Abstract
The spliceosomal snRNP cores, each comprised of a snRNA and a seven-membered Sm ring (D1/D2/F/E/G/D3/B), are assembled by twelve chaperoning proteins in human. However, only six assembly-assisting proteins, ICln and the SMN complex (SMN/Gemin2/Gemin6-8), have been found in Schizosaccharomyces pombe (Sp). Here, we used recombinant proteins to reconstitute the chaperone machinery and investigated the roles of these proteins systematically. We found that, like the human system, the assembly in S. pombe requires ICln and the SMN complex sequentially. However, there are several significant differences. For instance, h_F/E/G forms heterohexamers and heterotrimers, while Sp_F/E/G only forms heterohexamers; h_Gemin2 alone can bind D1/D2/F/E/G, but Sp_Gemin2 cannot. Moreover, we found that Sp_Gemin2 is essential using genetic approaches. These mechanistic studies reveal that these six proteins are necessary and sufficient for Sm core assembly at the molecular level, and enrich our understanding of the chaperone systems in species variation and evolution.
Collapse
Affiliation(s)
- Yan Hu
- Department of Ophthalmology, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
| | - Yan Hou
- Department of Ophthalmology, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
| | - Shijie Zhou
- Department of Ophthalmology, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
| | - Yingzhi Wang
- Department of Ophthalmology, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
| | - Congcong Shen
- Department of Ophthalmology, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
| | - Li Mu
- Department of Ophthalmology, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
| | - Dan Su
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
| | - Rundong Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P.R. China
| |
Collapse
|
31
|
Cui Q, Wang W, Namani A, Wang H, Hammad A, Huang P, Gao Y, Elshaer M, Wu Y, Wang XJ, Tang X. NRF2 has a splicing regulatory function involving the survival of motor neuron (SMN) in non-small cell lung cancer. Oncogene 2023; 42:2751-2763. [PMID: 37573407 DOI: 10.1038/s41388-023-02799-z] [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: 02/06/2021] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/14/2023]
Abstract
The nuclear factor erythroid 2-like 2 (NFE2L2; NRF2) signaling pathway is frequently deregulated in human cancers. The critical functions of NRF2, other than its transcriptional activation, in cancers remain largely unknown. Here, we uncovered a previously unrecognized role of NRF2 in the regulation of RNA splicing. Global splicing analysis revealed that NRF2 knockdown in non-small cell lung cancer (NSCLC) A549 cells altered 839 alternative splicing (AS) events in 485 genes. Mechanistic studies demonstrated that NRF2 transcriptionally regulated SMN mRNA expression by binding to two antioxidant response elements in the SMN1 promoter. Post-transcriptionally, NRF2 was physically associated with the SMN protein. The Neh2 domain of NRF2, as well as the YG box and the region encoded by exon 7 of SMN, were required for their interaction. NRF2 formed a complex with SMN and Gemin2 in nuclear gems and Cajal bodies. Furthermore, the NRF2-SMN interaction regulated RNA splicing by expressing SMN in NRF2-knockout HeLa cells, reverting some of the altered RNA splicing. Moreover, SMN overexpression was significantly associated with alterations in the NRF2 pathway in patients with lung squamous cell carcinoma from The Cancer Genome Atlas. Taken together, our findings suggest a novel therapeutic strategy for cancers involving an aberrant NRF2 pathway.
Collapse
Affiliation(s)
- Qinqin Cui
- Department of Biochemistry and Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Wei Wang
- Department of Biochemistry and Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Akhileshwar Namani
- Department of Biochemistry and Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Hongyan Wang
- Department of Biochemistry and Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Ahmed Hammad
- Department of Biochemistry and Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Pu Huang
- Department of Biochemistry and Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Yang Gao
- Department of Biochemistry and Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Mohamed Elshaer
- Department of Biochemistry and Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Yihe Wu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Xiu Jun Wang
- Department of Pharmacology and Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China.
| | - Xiuwen Tang
- Department of Biochemistry and Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China.
- Zhejiang Provincial Key Laboratory of Immunity and Inflammatory Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China.
| |
Collapse
|
32
|
Cottam NC, Bamfo T, Harrington MA, Charvet CJ, Hekmatyar K, Tulin N, Sun J. Cerebellar structural, astrocytic, and neuronal abnormalities in the SMNΔ7 mouse model of spinal muscular atrophy. Brain Pathol 2023; 33:e13162. [PMID: 37218083 PMCID: PMC10467044 DOI: 10.1111/bpa.13162] [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: 11/18/2022] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Spinalmuscular atrophy (SMA) is a neuromuscular disease that affects as many as 1 in 6000 individuals at birth, making it the leading genetic cause of infant mortality. A growing number of studies indicate that SMA is a multi-system disease. The cerebellum has received little attention even though it plays an important role in motor function and widespread pathology has been reported in the cerebella of SMA patients. In this study, we assessed SMA pathology in the cerebellum using structural and diffusion magnetic resonance imaging, immunohistochemistry, and electrophysiology with the SMNΔ7 mouse model. We found a significant disproportionate loss in cerebellar volume, decrease in afferent cerebellar tracts, selective lobule-specific degeneration of Purkinje cells, abnormal lobule foliation and astrocyte integrity, and a decrease in spontaneous firing of cerebellar output neurons in the SMA mice compared to controls. Our data suggest that defects in cerebellar structure and function due to decreased survival motor neuron (SMN) levels impair the functional cerebellar output affecting motor control, and that cerebellar pathology should be addressed to achieve comprehensive treatment and therapy for SMA patients.
