1
|
Berzal-Serrano A, García-Bohórquez B, Aller E, Jaijo T, Pitarch-Castellano I, Rausell D, García-García G, Millán JM. Outcomes of a Pilot Newborn Screening Program for Spinal Muscular Atrophy in the Valencian Community. Int J Neonatal Screen 2025; 11:7. [PMID: 39846593 PMCID: PMC11755645 DOI: 10.3390/ijns11010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/24/2025] Open
Abstract
Spinal muscular atrophy (SMA) is a degenerative neuromuscular condition resulting from a homozygous deletion of the survival motor neuron 1 (SMN1) gene in 95% of patients. A timely diagnosis via newborn screening (NBS) and initiating treatment before the onset of symptoms are critical for improving health outcomes in affected individuals. We carried out a screening test by quantitative PCR (qPCR) to amplify the exon seven of SMN1 using dried blood spot (DBS) samples. From October 2021 to August 2024, a total of 31,560 samples were tested in the Valencian Community (Spain) and 4 of them were positive for SMA, indicating an incidence of 1/7890. Genetic confirmation was performed using multiplex ligation-dependent probe amplification (MLPA) and AmplideX PCR/CE SMN1/2 Plus kit, in parallel obtaining concordant results in survival motor neuron 2 (SMN2) gene copy number. Within the first few weeks of their lives, two of the four patients detected by NBS showed signs of severe hypotonia, becoming ineligible for treatment. The other two patients were the first presymptomatic patients with two copies of SMN2 to receive treatment with Risdiplam in Spain. In order to treat positive cases in their early stages, we conclude that the official deployment of SMA newborn screening is necessary.
Collapse
Affiliation(s)
- Alba Berzal-Serrano
- Cellular, Molecular and Genomics Biomedicine Group, La Fe Health Research Institute, 46026 Valencia, Spain; (A.B.-S.); (B.G.-B.); (E.A.); (T.J.)
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
| | - Belén García-Bohórquez
- Cellular, Molecular and Genomics Biomedicine Group, La Fe Health Research Institute, 46026 Valencia, Spain; (A.B.-S.); (B.G.-B.); (E.A.); (T.J.)
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Carlos III Health Institute, 28029 Madrid, Spain
| | - Elena Aller
- Cellular, Molecular and Genomics Biomedicine Group, La Fe Health Research Institute, 46026 Valencia, Spain; (A.B.-S.); (B.G.-B.); (E.A.); (T.J.)
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Genetics, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain
| | - Teresa Jaijo
- Cellular, Molecular and Genomics Biomedicine Group, La Fe Health Research Institute, 46026 Valencia, Spain; (A.B.-S.); (B.G.-B.); (E.A.); (T.J.)
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Genetics, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain
| | | | - Dolores Rausell
- Clinical Analysis Service, Metabolic Disorders Laboratory, La Fe University and Polytechnic Hospital, 46026 Valencia, Spain;
| | - Gema García-García
- Cellular, Molecular and Genomics Biomedicine Group, La Fe Health Research Institute, 46026 Valencia, Spain; (A.B.-S.); (B.G.-B.); (E.A.); (T.J.)
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Carlos III Health Institute, 28029 Madrid, Spain
| | - José M. Millán
- Cellular, Molecular and Genomics Biomedicine Group, La Fe Health Research Institute, 46026 Valencia, Spain; (A.B.-S.); (B.G.-B.); (E.A.); (T.J.)
- Joint Unit CIPF-IIS La Fe Molecular, Cellular and Genomic Biomedicine, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
2
|
Tizzano EF, Quijano-Roy S, Servais L, Parsons JA, Aharoni S, Lakhotia A, Finkel RS. Outcomes for patients in the RESTORE registry with spinal muscular atrophy and four or more SMN2 gene copies treated with onasemnogene abeparvovec. Eur J Paediatr Neurol 2024; 53:18-24. [PMID: 39260228 DOI: 10.1016/j.ejpn.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024]
Abstract
OBJECTIVE We describe outcomes following onasemnogene abeparvovec monotherapy for patients with ≥four survival motor neuron 2 (SMN2) gene copies in RESTORE, a noninterventional spinal muscular atrophy patient registry. METHODS We evaluated baseline characteristics, motor milestone achievement, post-treatment motor function, use of ventilatory/nutritional support, and adverse events as of December 22, 2022. RESULTS At data cutoff, 19 patients in RESTORE had ≥four SMN2 copies and were treated with onasemnogene abeparvovec monotherapy (n=12 [63.2%] four copies; n=7 [36.8%] >four copies). All patients were identified by newborn screening and were reported as asymptomatic at diagnosis. Median age at onasemnogene abeparvovec administration was 3.0 months. Median time from treatment to last recorded visit was 15.4 months, with a range of post-treatment follow-up of 0.03-39.4 months. All 12 children who were assessed for motor development achieved new milestones, including standing alone (n=2) and walking alone (n=5). Five children reported one or more treatment-emergent adverse events (one Grade 3 or greater). No deaths or use of ventilatory/nutritional support were reported. CONCLUSIONS Real-world findings from the RESTORE registry indicate that patients with ≥four SMN2 gene copies treated with onasemnogene abeparvovec monotherapy demonstrated improvements in motor function. Adverse events experienced by these patients were consistent with previously reported findings.
Collapse
Affiliation(s)
- Eduardo F Tizzano
- Department of Clinical and Molecular Genetics, Hospital Vall d'Hebron, Passeig de la Vall d'Hebron, 119-129, Horta-Guinardó, 08035, Barcelona, Spain.
| | - Susana Quijano-Roy
- Garches Neuromuscular Reference Center, APHP Raymond Poincaré University Hospital (UVSQ Paris Saclay), 104 Bd Raymond Poincaré, 92380, Garches, France
| | - Laurent Servais
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre, & NIHR Oxford Biomedical Research, University of Oxford, Headly Way, Headington, OX3 9DU, Oxford, UK; Department of Pediatrics, Neuromuscular Reference Center, University and University Hospital of Liège, Bât. B35 Département des Sciences Cliniques, Quartier Hôpital, Avenue de l'Hôpital 13, 4000, Liège, Belgium
| | - Julie A Parsons
- Children's Hospital Colorado, University of Colorado School of Medicine, 13001 East 17th Place, Aurora, CO, 80045, USA
| | - Sharon Aharoni
- Institute of Pediatric Neurology, Schneider Children's Medical Center of Israel, Kaplan St 14, Petah Tikva, Israel; Faculty of Medical and Health Sciences, Tel-Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Arpita Lakhotia
- University of Louisville, Norton Children's Medical Group, 411 East Chestnut Street, Floor 6, Louisville, KY, 40202, USA
| | - Richard S Finkel
- Center for Experimental Neurotherapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| |
Collapse
|
3
|
Maretina M, Koroleva V, Shchugareva L, Glotov A, Kiselev A. The Relevance of Spinal Muscular Atrophy Biomarkers in the Treatment Era. Biomedicines 2024; 12:2486. [PMID: 39595052 PMCID: PMC11591959 DOI: 10.3390/biomedicines12112486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a severe neuromuscular disorder that currently has an approved treatment for all forms of the disease. Previously, biomarkers were primarily used for diagnostic purposes, such as detecting the presence of the disease or determining a specific clinical type of SMA. Currently, with the availability of therapy, biomarkers have become more valuable due to their potential for prognostic, predictive, and pharmacodynamic applications. This review describes the most promising physiological, functional, imaging and molecular biomarkers for SMA, derived from different patients' tissues. The review summarizes information about classical biomarkers that are already used in clinical practice as well as fresh findings on promising biomarkers that have been recently disclosed. It highlights the usefulness, limitations, and strengths of each potential biomarker, indicating the purposes for which each is best suited and when combining them may be most beneficial.
Collapse
Affiliation(s)
- Marianna Maretina
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia; (M.M.); (A.G.)
| | - Valeria Koroleva
- Municipal Hospital for Children No. 1, 198205 Saint-Petersburg, Russia; (V.K.); (L.S.)
| | - Lyudmila Shchugareva
- Municipal Hospital for Children No. 1, 198205 Saint-Petersburg, Russia; (V.K.); (L.S.)
- Department of Pediatric Neuropathology and Neurosurgery, North-Western State Medical University Named After I.I. Mechnikov, 191015 Saint-Petersburg, Russia
| | - Andrey Glotov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia; (M.M.); (A.G.)
| | - Anton Kiselev
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia; (M.M.); (A.G.)
| |
Collapse
|
4
|
Palma S, Pereira P. Subclinical spinal muscular atrophy in a 60-year-old man. Neuromuscul Disord 2024; 39:42-45. [PMID: 38772073 DOI: 10.1016/j.nmd.2024.05.004] [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: 11/14/2023] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 05/23/2024]
Abstract
Spinal muscular atrophy is an autosomal recessive genetic disease that can manifest with different phenotypes, classified as types 1 to 4, being type 4 the mildest form. We report a case of a 60-year-old man presenting with sudden onset of numbness in the right upper limb and with a family history of a 48-year-old brother with progressive weakness. At the first visit, his exam was unremarkable, except for a mild paresis of the right elbow extension and reduced right bicipital and tricipital reflexes. Electromyography revealed chronic motor neuronopathy and the genetic study confirmed a diagnosis of spinal muscular atrophy. At the follow-up visit his complains improved and his neurologic exam returned to normal. To our knowledge, this patient is the oldest asymptomatic SMA individual ever reported. This case highlights the need to exclude late onset spinal muscular atrophy in patients with indolent motor neuronopathy.