Collapse
Affiliation(s)
- Nicholas C. Cottam
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
| | - Tiffany Bamfo
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
| | | | - Christine J. Charvet
- Delaware Center for Neuroscience ResearchDelaware State UniversityDoverDelawareUSA
- Department of Anatomy, Physiology and PharmacologyAuburn UniversityAuburnAlabamaUSA
- Department of PsychologyDelaware State UniversityDoverDEUnited States
| | - Khan Hekmatyar
- Center for Biomedical and Brain ImagingUniversity of DelawareNewarkDelawareUSA
- Bioimaging Research Center for Biomedical and Brain ImagingUniversity of GeorgiaAthensGeorgiaUSA
| | - Nikita Tulin
- Department of NeuroscienceTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Jianli Sun
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
- Delaware Center for Neuroscience ResearchDelaware State UniversityDoverDelawareUSA
| |
Collapse
|
33
|
Fortuna TR, Kour S, Chimata AV, Muiños-Bühl A, Anderson EN, Nelson Iv CH, Ward C, Chauhan O, O'Brien C, Rajasundaram D, Rajan DS, Wirth B, Singh A, Pandey UB. SMN regulates GEMIN5 expression and acts as a modifier of GEMIN5-mediated neurodegeneration. Acta Neuropathol 2023; 146:477-498. [PMID: 37369805 PMCID: PMC11348892 DOI: 10.1007/s00401-023-02607-8] [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: 05/17/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/29/2023]
Abstract
GEMIN5 is essential for core assembly of small nuclear Ribonucleoproteins (snRNPs), the building blocks of spliceosome formation. Loss-of-function mutations in GEMIN5 lead to a neurodevelopmental syndrome among patients presenting with developmental delay, motor dysfunction, and cerebellar atrophy by perturbing SMN complex protein expression and assembly. Currently, molecular determinants of GEMIN5-mediated disease have yet to be explored. Here, we identified SMN as a genetic suppressor of GEMIN5-mediated neurodegeneration in vivo. We discovered that an increase in SMN expression by either SMN gene therapy replacement or the antisense oligonucleotide (ASO), Nusinersen, significantly upregulated the endogenous levels of GEMIN5 in mammalian cells and mutant GEMIN5-derived iPSC neurons. Further, we identified a strong functional association between the expression patterns of SMN and GEMIN5 in patient Spinal Muscular Atrophy (SMA)-derived motor neurons harboring loss-of-function mutations in the SMN gene. Interestingly, SMN binds to the C-terminus of GEMIN5 and requires the Tudor domain for GEMIN5 binding and expression regulation. Finally, we show that SMN upregulation ameliorates defective snRNP biogenesis and alternative splicing defects caused by loss of GEMIN5 in iPSC neurons and in vivo. Collectively, these studies indicate that SMN acts as a regulator of GEMIN5 expression and neuropathologies.
Collapse
Affiliation(s)
- Tyler R Fortuna
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sukhleen Kour
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Anixa Muiños-Bühl
- Institute of Human Genetics, Center for Molecular Medicine, Center for Rare Disorders, University of Cologne, Cologne, Germany
| | - Eric N Anderson
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Charlie H Nelson Iv
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Caroline Ward
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Om Chauhan
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Casey O'Brien
- Department of Pediatrics, Division of Health Informatics, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Division of Health Informatics, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Deepa S Rajan
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Medicine, Center for Rare Disorders, University of Cologne, Cologne, Germany
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH, USA
| | - Udai Bhan Pandey
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
34
|
Tuncel G, Sanlıdag B, Dirik E, Baris T, Ergoren MC, Temel SG. Lessons from Real Life Experience: Importance of In-House Sequencing and Smart Ratio-Based Real-Time PCR Outperform Multiplex Ligation-Dependent Probe Amplification in Prenatal Diagnosis for Spinal Muscular Atrophy: Bench to Bedside Diagnosis. Glob Med Genet 2023; 10:240-246. [PMID: 37663644 PMCID: PMC10471427 DOI: 10.1055/s-0043-1774307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a rare, recessively inherited neurodegenerative disorder caused by the presence of pathogenic variants in the SMN gene. As it is the leading inherited cause of infant mortality, identification of SMN gene pathogenic variant carriers is important for diagnostic purposes with effective genetic counseling. Multiple ligation probe analysis (MLPA), a probe-based method, is considered as the gold standard for SMA carrier analysis. However, MLPA might give false-negative results in cases with variations in the probe-binding regions. Here, we present a case born to consanguineous SMA carrier parents. Prenatal diagnosis with MLPA failed to detect the compound heterozygous mutant state of the proband and she was born unfortunately with SMA phenotype. Further analysis with a real-time polymerase chain reaction kit was able to detect the compound heterozygous state of the patient and was confirmed with targeted next-generation sequencing technology.
Collapse
Affiliation(s)
- Gulten Tuncel
- Department of Medical Genetics, Faculty of Medicine, Near East University, Nicosia, Cyprus
- DESAM Research Institute, Near East University, Nicosia, Cyprus
| | - Burcin Sanlıdag
- Department of Paediatrics, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Eray Dirik
- Department of Paediatrics, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Tugba Baris
- Gelişim Tıp Laboratuvarları, Istanbul, Turkey
| | - Mahmut Cerkez Ergoren
- Department of Medical Genetics, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Sehime Gulsun Temel
- Department of Medical Genetics, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
- Department of Translational Medicine, Institute of Health Science, Bursa Uludag University, Bursa, Turkey
- Department of Histology and Embryology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
35
|
Shell RD, McGrattan KE, Hurst-Davis R, Young SD, Baranello G, Lavrov A, O'Brien E, Wallach S, LaMarca N, Reyna SP, Darras BT. Onasemnogene abeparvovec preserves bulbar function in infants with presymptomatic spinal muscular atrophy: a post-hoc analysis of the SPR1NT trial. Neuromuscul Disord 2023; 33:670-676. [PMID: 37455203 DOI: 10.1016/j.nmd.2023.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
Bulbar function in spinal muscular atrophy has been defined as the ability to meet nutritional needs by mouth while maintaining airway protection and communicate verbally. The effects of disease-modifying treatment on bulbar function are not clear. A multidisciplinary team conducted post-hoc analyses of phase 3 SPR1NT trial data to evaluate bulbar function of infants at risk for spinal muscular atrophy who received one-time gene replacement therapy (onasemnogene abeparvovec) before symptom onset. Three endpoints represented adequate bulbar function in SPR1NT: (1) absence of physiologic swallowing impairment, (2) full oral nutrition, and (3) absence of adverse events indicating pulmonary instability. Communication was not assessed in SPR1NT. We descriptively assessed numbers/percentages of children who achieved each endpoint and all three collectively. SPR1NT included infants <6 postnatal weeks with two (n = 14) or three (n = 15) copies of the survival motor neuron 2 gene. At study end (18 [two-copy cohort] or 24 [three-copy cohort] months of age), 100% (29/29) of patients swallowed normally, achieved full oral nutrition, maintained pulmonary stability, and achieved the composite endpoint. When administered to infants before clinical symptom onset, onasemnogene abeparvovec allowed children at risk for spinal muscular atrophy to achieve milestones within published normal ranges of development and preserve bulbar function.
Collapse
Affiliation(s)
- Richard D Shell
- Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| | | | | | | | - Giovanni Baranello
- The Dubowitz Neuromuscular Centre, Developmental Neuroscience Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre & Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | | | | | | | | | | |
Collapse
|
36
|
Nishio H, Niba ETE, Saito T, Okamoto K, Takeshima Y, Awano H. Spinal Muscular Atrophy: The Past, Present, and Future of Diagnosis and Treatment. Int J Mol Sci 2023; 24:11939. [PMID: 37569314 PMCID: PMC10418635 DOI: 10.3390/ijms241511939] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a lower motor neuron disease with autosomal recessive inheritance. The first cases of SMA were reported by Werdnig in 1891. Although the phenotypic variation of SMA led to controversy regarding the clinical entity of the disease, the genetic homogeneity of SMA was proved in 1990. Five years later, in 1995, the gene responsible for SMA, SMN1, was identified. Genetic testing of SMN1 has enabled precise epidemiological studies, revealing that SMA occurs in 1 of 10,000 to 20,000 live births and that more than 95% of affected patients are homozygous for SMN1 deletion. In 2016, nusinersen was the first drug approved for treatment of SMA in the United States. Two other drugs were subsequently approved: onasemnogene abeparvovec and risdiplam. Clinical trials with these drugs targeting patients with pre-symptomatic SMA (those who were diagnosed by genetic testing but showed no symptoms) revealed that such patients could achieve the milestones of independent sitting and/or walking. Following the great success of these trials, population-based newborn screening programs for SMA (more precisely, SMN1-deleted SMA) have been increasingly implemented worldwide. Early detection by newborn screening and early treatment with new drugs are expected to soon become the standards in the field of SMA.
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, Juntendo University Graduate School of Medicine, 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;
| | - Yasuhiro Takeshima
- Department of Pediatrics, Hyogo Medical University, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan;
| | - Hiroyuki Awano
- Organization for Research Initiative and Promotion, Research Initiative Center, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan;
| |
Collapse
|
37
|
Davidson JE, Russell JS, Martinez NN, Mowat DR, Jones KJ, Kirk EP, Kariyawasam D, Farrar M, D’Silva A. The Carrier Frequency of Two SMN1 Genes in Parents of Symptomatic Children with SMA and the Significance of SMN1 Exon 8 in Carriers. Genes (Basel) 2023; 14:1403. [PMID: 37510307 PMCID: PMC10379112 DOI: 10.3390/genes14071403] [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: 05/24/2023] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Current carrier screening methods do not identify a proportion of carriers that may have children affected by spinal muscular atrophy (SMA). Additional genetic data is essential to inform accurate risk assessment and genetic counselling of SMA carriers. This study aims to quantify the various genotypes among parents of children with SMA. METHOD A retrospective cohort study was undertaken at Sydney Children's Hospital Network, the major SMA referral centre for New South Wales, Australia. Participants included children with genetically confirmed SMA born between 2005 and 2021. Data was collected on parent genotype inclusive of copy number of SMN1 exons 7 and 8. The number of SMN2 exon 7 copies were recorded for the affected children. Descriptive statistics were used to determine the proportion of carriers of 2+0 genotype classified as silent carriers. Chi-square test was used to correlate the association between parents with a heterozygous SMN1 exon 7 deletion and two copies of exon 8 and ≥3 SMN2 copy number in the proband. RESULTS SMA carrier testing was performed in 118/154 (76.6%) parents, incorporating 59 probands with homozygous SMN1 deletions and one proband with compound heterozygote pathogenic variants. Among parents with a child with SMA, 7.6% had two copies of SMN1 exon 7. When only probands with a homozygous SMN1 exon 7 deletion were included, 6.9% of parents had two copies of SMN1 exon 7. An association was observed between heterozygous deletion of SMN1 exon 7 with two copies of exon 8 in a parent and ≥3 SMN2 copy number in the affected proband (p = 0.07). CONCLUSIONS This study confirmed a small but substantial proportion of silent carriers not identified by conventional screening within an Australian context. Accordingly, the effectiveness of carrier screening for SMA is linked with genetic counselling to enable health literacy regarding high and low risk results and is complemented by new-born screening and maintaining clinical awareness for SMA. Gene conversion events may underpin the associations between parent carrier status and proband SMN2 copy number.