Collapse
Affiliation(s)
- Sandra Palma
- Hospital Garcia de Orta (Portugal), Avenida Torrado da Silva, 2805-267, Almada, Portugal.
| | - Pedro Pereira
- Hospital Garcia de Orta (Portugal), Avenida Torrado da Silva, 2805-267, Almada, Portugal.
| |
Collapse
|
5
|
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
|
6
|
Dosi C, Masson R. The impact of three SMN2 gene copies on clinical characteristics and effect of disease-modifying treatment in patients with spinal muscular atrophy: a systematic literature review. Front Neurol 2024; 15:1308296. [PMID: 38487326 PMCID: PMC10937544 DOI: 10.3389/fneur.2024.1308296] [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: 10/06/2023] [Accepted: 02/12/2024] [Indexed: 03/17/2024] Open
Abstract
Objective To review the clinical characteristics and effect of treatment in patients with spinal muscular atrophy (SMA) and three copies of the SMN2 gene. Methods We conducted a literature search in October 2022 to identify English-language clinical research on SMA that included SMN2 copy number according to PRISMA guidelines. Results Our search identified 44 studies examining the impact of three SMN2 copies on clinical characteristics (21 on phenotype, 13 on natural history, and 15 on functional status and other signs/symptoms). In children with type I SMA or presymptomatic infants with an SMN1 deletion, three SMN2 copies was associated with later symptom onset, slower decline in motor function and longer survival compared with two SMN2 copies. In patients with SMA type II or III, three SMN2 copies is associated with earlier symptom onset, loss of ambulation, and ventilator dependence compared with four SMN2 copies. Eleven studies examined treatment effects with nusinersen (nine studies), onasemnogene abeparvovec (one study), and a range of treatments (one study) in patients with three SMN2 copies. In presymptomatic infants, early treatment delayed the onset of symptoms and maintained motor function in those with three SMN2 copies. The impact of copy number on treatment response in symptomatic patients is still unclear. Conclusion SMN2 copy number is strongly correlated with SMA phenotype in patients with SMN1 deletion, while no correlation was found in patients with an SMN1 mutation. Patients with three SMN2 copies show a highly variable clinical phenotype. Early initiation of treatment is highly effective in presymptomatic patients with three SMN2 copies.
Collapse
Affiliation(s)
| | - Riccardo Masson
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Developmental Neurology Unit, Milan, Italy
| |
Collapse
|
7
|
Kiselev A, Maretina M, Shtykalova S, Al-Hilal H, Maslyanyuk N, Plokhih M, Serebryakova E, Frolova M, Shved N, Krylova N, Il’ina A, Freund S, Osinovskaya N, Sultanov I, Egorova A, Lobenskaya A, Koroteev A, Sosnina I, Gorelik Y, Bespalova O, Baranov V, Kogan I, Glotov A. Establishment of a Pilot Newborn Screening Program for Spinal Muscular Atrophy in Saint Petersburg. Int J Neonatal Screen 2024; 10:9. [PMID: 38390973 PMCID: PMC10885106 DOI: 10.3390/ijns10010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Spinal muscular atrophy 5q (SMA) is one of the most common neuromuscular inherited diseases and is the most common genetic cause of infant mortality. SMA is associated with homozygous deletion of exon 7 in the SMN1 gene. Recently developed drugs can improve the motor functions of infants with SMA when they are treated in the pre-symptomatic stage. With aim of providing an early diagnosis, newborn screening (NBS) for SMA using a real-time PCR assay with dried blood spots (DBS) was performed from January 2022 through November 2022 in Saint Petersburg, which is a representative Russian megapolis. Here, 36,140 newborns were screened by the GenomeX real-time PCR-based screening test, and three genotypes were identified: homozygous deletion carriers (4 newborns), heterozygous carriers (772 newborns), and wild-type individuals (35,364 newborns). The disease status of all four newborns that screened positive for the homozygous SMN1 deletion was confirmed by alternate methods. Two of the newborns had two copies of SMN2, and two of the newborns had three copies. We determined the incidence of spinal muscular atrophy in Saint Petersburg to be 1 in 9035 and the SMA carrier frequency to be 1 in 47. In conclusion, providing timely information regarding SMN1, confirmation of disease status, and SMN2 copy number as part of the SMA newborn-screening algorithm can significantly improve clinical follow-up, testing of family members, and treatment of patients with SMA.
Collapse
Affiliation(s)
- Anton Kiselev
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Marianna Maretina
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Sofia Shtykalova
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Haya Al-Hilal
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Natalia Maslyanyuk
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Mariya Plokhih
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Elena Serebryakova
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
- Saint Petersburg State Medical Diagnostic Center (Genetic Medical Center), Tobolskaya Street 5, 353912 Saint Petersburg, Russia; (M.F.); (A.L.); (A.K.)
| | - Marina Frolova
- Saint Petersburg State Medical Diagnostic Center (Genetic Medical Center), Tobolskaya Street 5, 353912 Saint Petersburg, Russia; (M.F.); (A.L.); (A.K.)
| | - Natalia Shved
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Nadezhda Krylova
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Arina Il’ina
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Svetlana Freund
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Natalia Osinovskaya
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Iskender Sultanov
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Anna Egorova
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Anastasia Lobenskaya
- Saint Petersburg State Medical Diagnostic Center (Genetic Medical Center), Tobolskaya Street 5, 353912 Saint Petersburg, Russia; (M.F.); (A.L.); (A.K.)
| | - Alexander Koroteev
- Saint Petersburg State Medical Diagnostic Center (Genetic Medical Center), Tobolskaya Street 5, 353912 Saint Petersburg, Russia; (M.F.); (A.L.); (A.K.)
| | - Irina Sosnina
- Saint Petersburg State Budgetary Healthcare Institution “Consulting and Diagnostic Center for Children”, Aleksa Dundić Street 36/2, 192289 Saint Petersburg, Russia;
| | - Yulia Gorelik
- Children’s City Multidisciplinary Clinical Specialized Center of High Medical Technologies, Avangardnaya Street 14, 198205 Saint Petersburg, Russia;
| | - Olesya Bespalova
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Vladislav Baranov
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Igor Kogan
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| | - Andrey Glotov
- Department of Genomic Medicine Named after V.S. Baranov, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint Petersburg, Russia; (M.M.); (S.S.); (H.A.-H.); (N.M.); (M.P.); (E.S.); (N.S.); (N.K.); (A.I.); (S.F.); (I.S.); (A.E.); (O.B.); (I.K.); (A.G.)
| |
Collapse
|
8
|
Tan J, Zhang J, Sun R, Jiang Z, Wang Y, Ma D, Jiao J, Chen H, Lin Y, Zhang Q, Xu Z, Hu P. Evaluating the performance of four assays for carrier screening of spinal muscular atrophy. Clin Chim Acta 2023; 548:117496. [PMID: 37479010 DOI: 10.1016/j.cca.2023.117496] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/19/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND AND AIMS Spinal muscular atrophy (SMA) is an autosomal recessive inherited neuromuscular condition caused by biallelic mutations in the survival of motor neuron 1 (SMN1) gene. A homozygous deletion of the SMN1 gene accounts for approximately 95-98% of SMA patients. A highly homologous gene survival motor neuron 2 (SMN2) can partially compensate for SMN1 deletion, and its copy number is associated with disease severity. Population-based carrier screening by simultaneous quantification of SMN1 and SMN2 copy numbers is the best method to prevent SMA. MATERIALS AND METHODS In this study, a total of 516 samples were re-tested for the SMN1 copy number by using quantitative polymerase chain reaction (qPCR), multiplex ligation probe amplification (MLPA), droplet digital PCR (ddPCR), high-resolution melting (HRM) analysis, and PCR-based capillary electrophoresis (PCR/CE) simultaneously. Then, the performance of these methods was compared by using MLPA results as the reference. RESULTS The results of qPCR, ddPCR, HRM, and PCR/CE in detecting heterozygous deletion of SMN1 exon 7 and the results of ddPCR, HRM, and PCR/CE in detecting ≥2 copies of SMN1 exon7 are totally consistent with those of MLPA. The sensitivity and specificity of qPCR for detection of 2 copies of SMN1 exon 7 were 99.7% and 98.8%, respectively. The sensitivity and specificity of qPCR for detection of >2 copies of SMN1 exon 7 were 96.3% and 99.8%, respectively. Compared with the MLPA results, the sensitivity and specificity of qPCR and HRM for detection of heterozygous deletion of SMN1 exon 8 were 100% and 100%, respectively. They were 99.4% and 100%, respectively for detection of 2 copies, and 100% and 100%, respectively for detection of >2 copies. The results of PCR/CE in detecting SMN1 exon 8 were consistent with those of MLPA. CONCLUSION All these four methods show excellent performance in detecting heterozygous deletion of SMN1 exon 7. All PCR/CE results are totally concordant with those of MLPA. As the most cost-effective method, qPCR also shows high sensitivity and specificity in detecting SMN1. Taken together, our study provides useful information to select appropriate methods for SMA carrier screening.