Collapse
Affiliation(s)
- Joanne E Davidson
- Department of Neurology, Sydney Children’s Hospitals Network, Sydney, NSW 2031, Australia; (J.E.D.)
| | - Jacqueline S Russell
- Centre for Clinical Genetics, Sydney Children’s Hospital, Randwick, Sydney, NSW 2031, Australia; (J.S.R.)
| | - Noelia Nunez Martinez
- Centre for Clinical Genetics, Sydney Children’s Hospital, Randwick, Sydney, NSW 2031, Australia; (J.S.R.)
| | - David R Mowat
- Centre for Clinical Genetics, Sydney Children’s Hospital, Randwick, Sydney, NSW 2031, Australia; (J.S.R.)
| | - Kristi J Jones
- Department of Clinical Genetics, The Children’s Hospital at Westmead, and Discipline of Paediatrics and Child Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Edwin P Kirk
- Centre for Clinical Genetics, Sydney Children’s Hospital, Randwick, Sydney, NSW 2031, Australia; (J.S.R.)
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, NSW 2052, Australia
| | - Didu Kariyawasam
- Department of Neurology, Sydney Children’s Hospitals Network, Sydney, NSW 2031, Australia; (J.E.D.)
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, NSW 2052, Australia
| | - Michelle Farrar
- Department of Neurology, Sydney Children’s Hospitals Network, Sydney, NSW 2031, Australia; (J.E.D.)
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, NSW 2052, Australia
| | - Arlene D’Silva
- Department of Neurology, Sydney Children’s Hospitals Network, Sydney, NSW 2031, Australia; (J.E.D.)
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, University of New South Wales Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
38
|
Balaji L, Farrar MA, D'Silva AM, Kariyawasam DS. Decision-making and challenges within the evolving treatment algorithm in spinal muscular atrophy: a clinical perspective. Expert Rev Neurother 2023; 23:571-586. [PMID: 37227306 DOI: 10.1080/14737175.2023.2218549] [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: 02/07/2023] [Accepted: 05/23/2023] [Indexed: 05/26/2023]
Abstract
INTRODUCTION The clinical application of disease modifying therapies has dramatically changed the paradigm of the management of people with spinal muscular atrophy (SMA), from sole reliance on symptomatic care directed toward the downstream consequences of muscle weakness, to proactive intervention and even preventative care. AREAS COVERED In this perspective, the authors evaluate the contemporary therapeutic landscape of SMA and discuss the evolution of novel phenotypes and the treatment algorithm, including the key factors that define individual treatment choice and treatment response. The benefits achieved by early diagnosis and treatment through newborn screening are highlighted, alongside an appraisal of emerging prognostic methods and classification frameworks to inform clinicians, patients, and families about disease course, manage expectations, and improve care planning. A future perspective of unmet needs and challenges is provided, emphasizing the key role of research. EXPERT OPINION SMN-augmenting therapies have improved health outcomes for people with SMA and powered the practice of personalized medicine. Within this new proactive diagnostic and treatment paradigm, new phenotypes and different disease trajectories are emerging. Ongoing collaborative research efforts to understand the biology of SMA and define optimal response are critical to refining future approaches.
Collapse
Affiliation(s)
- Lakshmi Balaji
- Department of Neurology, Sydney Children's Hospital Network, Sydney, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health UNSW, Sydney, Australia
| | - Michelle A Farrar
- Department of Neurology, Sydney Children's Hospital Network, Sydney, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health UNSW, Sydney, Australia
- UNSW Kensington Campus, Sydney, Australia
| | - Arlene M D'Silva
- Department of Neurology, Sydney Children's Hospital Network, Sydney, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health UNSW, Sydney, Australia
- UNSW Kensington Campus, Sydney, Australia
| | - Didu S Kariyawasam
- Department of Neurology, Sydney Children's Hospital Network, Sydney, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health UNSW, Sydney, Australia
| |
Collapse
|
39
|
Yang XC, Desotell A, Lin MH, Paige AS, Malinowska A, Sun Y, Aik WS, Dadlez M, Tong L, Dominski Z. In vitro methylation of the U7 snRNP subunits Lsm11 and SmE by the PRMT5/MEP50/pICln methylosome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540203. [PMID: 37215023 PMCID: PMC10197641 DOI: 10.1101/2023.05.10.540203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
U7 snRNP is a multi-subunit endonuclease required for 3' end processing of metazoan replication-dependent histone pre-mRNAs. In contrast to the spliceosomal snRNPs, U7 snRNP lacks the Sm subunits D1 and D2 and instead contains two related proteins, Lsm10 and Lsm11. The remaining five subunits of the U7 heptameric Sm ring, SmE, F, G, B and D3, are shared with the spliceosomal snRNPs. The pathway that assembles the unique ring of U7 snRNP is unknown. Here, we show that a heterodimer of Lsm10 and Lsm11 tightly interacts with the methylosome, a complex of the arginine methyltransferase PRMT5, MEP50 and pICln known to methylate arginines in the C-terminal regions of the Sm proteins B, D1 and D3 during the spliceosomal Sm ring assembly. Both biochemical and Cryo-EM structural studies demonstrate that the interaction is mediated by PRMT5, which binds and methylates two arginine residues in the N-terminal region of Lsm11. Surprisingly, PRMT5 also methylates an N-terminal arginine in SmE, a subunit that does not undergo this type of modification during the biogenesis of the spliceosomal snRNPs. An intriguing possibility is that the unique methylation pattern of Lsm11 and SmE plays a vital role in the assembly of the U7 snRNP.
Collapse
|
40
|
Kataoka M, Sahashi K, Tsujikawa K, Takeda JI, Hirunagi T, Iida M, Katsunoa M. Dysregulation of Aldh1a2 underlies motor neuron degeneration in spinal muscular atrophy. Neurosci Res 2023:S0168-0102(23)00090-1. [PMID: 37146794 DOI: 10.1016/j.neures.2023.04.007] [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: 01/29/2023] [Revised: 04/22/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023]
Abstract
Lower motor neuron degeneration is the pathological hallmark of spinal muscular atrophy (SMA), a hereditary motor neuron disease caused by loss of the SMN1 gene and the resulting deficiency of ubiquitously expressed SMN protein. The molecular mechanisms underlying motor neuron degeneration, however, remain elusive. To clarify the cell-autonomous defect in developmental processes, we here performed transcriptome analyses of isolated embryonic motor neurons of SMA model mice to explore mechanisms of dysregulation of cell-type-specific gene expression. Of 12 identified genes that were differentially expressed between the SMA and control motor neurons, we focused on Aldh1a2, an essential gene for lower motor neuron development. In primary spinal motor neuron cultures, knockdown of Aldh1a2 led to the formation of axonal spheroids and neurodegeneration, reminiscent of the histopathological changes observed in human and animal cellular models. Conversely, Aldh1a2 rescued these pathological features in spinal motor neurons derived from SMA mouse embryos. Our findings suggest that developmental defects due to Aldh1a2 dysregulation enhances lower motor neuron vulnerability in SMA.
Collapse
Affiliation(s)
- Mayumi Kataoka
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Kentaro Sahashi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan.
| | - Koyo Tsujikawa
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Jun-Ichi Takeda
- Division of Neurogenetics, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Tomoki Hirunagi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Madoka Iida
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Masahisa Katsunoa
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan; Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan.
| |
Collapse
|
41
|
Shin HJ, Na JH, Lee H, Lee YM. Nusinersen for spinal muscular atrophy types II and III: a retrospective single-center study in South Korea. World J Pediatr 2023; 19:450-459. [PMID: 36441395 DOI: 10.1007/s12519-022-00638-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/11/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND This study investigated the efficacy and safety of nusinersen, an antisense oligonucleotide, in patients with spinal muscular atrophy (SMA) types II (OMIM: 253,550) or III (OMIM: 253,400), including those with severe scoliosis or requiring respiratory support via mechanical ventilation. METHODS Data from 40 patients with genetically confirmed SMA who were treated with nusinersen at our institute from March 2019 to April 2022 were retrospectively analyzed. Of these, 30 patients with an age of onset < 3 years and not on permanent ventilation were selected. Clinical and genetic characteristics were investigated, and motor function was evaluated based on the Hammersmith Functional Motor Scale-Expanded (HFMSE) score. RESULTS The mean age of symptom onset was 1.2 years. Most patients were diagnosed with SMA type II (27/30, 90%). Nusinersen was administered via computed tomography-guided or direct intrathecal injection in 87% (26/30) and 13% (4/30) of the patients, respectively. At the 6-, 14-, 22-, and 26-month follow-ups, 72%, 71%, 88%, and 86% of patients showed motor improvement, respectively, with mean changes in HFMSE scores of 2.10, 2.88, 4.21, and 5.29, respectively. Multivariable analysis showed that the use of noninvasive ventilation was associated with poorer outcomes of motor function. CONCLUSIONS Patients with SMA type II or III who received nusinersen treatment showed significant improvement in motor function. A longer treatment duration led to a higher number of patients with improved motor function. No significant side effects of nusinersen were observed. Patients with SMA, even those with severe scoliosis or on respiratory support, can be safely treated using nusinersen.
Collapse
Affiliation(s)
- Hui Jin Shin
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 135-720, Korea
| | - Ji-Hoon Na
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 135-720, Korea
| | - Hyunjoo Lee
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 135-720, Korea
| | - Young-Mock Lee
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, 135-720, Korea.