Collapse
Affiliation(s)
- Jianxin Tan
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China
| | - Jingjing Zhang
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China
| | - Ruihong Sun
- Department of Laboratory Medicine, The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Zhu Jiang
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China
| | - Yuguo Wang
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China
| | - Dingyuan Ma
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China
| | - Jiao Jiao
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China
| | - Hao Chen
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China
| | - Yingchun Lin
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China
| | - Qinxin Zhang
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China
| | - Zhengfeng Xu
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China.
| | - Ping Hu
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, People's Republic of China.
| |
Collapse
|
9
|
Maggi L, Bello L, Bonanno S, Govoni A, Caponnetto C, Passamano L, Grandis M, Trojsi F, Cerri F, Gardani A, Ferraro M, Gadaleta G, Zangaro V, Caumo L, Maioli M, Tanel R, Saccani E, Meneri M, Vacchiano V, Ricci G, Sorarù G, D'Errico E, Bortolani S, Pavesi G, Gellera C, Zanin R, Corti S, Silvestrini M, Politano L, Schenone A, Previtali SC, Berardinelli A, Turri M, Verriello L, Coccia M, Mantegazza R, Liguori R, Filosto M, Marrosu G, Tiziano FD, Siciliano G, Simone IL, Mongini T, Comi G, Pegoraro E. Adults with spinal muscular atrophy: a large-scale natural history study shows gender effect on disease. J Neurol Neurosurg Psychiatry 2022; 93:1253-1261. [PMID: 36220341 DOI: 10.1136/jnnp-2022-329320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Natural history of spinal muscular atrophy (SMA) in adult age has not been fully elucidated yet, including factors predicting disease progression and response to treatments. Aim of this retrospective, cross-sectional study, is to investigate motor function across different ages, disease patterns and gender in adult SMA untreated patients. METHODS Inclusion criteria were as follows: (1) clinical and molecular diagnosis of SMA2, SMA3 or SMA4 and (2) clinical assessments performed in adult age (>18 years). RESULTS We included 64 (38.8%) females and 101 (61.2%) males (p=0.0025), among which 21 (12.7%) SMA2, 141 (85.5%) SMA3 and 3 (1.8%) SMA4. Ratio of sitters/walkers within the SMA3 subgroup was significantly (p=0.016) higher in males (46/38) than in females (19/38). Median age at onset was significantly (p=0.0071) earlier in females (3 years; range 0-16) than in males (4 years; range 0.3-28), especially in patients carrying 4 SMN2 copies. Median Hammersmith Functional Rating Scale Expanded scores were significantly (p=0.0040) lower in males (16, range 0-64) than in females (40, range 0-62); median revised upper limb module scores were not significantly (p=0.059) different between males (24, 0-38) and females (33, range 0-38), although a trend towards worse performance in males was observed. In SMA3 patients carrying three or four SMN2 copies, an effect of female sex in prolonging ambulation was statistically significant (p=0.034). CONCLUSIONS Our data showed a relevant gender effect on SMA motor function with higher disease severity in males especially in the young adult age and in SMA3 patients.
Collapse
Affiliation(s)
- Lorenzo Maggi
- Neuroimmunology and Neuromuscular Disease Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Silvia Bonanno
- Neuroimmunology and Neuromuscular Disease Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Alessandra Govoni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Luigia Passamano
- Cardiomyology and Medical Genetics Unit, University Hospital "L Vanvitelli", Napoli, Italy
| | - Marina Grandis
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), Università di Genova, Genova, Italy
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, Università degli Studi della Campania "Luigi Vanvitelli", Napoli, Italy
| | - Federica Cerri
- Department of Neurology, IRCCS Ospedale San Raffaele, Milano, Italy
| | | | - Manfredi Ferraro
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Giulio Gadaleta
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Vittoria Zangaro
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Luca Caumo
- Department of Neurosciences, University of Padova, Padova, Italy
| | | | | | - Elena Saccani
- Specialistic Medicine Unit, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Megi Meneri
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Veria Vacchiano
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, Universita degli Studi di Bologna, Bologna, Italy
| | - Giulia Ricci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gianni Sorarù
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Eustachio D'Errico
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy
| | - Sara Bortolani
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Giovanni Pavesi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Riccardo Zanin
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Stefania Corti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mauro Silvestrini
- Department of Neurological Sciences, Ospedali Riuniti di Ancona, Ancona, Italy.,Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics Unit, University Hospital "L Vanvitelli", Napoli, Italy
| | - Angelo Schenone
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), Università di Genova, Genova, Italy
| | | | | | - Mara Turri
- Department of Neurology/Stroke Unit, San Maurizio Hospital, Bolzano, Italy
| | - Lorenzo Verriello
- Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Michela Coccia
- Department of Neurological Sciences, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Renato Mantegazza
- Neuroimmunology and Neuromuscular Disease Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Rocco Liguori
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, Universita degli Studi di Bologna, Bologna, Italy
| | - Massimiliano Filosto
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,NeMO-Brescia Clinical Center for Neuromuscular Diseases, Brescia, Italy
| | | | - Francesco Danilo Tiziano
- Department of Life Sciences and Public Health, Section of Genomic Medicine, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Roma, Italy.,Department of Laboratory Science and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Isabella Laura Simone
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy
| | - Tiziana Mongini
- Department of Neurosciences Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Giacomo Comi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, Milano, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padova, Padova, Italy
| |
Collapse
|
10
|
Lee BH, Deng S, Chiriboga CA, Kay DM, Irumudomon O, Laureta E, Delfiner L, Treidler SO, Anziska Y, Sakonju A, Kois C, Farooq O, Engelstad K, Laurenzano A, Hogan K, Caggana M, Saavedra-Matiz CA, Stevens CF, Ciafaloni E. Newborn Screening for Spinal Muscular Atrophy in New York State: Clinical Outcomes From the First 3 Years. Neurology 2022; 99:e1527-e1537. [PMID: 35835557 PMCID: PMC9576300 DOI: 10.1212/wnl.0000000000200986] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/03/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Spinal muscular atrophy (SMA) was added to the Recommended Uniform Screening Panel in July 2018 largely on the basis of the availability and efficacy of newly approved disease-modifying therapies. New York State (NYS) started universal newborn screening for SMA in October 2018. The authors report the findings from the first 3 years of screening. METHODS Statewide neonatal screening was conducted using DNA extracted from dried blood spots using a real-time quantitative PCR assay. Retrospective follow-up data were collected from 9 referral centers across the state on 34 infants. RESULTS In the first 3 years since statewide implementation, nearly 650,000 infants have been screened for SMA. Thirty-four babies screened positive and were referred to a neuromuscular specialty care center. The incidence remains lower than previously predicted. The majority (94%), including all infants with 2-3 copies of survival motor neuron (SMN) 2, have received treatment. Among treated infants, the overwhelming majority (94%; 30/32) have received gene replacement. All infants in this cohort with 3 copies of SMN2 are clinically asymptomatic posttreatment based on early clinical follow-up data. Infants with 2 copies of SMN2 are more variable in their outcomes. Electrodiagnostic outcomes data obtained from a subgroup of patients (n = 11) demonstrated either improvement or no change in compound muscle action potential (CMAP) amplitude at last clinical follow-up compared with pretreatment baseline. Most infants were treated before 6 weeks of age (median = 34.5 days of life; range 11-180 days). Delays and barriers to treatment identified by treating clinicians followed 2 broad themes: medical and nonmedical. Medical delays most commonly reported were the presence of AAV9 antibodies and elevated troponin I levels. Nonmedical barriers included delays in obtaining insurance and insurance policies regarding specific treatment modalities. DISCUSSION The findings from the NYS cohort of newborn screen-identified infants are consistent with other reports of improved outcomes from early diagnosis and treatment. Additional biomarkers of motor neuron health including EMG can potentially be helpful in detecting preclinical decline.
Collapse
Affiliation(s)
- Bo Hoon Lee
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY.
| | - Stella Deng
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Claudia A Chiriboga
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Denise M Kay
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Obehioya Irumudomon
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Emma Laureta
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Leslie Delfiner
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Simona O Treidler
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Yaacov Anziska
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Ai Sakonju
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Chelsea Kois
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Osman Farooq
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Kristin Engelstad
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Alexandra Laurenzano
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Katherine Hogan
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Michele Caggana
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Carlos A Saavedra-Matiz
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Colleen F Stevens
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| | - Emma Ciafaloni
- From the Department of Neurology (B.H.L., S.D., E.C.), University of Rochester NY; Columbia University (C.A.C., K.E., A.L.), Department of Neurology, Division of Child Neurology, New York, NY; Newborn Screening Program (D.M.K., M.C., C.A.S.-M., C.F.S.), Division of Genetics, Wadsworth Center, New York State Department of Health, Albany; Cohen Children's Medical Center (O.I., E.L., K.H.), New Hyde Park, NY; Montefiore Medical Center (L.D.), Bronx, NY; Stony Brook University (S.O.T.), Department of Neurology, Stony Brook, NY; SUNY Downstate Medical Center (Y.A.), Brooklyn, NY; SUNY Upstate Medical Center (A.S.), Department of Neurology, Syracuse, NY; Albany Medical Center (C.K.), Department of Pediatrics, Genetics and Metabolism, NY; and University of Buffalo (O.F.), NY
| |
Collapse
|
11
|
Rosow L, Lomen-Hoerth C. Treatment and Management of Adult Motor Neuron Diseases. Neuromuscul Disord 2022. [DOI: 10.1016/b978-0-323-71317-7.00012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Papaliagkas V, Foroglou N, Toulios P, Moschou M, Gavriilaki M, Notas K, Chatzikyriakou E, Zafeiridou G, Arnaoutoglou M, Kimiskidis VK. Intrathecal Administration of Nusinersen Using the Ommaya Reservoir in an Adult with 5q-Related Spinal Muscular Atrophy Type 1 and Severe Spinal Deformity. Case Rep Neurol 2021; 13:710-715. [PMID: 34950009 PMCID: PMC8647073 DOI: 10.1159/000519831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/19/2021] [Indexed: 11/19/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a hereditary neuromuscular disorder, typically caused by survival motor neuron 1 (SMN1) gene deletion in chromosome 5q resulting in loss of SMN protein. SMA type 1 progresses rapidly leading to increased mortality usually before the age of 2 years. Nusinersen, the first approved disease-modifying treatment for all 5q-SMA types and ages, is an antisense oligonucleotide administered intrathecally via repeated lumbar punctures. However, adult SMA patients typically present with severe scoliosis and spinal deformity. We present a 28-year-old patient with SMA type 1 and severe spinal deformity, who received nusinersen via a subcutaneously implanted Ommaya reservoir connected with an intrathecal catheter at the thoracic level. The repetitive administrations were completed uneventfully, obviating the need for repeated laborious lumbar punctures and eliminating radiation exposure. In adult SMA patients, performing recurrent lumbar punctures can be technically challenging raising the need for an alternative route of administration. The use of Ommaya reservoirs is a viable, practical for repeated infusions, and safe option for the intrathecal delivery of nusinersen for select cases such as an adult SMA type 1 survivor with severe spinal deformity.
Collapse
Affiliation(s)
- Vasileios Papaliagkas
- Department of Biomedical Sciences, School of Health Sciences, International Hellenic University, Thessaloniki, Greece
| | - Nikolaos Foroglou
- 1st Department of Neurosurgery, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Maria Moschou
- 1st Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Gavriilaki
- 1st Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos Notas
- Laboratory of Clinical Neurophysiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Chatzikyriakou
- Laboratory of Clinical Neurophysiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Zafeiridou
- Laboratory of Clinical Neurophysiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Marianthi Arnaoutoglou
- Laboratory of Clinical Neurophysiology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vasilios K Kimiskidis
- 1st Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
13
|
The Importance of Digging into the Genetics of SMN Genes in the Therapeutic Scenario of Spinal Muscular Atrophy. Int J Mol Sci 2021; 22:ijms22169029. [PMID: 34445733 PMCID: PMC8396600 DOI: 10.3390/ijms22169029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
After 26 years of discovery of the determinant survival motor neuron 1 and the modifier survival motor neuron 2 genes (SMN1 and SMN2, respectively), three SMN-dependent specific therapies are already approved by FDA and EMA and, as a consequence, worldwide SMA patients are currently under clinical investigation and treatment. Bi-allelic pathogenic variants (mostly deletions) in SMN1 should be detected in SMA patients to confirm the disease. Determination of SMN2 copy number has been historically employed to correlate with the phenotype, predict disease evolution, stratify patients for clinical trials and to define those eligible for treatment. In view that discordant genotype-phenotype correlations are present in SMA, besides technical issues with detection of SMN2 copy number, we have hypothesized that copy number determination is only the tip of the iceberg and that more deepen studies of variants, sequencing and structures of the SMN2 genes are necessary for a better understanding of the disease as well as to investigate possible influences in treatment responses. Here, we highlight the importance of a comprehensive approach of SMN1 and SMN2 genetics with the perspective to apply for better prediction of SMA in positive neonatal screening cases and early diagnosis to start treatments.
Collapse
|
14
|
Chong LC, Gandhi G, Lee JM, Yeo WWY, Choi SB. Drug Discovery of Spinal Muscular Atrophy (SMA) from the Computational Perspective: A Comprehensive Review. Int J Mol Sci 2021; 22:8962. [PMID: 34445667 PMCID: PMC8396480 DOI: 10.3390/ijms22168962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 01/27/2021] [Indexed: 01/02/2023] Open
Abstract
Spinal muscular atrophy (SMA), one of the leading inherited causes of child mortality, is a rare neuromuscular disease arising from loss-of-function mutations of the survival motor neuron 1 (SMN1) gene, which encodes the SMN protein. When lacking the SMN protein in neurons, patients suffer from muscle weakness and atrophy, and in the severe cases, respiratory failure and death. Several therapeutic approaches show promise with human testing and three medications have been approved by the U.S. Food and Drug Administration (FDA) to date. Despite the shown promise of these approved therapies, there are some crucial limitations, one of the most important being the cost. The FDA-approved drugs are high-priced and are shortlisted among the most expensive treatments in the world. The price is still far beyond affordable and may serve as a burden for patients. The blooming of the biomedical data and advancement of computational approaches have opened new possibilities for SMA therapeutic development. This article highlights the present status of computationally aided approaches, including in silico drug repurposing, network driven drug discovery as well as artificial intelligence (AI)-assisted drug discovery, and discusses the future prospects.
Collapse
Affiliation(s)
- Li Chuin Chong
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (L.C.C.); (J.M.L.)
| | - Gayatri Gandhi
- Perdana University Graduate School of Medicine, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (G.G.); (W.W.Y.Y.)
| | - Jian Ming Lee
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (L.C.C.); (J.M.L.)
| | - Wendy Wai Yeng Yeo
- Perdana University Graduate School of Medicine, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (G.G.); (W.W.Y.Y.)
| | - Sy-Bing Choi
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Kuala Lumpur 50490, Malaysia; (L.C.C.); (J.M.L.)
| |
Collapse
|
15
|
Blatnik AJ, McGovern VL, Burghes AHM. What Genetics Has Told Us and How It Can Inform Future Experiments for Spinal Muscular Atrophy, a Perspective. Int J Mol Sci 2021; 22:8494. [PMID: 34445199 PMCID: PMC8395208 DOI: 10.3390/ijms22168494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
Proximal spinal muscular atrophy (SMA) is an autosomal recessive neurodegenerative disorder characterized by motor neuron loss and subsequent atrophy of skeletal muscle. SMA is caused by deficiency of the essential survival motor neuron (SMN) protein, canonically responsible for the assembly of the spliceosomal small nuclear ribonucleoproteins (snRNPs). Therapeutics aimed at increasing SMN protein levels are efficacious in treating SMA. However, it remains unknown how deficiency of SMN results in motor neuron loss, resulting in many reported cellular functions of SMN and pathways affected in SMA. Herein is a perspective detailing what genetics and biochemistry have told us about SMA and SMN, from identifying the SMA determinant region of the genome, to the development of therapeutics. Furthermore, we will discuss how genetics and biochemistry have been used to understand SMN function and how we can determine which of these are critical to SMA moving forward.
Collapse
Affiliation(s)
| | | | - Arthur H. M. Burghes
- Department of Biological Chemistry & Pharmacology, The Ohio State University Wexner Medical Center, Rightmire Hall, Room 168, 1060 Carmack Road, Columbus, OH 43210, USA; (A.J.B.III); (V.L.M.)
| |
Collapse
|
16
|
Butchbach MER. Genomic Variability in the Survival Motor Neuron Genes ( SMN1 and SMN2): Implications for Spinal Muscular Atrophy Phenotype and Therapeutics Development. Int J Mol Sci 2021; 22:ijms22157896. [PMID: 34360669 PMCID: PMC8348669 DOI: 10.3390/ijms22157896] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a leading genetic cause of infant death worldwide that is characterized by loss of spinal motor neurons leading to muscle weakness and atrophy. SMA results from the loss of survival motor neuron 1 (SMN1) gene but retention of its paralog SMN2. The copy numbers of SMN1 and SMN2 are variable within the human population with SMN2 copy number inversely correlating with SMA severity. Current therapeutic options for SMA focus on increasing SMN2 expression and alternative splicing so as to increase the amount of SMN protein. Recent work has demonstrated that not all SMN2, or SMN1, genes are equivalent and there is a high degree of genomic heterogeneity with respect to the SMN genes. Because SMA is now an actionable disease with SMN2 being the primary target, it is imperative to have a comprehensive understanding of this genomic heterogeneity with respect to hybrid SMN1–SMN2 genes generated by gene conversion events as well as partial deletions of the SMN genes. This review will describe this genetic heterogeneity in SMA and its impact on disease phenotype as well as therapeutic efficacy.