| |
Collapse
|
42
|
Ros LAA, Goedee HS, Franssen H, Asselman FL, Bartels B, Cuppen I, van Eijk RPA, Sleutjes BTHM, van der Pol WL, Wadman RI. Longitudinal prospective cohort study to assess peripheral motor function with extensive electrophysiological techniques in patients with Spinal Muscular Atrophy (SMA): the SMA Motor Map protocol. BMC Neurol 2023; 23:164. [PMID: 37095427 PMCID: PMC10124000 DOI: 10.1186/s12883-023-03207-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/14/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Hereditary spinal muscular atrophy (SMA) is a motor neuron disorder with a wide range in severity in children and adults. Two therapies that alter splicing of the Survival Motor Neuron 2 (SMN2) gene, i.e. nusinersen and risdiplam, improve motor function in SMA, but treatment effects vary. Experimental studies indicate that motor unit dysfunction encompasses multiple features, including abnormal function of the motor neuron, axon, neuromuscular junction and muscle fibres. The relative contributions of dysfunction of different parts of the motor unit to the clinical phenotype are unknown. Predictive biomarkers for clinical efficacy are currently lacking. The goals of this project are to study the association of electrophysiological abnormalities of the peripheral motor system in relation to 1) SMA clinical phenotypes and 2) treatment response in patients treated with SMN2-splicing modifiers (nusinersen or risdiplam). METHODS We designed an investigator-initiated, monocentre, longitudinal cohort study using electrophysiological techniques ('the SMA Motor Map') in Dutch children (≥ 12 years) and adults with SMA types 1-4. The protocol includes the compound muscle action potential scan, nerve excitability testing and repetitive nerve stimulation test, executed unilaterally at the median nerve. Part one cross-sectionally assesses the association of electrophysiological abnormalities in relation to SMA clinical phenotypes in treatment-naïve patients. Part two investigates the predictive value of electrophysiological changes at two-months treatment for a positive clinical motor response after one-year treatment with SMN2-splicing modifiers. We will include 100 patients in each part of the study. DISCUSSION This study will provide important information on the pathophysiology of the peripheral motor system of treatment-naïve patients with SMA through electrophysiological techniques. More importantly, the longitudinal analysis in patients on SMN2-splicing modifying therapies (i.e. nusinersen and risdiplam) intents to develop non-invasive electrophysiological biomarkers for treatment response in order to improve (individualized) treatment decisions. TRIAL REGISTRATION NL72562.041.20 (registered at https://www.toetsingonline.nl . 26-03-2020).
Collapse
Affiliation(s)
- Leandra A A Ros
- Department of Neurology & Neurosurgery, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - H Stephan Goedee
- Department of Neurology & Neurosurgery, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Hessel Franssen
- Department of Neurology & Neurosurgery, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Fay-Lynn Asselman
- Department of Neurology & Neurosurgery, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Bart Bartels
- Child Development and Exercise Centre, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Inge Cuppen
- Department of Neurology & Neurosurgery, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Ruben P A van Eijk
- Department of Neurology & Neurosurgery, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
- Biostatistics and Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Boudewijn T H M Sleutjes
- Department of Neurology & Neurosurgery, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - W Ludo van der Pol
- Department of Neurology & Neurosurgery, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - Renske I Wadman
- Department of Neurology & Neurosurgery, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands.
| |
Collapse
|
43
|
Tejero R, Alsakkal M, Hennlein L, Lopez-Cabello AM, Jablonka S, Tabares L. Nifedipine Ameliorates Cellular Differentiation Defects of Smn-Deficient Motor Neurons and Enhances Neuromuscular Transmission in SMA Mice. Int J Mol Sci 2023; 24:ijms24087648. [PMID: 37108811 PMCID: PMC10146780 DOI: 10.3390/ijms24087648] [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: 03/14/2023] [Revised: 04/11/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
In spinal muscular atrophy (SMA), mutations in or loss of the Survival Motor Neuron 1 (SMN1) gene reduce full-length SMN protein levels, which leads to the degeneration of a percentage of motor neurons. In mouse models of SMA, the development and maintenance of spinal motor neurons and the neuromuscular junction (NMJ) function are altered. Since nifedipine is known to be neuroprotective and increases neurotransmission in nerve terminals, we investigated its effects on cultured spinal cord motor neurons and motor nerve terminals of control and SMA mice. We found that application of nifedipine increased the frequency of spontaneous Ca2+ transients, growth cone size, cluster-like formations of Cav2.2 channels, and it normalized axon extension in SMA neurons in culture. At the NMJ, nifedipine significantly increased evoked and spontaneous release at low-frequency stimulation in both genotypes. High-strength stimulation revealed that nifedipine increased the size of the readily releasable pool (RRP) of vesicles in control but not SMA mice. These findings provide experimental evidence about the ability of nifedipine to prevent the appearance of developmental defects in SMA embryonic motor neurons in culture and reveal to which extent nifedipine could still increase neurotransmission at the NMJ in SMA mice under different functional demands.
Collapse
Affiliation(s)
- Rocio Tejero
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Mohammad Alsakkal
- Institute of Clinical Neurobiology, University Hospital Würzburg, 97078 Würzburg, Germany
| | - Luisa Hennlein
- Institute of Clinical Neurobiology, University Hospital Würzburg, 97078 Würzburg, Germany
| | - Ana M Lopez-Cabello
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Sibylle Jablonka
- Institute of Clinical Neurobiology, University Hospital Würzburg, 97078 Würzburg, Germany
| | - Lucia Tabares
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, 41009 Seville, Spain
| |
Collapse
|
44
|
Ishibashi Y, Sung CYW, Grati M, Chien W. Immune responses in the mammalian inner ear and their implications for AAV-mediated inner ear gene therapy. Hear Res 2023; 432:108735. [PMID: 36965335 DOI: 10.1016/j.heares.2023.108735] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 02/17/2023] [Accepted: 03/04/2023] [Indexed: 03/13/2023]
Abstract
Adeno-associated virus (AAV)-mediated inner ear gene therapy is a promising treatment option for hearing loss and dizziness. Several studies have shown that AAV-mediated inner ear gene therapy can be applied to various mouse models of hereditary hearing loss to improve their auditory function. Despite the increase in AAV-based animal and clinical studies aiming to rescue auditory and vestibular functions, little is currently known about the host immune responses to AAV in the mammalian inner ear. It has been reported that the host immune response plays an important role in the safety and efficacy of viral-mediated gene therapy. Therefore, in order for AAV-mediated gene therapy to be successfully and safely translated into patients with hearing loss and dizziness, a better understanding of the host immune responses to AAV in the inner ear is critical. In this review, we summarize the current knowledge on host immune responses to AAV-mediated gene therapy in the mammalian inner ear and other organ systems. We also outline the areas of research that are critical for ensuring the safety and efficacy of AAV-mediated inner ear gene therapy in future clinical and translational studies.