Collapse
Affiliation(s)
- Matthew E. R. Butchbach
- Center for Applied Clinical Genomics, Nemours Children’s Health Delaware, Wilmington, DE 19803, USA;
- Center for Pediatric Research, Nemours Children’s Health Delaware, Wilmington, DE 19803, USA
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
17
|
Sansa A, Hidalgo I, Miralles MP, de la Fuente S, Perez-Garcia MJ, Munell F, Soler RM, Garcera A. Spinal Muscular Atrophy autophagy profile is tissue-dependent: differential regulation between muscle and motoneurons. Acta Neuropathol Commun 2021; 9:122. [PMID: 34217376 PMCID: PMC8254901 DOI: 10.1186/s40478-021-01223-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/24/2021] [Indexed: 11/10/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular genetic disease caused by reduced survival motor neuron (SMN) protein. SMN is ubiquitous and deficient levels cause spinal cord motoneurons (MNs) degeneration and muscle atrophy. Nevertheless, the mechanism by which SMN reduction in muscle contributes to SMA disease is not fully understood. Therefore, studies evaluating atrophy mechanisms in SMA muscles will contribute to strengthening current knowledge of the pathology. Here we propose to evaluate autophagy in SMA muscle, a pathway altered in myotube atrophy. We analized autophagy proteins and mTOR in muscle biopsies, fibroblasts, and lymphoblast cell lines from SMA patients and in gastrocnemius muscles from a severe SMA mouse model. Human MNs differentiated from SMA and unaffected control iPSCs were also included in the analysis of the autophagy. Muscle biopsies, fibroblasts, and lymphoblast cell lines from SMA patients showed reduction of the autophagy marker LC3-II. In SMA mouse gastrocnemius, we observed lower levels of LC3-II, Beclin 1, and p62/SQSTM1 proteins at pre-symptomatic stage. mTOR phosphorylation at Ser2448 was decreased in SMA muscle cells. However, in mouse and human cultured SMA MNs mTOR phosphorylation and LC3-II levels were increased. These results suggest a differential regulation in SMA of the autophagy process in muscle cells and MNs. Opposite changes in autophagy proteins and mTOR phosphorylation between muscle cells and neurons were observed. These differences may reflect a specific response to SMN reduction, which could imply diverse tissue-dependent reactions to therapies that should be taken into account when treating SMA patients.
Collapse
|
18
|
Blasco-Pérez L, Paramonov I, Leno J, Bernal S, Alias L, Fuentes-Prior P, Cuscó I, Tizzano EF. Beyond copy number: A new, rapid, and versatile method for sequencing the entire SMN2 gene in SMA patients. Hum Mutat 2021; 42:787-795. [PMID: 33739559 PMCID: PMC8252042 DOI: 10.1002/humu.24200] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/13/2021] [Accepted: 03/11/2021] [Indexed: 01/16/2023]
Abstract
Spinal muscular atrophy (SMA) is caused by bi‐allelic loss or pathogenic variants in the SMN1 gene. SMN2, the highly homologous copy of SMN1, is considered the major phenotypic modifier of the disease. Determination of SMN2 copy number is essential to establish robust genotype–phenotype correlations and predict disease evolution, to stratify patients for clinical trials, as well as to define those eligible for treatment. Discordant genotype–phenotype correlations are not uncommon in SMA, some of which are due to intragenic SMN2 variants that may influence the amount of complete SMN transcripts and, therefore, of full‐length SMN protein. Detection of these variants is crucial to predict SMA phenotypes in the present scenario of therapeutic advances and with the perspective of SMA neonatal screening and early diagnosis to start treatments. Here, we present a novel, affordable, and versatile method for complete sequencing of the SMN2 gene based on long‐range polymerase chain reaction and next‐generation sequencing. The method was validated by analyzing samples from 53 SMA patients who lack SMN1, allowing to characterize paralogous, rare variants, and single‐nucleotide polymorphisms of SMN2 as well as SMN2–SMN1 hybrid genes. The method identifies partial deletions and can be adapted to determine rare pathogenic variants in patients with at least one SMN1 copy.
Collapse
Affiliation(s)
- Laura Blasco-Pérez
- Medicine Genetics Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,Department of Clinical and Molecular Genetics, Hospital Vall d'Hebron, Barcelona, Spain
| | - Ida Paramonov
- Medicine Genetics Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,Department of Clinical and Molecular Genetics, Hospital Vall d'Hebron, Barcelona, Spain
| | - Jordi Leno
- Medicine Genetics Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,Department of Clinical and Molecular Genetics, Hospital Vall d'Hebron, Barcelona, Spain
| | - Sara Bernal
- Genetics Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)
| | - Laura Alias
- Genetics Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)
| | - Pablo Fuentes-Prior
- Molecular Bases of Disease, Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ivon Cuscó
- Medicine Genetics Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,Department of Clinical and Molecular Genetics, Hospital Vall d'Hebron, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)
| | - Eduardo F Tizzano
- Medicine Genetics Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,Department of Clinical and Molecular Genetics, Hospital Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
19
|
Lusakowska A, Jedrzejowska M, Kaminska A, Janiszewska K, Grochowski P, Zimowski J, Sierdzinski J, Kostera-Pruszczyk A. Observation of the natural course of type 3 spinal muscular atrophy: data from the polish registry of spinal muscular atrophy. Orphanet J Rare Dis 2021; 16:150. [PMID: 33761963 PMCID: PMC7992780 DOI: 10.1186/s13023-021-01771-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 03/09/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is one of the most frequent and severe genetic diseases leading to premature death or severe motor disability. New therapies have been developed in recent years that change the natural history of the disease. The aim of this study is to describe patients included in the Polish Registry of SMA, with a focus on the course of type 3 SMA (SMA3) before the availability of disease-modifying treatments. RESULTS 790 patients with SMA were included in the registry (173 with type 1 [SMA1], 218 with type 2 [SMA2], 393 with SMA3, and six with type 4 SMA [SMA4]), most (52%) of whom were adults. Data on SMN2 gene copy number were available for 672 (85%) patients. The mean age of onset was 5 months for SMA1, 11.5 months for SMA2, and 4.5 years for SMA3. In patients with SMA3, the first symptoms occurred earlier in those with three copies of SMN2 than in those with four copies of SMN2 (3.2 years vs. 6.7 years). The age of onset of SMA3 was younger in girls than in boys (3.1 years vs. 5.7 years), with no new cases observed in women older than 16 years. Male patients outnumbered female patients, especially among patients with SMA3b (49 female vs. 85 male patients) and among patients with SMA3 with four copies of SMN2 (30 female vs. 69 male patients). 44% of patients with SMA3 were still able to walk; in those who were not still able to walk, the mean age of immobilization was 14.0 years. Patients with SMA3a (age of onset < 3 years) and three copies of SMN2 had significantly worse prognosis for remaining ambulant than patients with SMA3b (age of onset ≥ 3 years) and four copies of SMN2. CONCLUSIONS The Registry of SMA is an effective tool for assessing the disease course in the real world setting. SMN2 copy number is an important prognostic factor for the age of onset and ambulation in SMA3. Sex and age of disease onset also strongly affect the course of SMA. Data supplied by this study can aid treatment decisions.
Collapse
Affiliation(s)
- Anna Lusakowska
- Department of Neurology, European Reference Network EURO-NMD, Medical University of Warsaw, Warsaw, Poland
| | - Maria Jedrzejowska
- Rare Diseases Research Platform, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Kaminska
- Department of Neurology, European Reference Network EURO-NMD, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Janiszewska
- Department of Neurology, European Reference Network EURO-NMD, Medical University of Warsaw, Warsaw, Poland
| | - Przemysław Grochowski
- Student Research Group of Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Janusz Zimowski
- Department of Genetics, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Janusz Sierdzinski
- Department of Medical Informatics and Telemedicine, Medical University of Warsaw, Warsaw, Poland
| | - Anna Kostera-Pruszczyk
- Department of Neurology, European Reference Network EURO-NMD, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
20
|
Cuscó I, Bernal S, Blasco-Pérez L, Calucho M, Alias L, Fuentes-Prior P, Tizzano EF. Practical guidelines to manage discordant situations of SMN2 copy number in patients with spinal muscular atrophy. NEUROLOGY-GENETICS 2020; 6:e530. [PMID: 33324756 PMCID: PMC7713720 DOI: 10.1212/nxg.0000000000000530] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/29/2020] [Indexed: 12/26/2022]
Abstract
Objective Assessment of SMN2 copy number in patients with spinal muscular atrophy (SMA) is essential to establish careful genotype-phenotype correlations and predict disease evolution. This issue is becoming crucial in the present scenario of therapeutic advances with the perspective of SMA neonatal screening and early diagnosis to initiate treatment, as this value is critical to stratify patients for clinical trials and to define those eligible to receive medication. Several technical pitfalls and interindividual variations may account for reported discrepancies in the estimation of SMN2 copy number and establishment of phenotype-genotype correlations. Methods We propose a management guide based on a sequence of specified actions once SMN2 copy number is determined for a given patient. Regardless of the method used to estimate the number of SMN2 copies, our approach focuses on the manifestations of the patient to recommend how to proceed in each case. Results We defined situations according to SMN2 copy number in a presymptomatic scenario of screening, in which we predict the possible evolution, and when a symptomatic patient is genetically confirmed. Unexpected discordant cases include patients having a single SMN2 copy but noncongenital disease forms, 2 SMN2 copies compatible with type II or III SMA, and 3 or 4 copies of the gene showing more severe disease than expected. Conclusions Our proposed guideline would help to systematically identify discordant SMA cases that warrant further genetic investigation. The SMN2 gene, as the main modifier of SMA phenotype, deserves a more in-depth study to provide more accurate genotype-phenotype correlations.
Collapse
Affiliation(s)
- Ivon Cuscó
- Medicine Genetics Group (I.C., L.B.-P., M.C., E.F.T.), Vall dHebron Research Institute (VHIR), Barcelona; Department of Clinical and Molecular Genetics (I.C., L.B.-P., M.C., E.F.T.), Hospital Vall dHebron, Barcelona; Department of Genetics (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Biomedical Research Institute Sant Pau (IIB Sant Pau) (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII, U-705 Barcelona) (S.B., L.A.), Madrid; Molecular Bases of Disease (P.F.-P.), Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Sara Bernal
- Medicine Genetics Group (I.C., L.B.-P., M.C., E.F.T.), Vall dHebron Research Institute (VHIR), Barcelona; Department of Clinical and Molecular Genetics (I.C., L.B.-P., M.C., E.F.T.), Hospital Vall dHebron, Barcelona; Department of Genetics (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Biomedical Research Institute Sant Pau (IIB Sant Pau) (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII, U-705 Barcelona) (S.B., L.A.), Madrid; Molecular Bases of Disease (P.F.-P.), Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Laura Blasco-Pérez
- Medicine Genetics Group (I.C., L.B.-P., M.C., E.F.T.), Vall dHebron Research Institute (VHIR), Barcelona; Department of Clinical and Molecular Genetics (I.C., L.B.-P., M.C., E.F.T.), Hospital Vall dHebron, Barcelona; Department of Genetics (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Biomedical Research Institute Sant Pau (IIB Sant Pau) (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII, U-705 Barcelona) (S.B., L.A.), Madrid; Molecular Bases of Disease (P.F.-P.), Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Maite Calucho
- Medicine Genetics Group (I.C., L.B.-P., M.C., E.F.T.), Vall dHebron Research Institute (VHIR), Barcelona; Department of Clinical and Molecular Genetics (I.C., L.B.-P., M.C., E.F.T.), Hospital Vall dHebron, Barcelona; Department of Genetics (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Biomedical Research Institute Sant Pau (IIB Sant Pau) (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII, U-705 Barcelona) (S.B., L.A.), Madrid; Molecular Bases of Disease (P.F.-P.), Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Laura Alias
- Medicine Genetics Group (I.C., L.B.-P., M.C., E.F.T.), Vall dHebron Research Institute (VHIR), Barcelona; Department of Clinical and Molecular Genetics (I.C., L.B.-P., M.C., E.F.T.), Hospital Vall dHebron, Barcelona; Department of Genetics (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Biomedical Research Institute Sant Pau (IIB Sant Pau) (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII, U-705 Barcelona) (S.B., L.A.), Madrid; Molecular Bases of Disease (P.F.-P.), Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Pablo Fuentes-Prior
- Medicine Genetics Group (I.C., L.B.-P., M.C., E.F.T.), Vall dHebron Research Institute (VHIR), Barcelona; Department of Clinical and Molecular Genetics (I.C., L.B.-P., M.C., E.F.T.), Hospital Vall dHebron, Barcelona; Department of Genetics (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Biomedical Research Institute Sant Pau (IIB Sant Pau) (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII, U-705 Barcelona) (S.B., L.A.), Madrid; Molecular Bases of Disease (P.F.-P.), Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Eduardo F Tizzano
- Medicine Genetics Group (I.C., L.B.-P., M.C., E.F.T.), Vall dHebron Research Institute (VHIR), Barcelona; Department of Clinical and Molecular Genetics (I.C., L.B.-P., M.C., E.F.T.), Hospital Vall dHebron, Barcelona; Department of Genetics (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Biomedical Research Institute Sant Pau (IIB Sant Pau) (S.B., L.A.), Hospital de la Santa Creu i Sant Pau, Barcelona; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII, U-705 Barcelona) (S.B., L.A.), Madrid; Molecular Bases of Disease (P.F.-P.), Biomedical Research Institute Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| |
Collapse
|
21
|
Müller-Felber W, Vill K, Schwartz O, Gläser D, Nennstiel U, Wirth B, Burggraf S, Röschinger W, Becker M, Durner J, Eggermann K, Müller C, Hannibal I, Olgemöller B, Schara U, Blaschek A, Kölbel H. Infants Diagnosed with Spinal Muscular Atrophy and 4 SMN2 Copies through Newborn Screening - Opportunity or Burden? J Neuromuscul Dis 2020; 7:109-117. [PMID: 32144995 PMCID: PMC7175938 DOI: 10.3233/jnd-200475] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Although the value of newborn screening (NBS) for early detection and treatment opportunity in SMA patients is generally accepted, there is still an ongoing discussion about the best strategy in children with 4 and more copies of the SMN2 gene. This gene is known to be the most important but not the only disease modifier. In our SMA-NBS pilot project in Germany comprising 278,970 infants screened between January 2018 and November 2019 were 38 positive cases with a homozygous SMN1 deletion. 40% of them had 4 or more SMN2 copies. The incidence for homozygous SMN1 deletion was 1 : 7350, which is within the known range of SMA incidence in Germany. Of the 15 SMA children with 4 SMN2 copies, one child developed physical signs of SMA by the age of 8 months. Reanalysis of the SMN2 copy number by a different test method revealed 3 copies. Two children had affected siblings with SMA Type III, who were diagnosed only after detection of the index patient in the NBS. One had a positive family history with an affected aunt (onset of disease at the age of 3 years). Three families were lost to medical follow up; two because of socioeconomic reasons and one to avoid the psychological stress associated with the appointments. Decisions on how to handle patients with 4 SMN2 copies are discussed in the light of the experience gathered from our NBS pilot SMA program.
Collapse
Affiliation(s)
- Wolfgang Müller-Felber
- Dr. v. Hauner Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, LMU - University of Munich, Munich, Germany
| | - Katharina Vill
- Dr. v. Hauner Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, LMU - University of Munich, Munich, Germany
| | - Oliver Schwartz
- Department of Pediatric Neurology, Muenster University Hospital, Münster, Germany
| | - Dieter Gläser
- Genetikum ®, Center for Human Genetics, Neu-Ulm, Germany
| | - Uta Nennstiel
- Screening Center of the Bavarian Health and Food Safety Authority, Oberschleissheim, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Genetics Cologne and Center for Rare Diseases Cologne, University of Cologne, Cologne, Germany
| | | | | | | | - Jürgen Durner
- Labor Becker und Kollegen, Munich, Germany.,Department of Operative/Restorative Dentistry, Periodontology and Pedodontics, Ludwig-Maximilians-Universität München, Goethestr. 70, 80336 Munich, Germany
| | - Katja Eggermann
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Christine Müller
- Dr. v. Hauner Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, LMU - University of Munich, Munich, Germany
| | - Iris Hannibal
- Dr. v. Hauner Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, LMU - University of Munich, Munich, Germany
| | | | - Ulrike Schara
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, University of Essen, Germany
| | - Astrid Blaschek
- Dr. v. Hauner Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, LMU - University of Munich, Munich, Germany
| | - Heike Kölbel
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, University of Essen, Germany
| |
Collapse
|
22
|
Genetic screening method for analyzing survival motor neuron copy number in spinal muscular atrophy by multiplex ligation-dependent probe amplification and droplet digital polymerase chain reaction. Chin Med J (Engl) 2020; 133:2510-2511. [PMID: 32960844 PMCID: PMC7575165 DOI: 10.1097/cm9.0000000000001102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
23
|
Jones CC, Cook SF, Jarecki J, Belter L, Reyna SP, Staropoli J, Farwell W, Hobby K. Spinal Muscular Atrophy (SMA) Subtype Concordance in Siblings: Findings From the Cure SMA Cohort. J Neuromuscul Dis 2020; 7:33-40. [PMID: 31707372 PMCID: PMC7029365 DOI: 10.3233/jnd-190399] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Background: Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder caused by homozygous survival of motor neuron 1 (SMN1) gene disruption. Despite a genetic etiology, little is known about subtype concordance among siblings. Objective: To investigate subtype concordance among siblings with SMA. Methods: Cure SMA maintains a database of newly diagnosed patients with SMA, which was utilized for this research. Results: Among 303 sibships identified between 1996 and 2016, 84.8% were subtype concordant. Of concordant sibships, subtype distribution was as follows: Type I, 54.5%; Type II, 31.9%; Type III, 13.2%; Type IV, 0.4%. Subtype and concordance/discordance association was significant (Fisher’s exact test; p < 0.0001). Among discordant sibships (chi-square test, p < 0.0001), Types II/III (52.2%) and Types I/II (28.3%) were the most common pairs. No association was found between sibling sex and concordance. Our findings show that most siblings with SMA shared the same subtype concordance (most commonly Type I). Conclusions: These data are valuable for understanding familial occurrence of SMA subtypes, enabling better individual treatment and management planning in view of new treatment options and newborn screening initiatives.