Collapse
Affiliation(s)
- Yasuko Ishibashi
- Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, 35A 1F220, 35A Covent Dr., Bethesda, MD 20892, USA; Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, USA
| | - Cathy Yea Won Sung
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, MD, USA
| | - Mhamed Grati
- Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, 35A 1F220, 35A Covent Dr., Bethesda, MD 20892, USA
| | - Wade Chien
- Inner Ear Gene Therapy Program, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, 35A 1F220, 35A Covent Dr., Bethesda, MD 20892, USA; Department of Otolaryngology-Head & Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
45
|
Ikenaka A, Kitagawa Y, Yoshida M, Lin CY, Niwa A, Nakahata T, Saito MK. SMN promotes mitochondrial metabolic maturation during myogenesis by regulating the MYOD-miRNA axis. Life Sci Alliance 2023; 6:e202201457. [PMID: 36604149 PMCID: PMC9834662 DOI: 10.26508/lsa.202201457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 01/07/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a congenital neuromuscular disease caused by the mutation or deletion of the survival motor neuron 1 (SMN1) gene. Although the primary cause of progressive muscle atrophy in SMA has classically been considered the degeneration of motor neurons, recent studies have indicated a skeletal muscle-specific pathological phenotype such as impaired mitochondrial function and enhanced cell death. Here, we found that the down-regulation of SMN causes mitochondrial dysfunction and subsequent cell death in in vitro models of skeletal myogenesis with both a murine C2C12 cell line and human induced pluripotent stem cells. During myogenesis, SMN binds to the upstream genomic regions of MYOD1 and microRNA (miR)-1 and miR-206. Accordingly, the loss of SMN down-regulates these miRs, whereas supplementation of the miRs recovers the mitochondrial function, cell survival, and myotube formation of SMN-deficient C2C12, indicating the SMN-miR axis is essential for myogenic metabolic maturation. In addition, the introduction of the miRs into ex vivo muscle stem cells derived from Δ7-SMA mice caused myotube formation and muscle contraction. In conclusion, our data revealed novel transcriptional roles of SMN during myogenesis, providing an alternative muscle-oriented therapeutic strategy for SMA patients.
Collapse
Affiliation(s)
- Akihiro Ikenaka
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yohko Kitagawa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Michiko Yoshida
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chuang-Yu Lin
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Akira Niwa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Tatsutoshi Nakahata
- Drug Discovery Technology Development Office, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Megumu K Saito
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| |
Collapse
|
46
|
Riboldi GM, Faravelli I, Rinchetti P, Lotti F. SMN post-translational modifications in spinal muscular atrophy. Front Cell Neurosci 2023; 17:1092488. [PMID: 36874214 PMCID: PMC9981653 DOI: 10.3389/fncel.2023.1092488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/26/2023] [Indexed: 02/19/2023] Open
Abstract
Since its first identification as the gene responsible for spinal muscular atrophy (SMA), the range of survival motor neuron (SMN) protein functions has increasingly expanded. This multimeric complex plays a crucial role in a variety of RNA processing pathways. While its most characterized function is in the biogenesis of ribonucleoproteins, several studies have highlighted the SMN complex as an important contributor to mRNA trafficking and translation, axonal transport, endocytosis, and mitochondria metabolism. All these multiple functions need to be selectively and finely modulated to maintain cellular homeostasis. SMN has distinct functional domains that play a crucial role in complex stability, function, and subcellular distribution. Many different processes were reported as modulators of the SMN complex activities, although their contribution to SMN biology still needs to be elucidated. Recent evidence has identified post-translational modifications (PTMs) as a way to regulate the pleiotropic functions of the SMN complex. These modifications include phosphorylation, methylation, ubiquitination, acetylation, sumoylation, and many other types. PTMs can broaden the range of protein functions by binding chemical moieties to specific amino acids, thus modulating several cellular processes. Here, we provide an overview of the main PTMs involved in the regulation of the SMN complex with a major focus on the functions that have been linked to SMA pathogenesis.
Collapse
Affiliation(s)
| | | | | | - Francesco Lotti
- Center for Motor Neuron Biology and Diseases, Departments of Pathology & Cell Biology, and Neurology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
47
|
Faravelli I, Riboldi GM, Rinchetti P, Lotti F. The SMN Complex at the Crossroad between RNA Metabolism and Neurodegeneration. Int J Mol Sci 2023; 24:2247. [PMID: 36768569 PMCID: PMC9917330 DOI: 10.3390/ijms24032247] [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: 12/03/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
In the cell, RNA exists and functions in a complex with RNA binding proteins (RBPs) that regulate each step of the RNA life cycle from transcription to degradation. Central to this regulation is the role of several molecular chaperones that ensure the correct interactions between RNA and proteins, while aiding the biogenesis of large RNA-protein complexes (ribonucleoproteins or RNPs). Accurate formation of RNPs is fundamentally important to cellular development and function, and its impairment often leads to disease. The survival motor neuron (SMN) protein exemplifies this biological paradigm. SMN is part of a multi-protein complex essential for the biogenesis of various RNPs that function in RNA metabolism. Mutations leading to SMN deficiency cause the neurodegenerative disease spinal muscular atrophy (SMA). A fundamental question in SMA biology is how selective motor system dysfunction results from reduced levels of the ubiquitously expressed SMN protein. Recent clarification of the central role of the SMN complex in RNA metabolism and a thorough characterization of animal models of SMA have significantly advanced our knowledge of the molecular basis of the disease. Here we review the expanding role of SMN in the regulation of gene expression through its multiple functions in RNP biogenesis. We discuss developments in our understanding of SMN activity as a molecular chaperone of RNPs and how disruption of SMN-dependent RNA pathways can contribute to the SMA phenotype.