Collapse
|
24
|
Fang YL, Li N, Zhi XF, Zheng J, Liu Y, Pu LJ, Gu CY, Shu JB, Cai CQ. Discovery of specific mutations in spinal muscular atrophy patients by next-generation sequencing. Neurol Sci 2020; 42:1827-1833. [PMID: 32895776 DOI: 10.1007/s10072-020-04697-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/28/2020] [Indexed: 12/28/2022]
Abstract
Spinal muscular atrophy (SMA) is a type of autosomal recessive genetic disease, which seriously threatens the health and lives of children and adolescents. We attempted to find some genes and mutations related to the onset of SMA. Eighty-three whole-blood samples were collected from 28 core families, including 28 probands with clinically suspected SMA (20 SMA patients, 5 non-SMA children, and 3 patients with unknown etiology) and their parents. The multiplex ligation probe amplification (MLPA) was performed for preliminary diagnosis. The high-throughput sequencing technology was used to conduct the whole-exome sequencing analysis. We analyzed the mutations in adjacent genes of SMN1 gene and the unique mutations that only occurred in SMA patients. According to the MLPA results, 20 probands were regarded as experimental group and 5 non-SMA children as control group. A total of 10 mutations were identified in the adjacent genes of SMN1 gene. GUSBP1 g.[69515863G>A], GUSBP1 g.[69515870C>T], and SMA4 g.[69515738C>A] were the top three most frequent sites. SMA4 g.[69515726A>G] and OCLN c.[818G>T] have not been reported in the existing relevant researches. Seventeen point mutations in the DYNC1H1 gene were only recognized in SMA children, and the top two most common mutations were c.[2869-34A>T] and c.[345-89A>G]; c.[7473+105C>T] was the splicing mutation that might change the mRNA splicing site. The mutations of SMA4 g.[69515726A>G], OCLN c.[818G>T], DYNC1H1 c.[2869-34A>T], DYNC1H1 c.[345-89A>G], and DYNC1H1 c.[7473+105C>T] in the adjacent genes of SMN1 gene and other genes might be related to the onset of SMA.
Collapse
Affiliation(s)
- Yu-Lian Fang
- Institute of Pediatrics, Tianjin Children's Hospital, 238 Longyan Road, Beichen District, Tianjin, 300134, China
| | - Na Li
- Department of Neonatology, The Pediatric Clinical College, Tianjin Medical University, Tianjin, 300134, China.,Department of Neonatology, Tianjin Children's Hospital, Tianjin, 300134, China
| | - Xiu-Fang Zhi
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
| | - Jie Zheng
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
| | - Yang Liu
- Department of Neonatology, The Pediatric Clinical College, Tianjin Medical University, Tianjin, 300134, China.,Department of Neonatology, Tianjin Children's Hospital, Tianjin, 300134, China
| | - Lin-Jie Pu
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
| | - Chun-Yu Gu
- Graduate School, Tianjin Medical University, Tianjin, 300070, China
| | - Jian-Bo Shu
- Institute of Pediatrics, Tianjin Children's Hospital, 238 Longyan Road, Beichen District, Tianjin, 300134, China. .,Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, 300134, China.
| | - Chun-Quan Cai
- Institute of Pediatrics, Tianjin Children's Hospital, 238 Longyan Road, Beichen District, Tianjin, 300134, China. .,Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, 300134, China. .,Department of Neurosurgery, Tianjin Children's Hospital, Tianjin, 300134, China.
| |
Collapse
|
25
|
Smeriglio P, Langard P, Querin G, Biferi MG. The Identification of Novel Biomarkers Is Required to Improve Adult SMA Patient Stratification, Diagnosis and Treatment. J Pers Med 2020; 10:jpm10030075. [PMID: 32751151 PMCID: PMC7564782 DOI: 10.3390/jpm10030075] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
Spinal muscular atrophy (SMA) is currently classified into five different subtypes, from the most severe (type 0) to the mildest (type 4) depending on age at onset, best motor function achieved, and copy number of the SMN2 gene. The two recent approved treatments for SMA patients revolutionized their life quality and perspectives. However, upon treatment with Nusinersen, the most widely administered therapy up to date, a high degree of variability in therapeutic response was observed in adult SMA patients. These data, together with the lack of natural history information and the wide spectrum of disease phenotypes, suggest that further efforts are needed to develop precision medicine approaches for all SMA patients. Here, we compile the current methods for functional evaluation of adult SMA patients treated with Nusinersen. We also present an overview of the known molecular changes underpinning disease heterogeneity. We finally highlight the need for novel techniques, i.e., -omics approaches, to capture phenotypic differences and to understand the biological signature in order to revise the disease classification and device personalized treatments.
Collapse
Affiliation(s)
- Piera Smeriglio
- Centre of Research in Myology, Institute of Myology, Sorbonne Université, INSERM, 75013 Paris, France; (P.L.); (G.Q.)
- Correspondence: (P.S.); (M.G.B.)
| | - Paul Langard
- Centre of Research in Myology, Institute of Myology, Sorbonne Université, INSERM, 75013 Paris, France; (P.L.); (G.Q.)
| | - Giorgia Querin
- Centre of Research in Myology, Institute of Myology, Sorbonne Université, INSERM, 75013 Paris, France; (P.L.); (G.Q.)
- Association Institut de Myologie, Plateforme Essais Cliniques Adultes, 75013 Paris, France
- APHP, Service de Neuromyologie, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Maria Grazia Biferi
- Centre of Research in Myology, Institute of Myology, Sorbonne Université, INSERM, 75013 Paris, France; (P.L.); (G.Q.)
- Correspondence: (P.S.); (M.G.B.)
| |
Collapse
|
26
|
Nusinersen ameliorates motor function and prevents motoneuron Cajal body disassembly and abnormal poly(A) RNA distribution in a SMA mouse model. Sci Rep 2020; 10:10738. [PMID: 32612161 PMCID: PMC7330045 DOI: 10.1038/s41598-020-67569-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/08/2020] [Indexed: 11/09/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a devastating autosomal recessive neuromuscular disease characterized by degeneration of spinal cord alpha motor neurons (αMNs). SMA is caused by the homozygous deletion or mutation of the survival motor neuron 1 (SMN1) gene, resulting in reduced expression of SMN protein, which leads to αMN degeneration and muscle atrophy. The majority of transcripts of a second gene (SMN2) generate an alternative spliced isoform that lacks exon 7 and produces a truncated nonfunctional form of SMN. A major function of SMN is the biogenesis of spliceosomal snRNPs, which are essential components of the pre-mRNA splicing machinery, the spliceosome. In recent years, new potential therapies have been developed to increase SMN levels, including treatment with antisense oligonucleotides (ASOs). The ASO-nusinersen (Spinraza) promotes the inclusion of exon 7 in SMN2 transcripts and notably enhances the production of full-length SMN in mouse models of SMA. In this work, we used the intracerebroventricular injection of nusinersen in the SMN∆7 mouse model of SMA to evaluate the effects of this ASO on the behavior of Cajal bodies (CBs), nuclear structures involved in spliceosomal snRNP biogenesis, and the cellular distribution of polyadenylated mRNAs in αMNs. The administration of nusinersen at postnatal day (P) 1 normalized SMN expression in the spinal cord but not in skeletal muscle, rescued the growth curve and improved motor behavior at P12 (late symptomatic stage). Importantly, this ASO recovered the number of canonical CBs in MNs, significantly reduced the abnormal accumulation of polyadenylated RNAs in nuclear granules, and normalized the expression of the pre-mRNAs encoding chondrolectin and choline acetyltransferase, two key factors for αMN homeostasis. We propose that the splicing modulatory function of nusinersen in SMA αMN is mediated by the rescue of CB biogenesis, resulting in enhanced polyadenylated pre-mRNA transcription and splicing and nuclear export of mature mRNAs for translation. Our results support that the selective restoration of SMN expression in the spinal cord has a beneficial impact not only on αMNs but also on skeletal myofibers. However, the rescue of SMN expression in muscle appears to be necessary for the complete recovery of motor function.
Collapse
|
27
|
Deguise MO, De Repentigny Y, Tierney A, Beauvais A, Michaud J, Chehade L, Thabet M, Paul B, Reilly A, Gagnon S, Renaud JM, Kothary R. Motor transmission defects with sex differences in a new mouse model of mild spinal muscular atrophy. EBioMedicine 2020; 55:102750. [PMID: 32339936 PMCID: PMC7184161 DOI: 10.1016/j.ebiom.2020.102750] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/25/2020] [Accepted: 03/19/2020] [Indexed: 12/31/2022] Open
Abstract
Background Mouse models of mild spinal muscular atrophy (SMA) have been extremely challenging to generate. This paucity of model systems has limited our understanding of pathophysiological events in milder forms of the disease and of the effect of SMN depletion during aging. Methods A mild mouse model of SMA, termed Smn2B/−;SMN2+/−, was generated by crossing Smn−/−;SMN2 and Smn2B/2B mice. This new model was characterized using behavioral testing, histology, western blot, muscle-nerve electrophysiology as well as ultrasonography to study classical SMA features and extra-neuronal involvement. Findings Smn2B/−;SMN2+/− mice have normal survival, mild but sustained motor weakness, denervation and neuronal/neuromuscular junction (NMJ) transmission defects, and neurogenic muscle atrophy that are more prominent in male mice. Increased centrally located nuclei, intrinsic contractile and relaxation muscle defects were also identified in both female and male mice, with some male predominance. There was an absence of extra-neuronal pathology. Interpretation The Smn2B/−;SMN2+/− mouse provides a model of mild SMA, displaying some hallmark features including reduced weight, sustained motor weakness, electrophysiological transmission deficit, NMJ defects, and muscle atrophy. Early and prominent increase central nucleation and intrinsic electrophysiological deficits demonstrate the potential role played by muscle in SMA disease. The use of this model will allow for the understanding of the most susceptible pathogenic molecular changes in motor neurons and muscles, investigation of the effects of SMN depletion in aging, sex differences and most importantly will provide guidance for the currently aging SMA patients treated with the recently approved genetic therapies. Funding : This work was supported by Cure SMA/Families of SMA Canada (grant numbers KOT-1819 and KOT-2021); Muscular Dystrophy Association (USA) (grant number 575466); and Canadian Institutes of Health Research (CIHR) (grant number PJT-156379).