Collapse
Affiliation(s)
- Irene Faravelli
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Center for Motor Neuron Biology and Diseases, Departments of Pathology & Cell Biology, and Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Giulietta M. Riboldi
- Center for Motor Neuron Biology and Diseases, Departments of Pathology & Cell Biology, and Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- The Marlene and Paolo Fresco Institute for Parkinson’s and Movement Disorders, NYU Langone Health, New York, NY 10017, USA
| | - Paola Rinchetti
- Center for Motor Neuron Biology and Diseases, Departments of Pathology & Cell Biology, and Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Francesco Lotti
- Center for Motor Neuron Biology and Diseases, Departments of Pathology & Cell Biology, and Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
48
|
Lejman J, Panuciak K, Nowicka E, Mastalerczyk A, Wojciechowska K, Lejman M. Gene Therapy in ALS and SMA: Advances, Challenges and Perspectives. Int J Mol Sci 2023; 24:ijms24021130. [PMID: 36674643 PMCID: PMC9860634 DOI: 10.3390/ijms24021130] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/31/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Gene therapy is defined as the administration of genetic material to modify, manipulate gene expression or alter the properties of living cells for therapeutic purposes. Recent advances and improvements in this field have led to many breakthroughs in the treatment of various diseases. As a result, there has been an increasing interest in the use of these therapies to treat motor neuron diseases (MNDs), for which many potential molecular targets have been discovered. MNDs are neurodegenerative disorders that, in their most severe forms, can lead to respiratory failure and death, for instance, spinal muscular atrophy (SMA) or amyotrophic lateral sclerosis (ALS). Despite the fact that SMA has been known for many years, it is still one of the most common genetic diseases causing infant mortality. The introduction of drugs based on ASOs-nusinersen; small molecules-risdiplam; and replacement therapy (GRT)-Zolgensma has shown a significant improvement in both event-free survival and the quality of life of patients after using these therapies in the available trial results. Although there is still no drug that would effectively alleviate the course of the disease in ALS, the experience gained from SMA gene therapy gives hope for a positive outcome of the efforts to produce an effective and safe drug. The aim of this review is to present current progress and prospects for the use of gene therapy in the treatment of both SMA and ALS.
Collapse
Affiliation(s)
- Jan Lejman
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence:
| | - Kinga Panuciak
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Emilia Nowicka
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Angelika Mastalerczyk
- Student Scientific Society, Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Katarzyna Wojciechowska
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
49
|
Younger DS. Spinal cord motor disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:3-42. [PMID: 37620076 DOI: 10.1016/b978-0-323-98817-9.00007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Spinal cord diseases are frequently devastating due to the precipitous and often permanently debilitating nature of the deficits. Spastic or flaccid paraparesis accompanied by dermatomal and myotomal signatures complementary to the incurred deficits facilitates localization of the insult within the cord. However, laboratory studies often employing disease-specific serology, neuroradiology, neurophysiology, and cerebrospinal fluid analysis aid in the etiologic diagnosis. While many spinal cord diseases are reversible and treatable, especially when recognized early, more than ever, neuroscientists are being called to investigate endogenous mechanisms of neural plasticity. This chapter is a review of the embryology, neuroanatomy, clinical localization, evaluation, and management of adult and childhood spinal cord motor disorders.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
50
|
A link between agrin signalling and Ca v3.2 at the neuromuscular junction in spinal muscular atrophy. Sci Rep 2022; 12:18960. [PMID: 36347955 PMCID: PMC9643518 DOI: 10.1038/s41598-022-23703-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
SMN protein deficiency causes motoneuron disease spinal muscular atrophy (SMA). SMN-based therapies improve patient motor symptoms to variable degrees. An early hallmark of SMA is the perturbation of the neuromuscular junction (NMJ), a synapse between a motoneuron and muscle cell. NMJ formation depends on acetylcholine receptor (AChR) clustering triggered by agrin and its co-receptors lipoprotein receptor-related protein 4 (LRP4) and transmembrane muscle-specific kinase (MuSK) signalling pathway. We have previously shown that flunarizine improves NMJs in SMA model mice, but the mechanisms remain elusive. We show here that flunarizine promotes AChR clustering in cell-autonomous, dose- and agrin-dependent manners in C2C12 myotubes. This is associated with an increase in protein levels of LRP4, integrin-beta-1 and alpha-dystroglycan, three agrin co-receptors. Furthermore, flunarizine enhances MuSK interaction with integrin-beta-1 and phosphotyrosines. Moreover, the drug acts on the expression and splicing of Agrn and Cacna1h genes in a muscle-specific manner. We reveal that the Cacna1h encoded protein Cav3.2 closely associates in vitro with the agrin co-receptor LRP4. In vivo, it is enriched nearby NMJs during neonatal development and the drug increases this immunolabelling in SMA muscles. Thus, flunarizine modulates key players of the NMJ and identifies Cav3.2 as a new protein involved in the NMJ biology.
Collapse
|