Collapse
Affiliation(s)
- Marc-Olivier Deguise
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Yves De Repentigny
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
| | - Alexandra Tierney
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
| | - Ariane Beauvais
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
| | - Jean Michaud
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Lucia Chehade
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Mohamed Thabet
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Brittany Paul
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada; Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Aoife Reilly
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Sabrina Gagnon
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
| | - Jean-Marc Renaud
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada; Department of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.
| |
Collapse
|
28
|
Lee BH, Collins E, Lewis L, Guntrum D, Eichinger K, Voter K, Abdel-Hamid HZ, Ciafaloni E. Combination therapy with nusinersen and AVXS-101 in SMA type 1. Neurology 2019; 93:640-641. [PMID: 31488615 DOI: 10.1212/wnl.0000000000008207] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/26/2019] [Indexed: 12/22/2022] Open
Affiliation(s)
- Bo Hoon Lee
- From Pediatric Neurology (B.H.L.), Department of Neurology (E.C., L.L., D.G., K.E., E.C.), and Department of Pediatric Pulmonology (K.V.), University of Rochester, NY; and Pediatric Neurology (H.Z.A.-H.), Children's Hospital of Pittsburgh of UPMC, PA
| | - Erin Collins
- From Pediatric Neurology (B.H.L.), Department of Neurology (E.C., L.L., D.G., K.E., E.C.), and Department of Pediatric Pulmonology (K.V.), University of Rochester, NY; and Pediatric Neurology (H.Z.A.-H.), Children's Hospital of Pittsburgh of UPMC, PA
| | - Leann Lewis
- From Pediatric Neurology (B.H.L.), Department of Neurology (E.C., L.L., D.G., K.E., E.C.), and Department of Pediatric Pulmonology (K.V.), University of Rochester, NY; and Pediatric Neurology (H.Z.A.-H.), Children's Hospital of Pittsburgh of UPMC, PA
| | - Debra Guntrum
- From Pediatric Neurology (B.H.L.), Department of Neurology (E.C., L.L., D.G., K.E., E.C.), and Department of Pediatric Pulmonology (K.V.), University of Rochester, NY; and Pediatric Neurology (H.Z.A.-H.), Children's Hospital of Pittsburgh of UPMC, PA
| | - Katy Eichinger
- From Pediatric Neurology (B.H.L.), Department of Neurology (E.C., L.L., D.G., K.E., E.C.), and Department of Pediatric Pulmonology (K.V.), University of Rochester, NY; and Pediatric Neurology (H.Z.A.-H.), Children's Hospital of Pittsburgh of UPMC, PA
| | - Karen Voter
- From Pediatric Neurology (B.H.L.), Department of Neurology (E.C., L.L., D.G., K.E., E.C.), and Department of Pediatric Pulmonology (K.V.), University of Rochester, NY; and Pediatric Neurology (H.Z.A.-H.), Children's Hospital of Pittsburgh of UPMC, PA
| | - Hoda Z Abdel-Hamid
- From Pediatric Neurology (B.H.L.), Department of Neurology (E.C., L.L., D.G., K.E., E.C.), and Department of Pediatric Pulmonology (K.V.), University of Rochester, NY; and Pediatric Neurology (H.Z.A.-H.), Children's Hospital of Pittsburgh of UPMC, PA
| | - Emma Ciafaloni
- From Pediatric Neurology (B.H.L.), Department of Neurology (E.C., L.L., D.G., K.E., E.C.), and Department of Pediatric Pulmonology (K.V.), University of Rochester, NY; and Pediatric Neurology (H.Z.A.-H.), Children's Hospital of Pittsburgh of UPMC, PA.
| |
Collapse
|
29
|
Lee BH. Comment: Accuracy and importance of SMN2 copy number. Neurology 2019; 93:270. [DOI: 10.1212/wnl.0000000000007839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
30
|
Park JM, Nishio H, Shin JH, Park JS. A Spinal Muscular Atrophy Family with Intrafamilial Phenotype Differences Despite the Same Copy-Number Variation in SMN2. J Clin Neurol 2019; 15:395-397. [PMID: 31286713 PMCID: PMC6620452 DOI: 10.3988/jcn.2019.15.3.395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/22/2018] [Accepted: 12/27/2018] [Indexed: 11/17/2022] Open
Affiliation(s)
- Jin Mo Park
- Department of Neurology, Dongguk University College of Medicine, Gyeongju, Korea
| | - Hisahide Nishio
- Department of Community Medicine and Social Healthcare Science, Kobe Univiersity Graduate School of Medicine, Kobe, Japan
| | - Jin Hong Shin
- Department of Neurology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Jin Sung Park
- Department of Neurology, School of Medicine, Kyungpook National University, Daegu, Korea.,Department of Neurology, Kyungpook National University Chilgok Hospital, Daegu, Korea.
| |
Collapse
|
31
|
Ruhno C, McGovern VL, Avenarius MR, Snyder PJ, Prior TW, Nery FC, Muhtaseb A, Roggenbuck JS, Kissel JT, Sansone VA, Siranosian JJ, Johnstone AJ, Nwe PH, Zhang RZ, Swoboda KJ, Burghes AHM. Complete sequencing of the SMN2 gene in SMA patients detects SMN gene deletion junctions and variants in SMN2 that modify the SMA phenotype. Hum Genet 2019; 138:241-256. [PMID: 30788592 PMCID: PMC6503527 DOI: 10.1007/s00439-019-01983-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/14/2019] [Indexed: 12/11/2022]
Abstract
Spinal muscular atrophy (SMA) is a progressive motor neuron disease caused by loss or mutation of the survival motor neuron 1 (SMN1) gene and retention of SMN2. We performed targeted capture and sequencing of the SMN2, CFTR, and PLS3 genes in 217 SMA patients. We identified a 6.3 kilobase deletion that occurred in both SMN1 and SMN2 (SMN1/2) and removed exons 7 and 8. The deletion junction was flanked by a 21 bp repeat that occurred 15 times in the SMN1/2 gene. We screened for its presence in 466 individuals with the known SMN1 and SMN2 copy numbers. In individuals with 1 SMN1 and 0 SMN2 copies, the deletion occurred in 63% of cases. We modeled the deletion junction frequency and determined that the deletion occurred in both SMN1 and SMN2. We have identified the first deletion junction where the deletion removes exons 7 and 8 of SMN1/2. As it occurred in SMN1, it is a pathogenic mutation. We called variants in the PLS3 and SMN2 genes, and tested for association with mild or severe exception patients. The variants A-44G, A-549G, and C-1897T in intron 6 of SMN2 were significantly associated with mild exception patients, but no PLS3 variants correlated with severity. The variants occurred in 14 out of 58 of our mild exception patients, indicating that mild exception patients with an intact SMN2 gene and without modifying variants occur. This sample set can be used in the association analysis of candidate genes outside of SMN2 that modify the SMA phenotype.
Collapse
Affiliation(s)
- Corey Ruhno
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, USA
| | - Vicki L McGovern
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, USA
| | | | - Pamela J Snyder
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Thomas W Prior
- Department of Pathology, Case Western Reserve Medical Center, Cleveland, OH, USA
| | - Flavia C Nery
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Abdurrahman Muhtaseb
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - John T Kissel
- Department of Neurology, The Ohio State University, Columbus, OH, USA
| | | | - Jennifer J Siranosian
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Alec J Johnstone
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Pann H Nwe
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ren Z Zhang
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kathryn J Swoboda
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Arthur H M Burghes
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
32
|
Vill K, Kölbel H, Schwartz O, Blaschek A, Olgemöller B, Harms E, Burggraf S, Röschinger W, Durner J, Gläser D, Nennstiel U, Wirth B, Schara U, Jensen B, Becker M, Hohenfellner K, Müller-Felber W. One Year of Newborn Screening for SMA - Results of a German Pilot Project. J Neuromuscul Dis 2019; 6:503-515. [PMID: 31594245 PMCID: PMC6918901 DOI: 10.3233/jnd-190428] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Spinal muscular atrophy (SMA) is the most common neurodegenerative disease in childhood. The study was conducted to assess the impact of early detection of SMA by newborn screening (NBS) on the clinical course of the disease. METHODS Screening was performed in two federal states of Germany, Bavaria and North Rhine Westphalia, between January 2018 and February 2019. The incidence in the screening population was calculated as number of detected patients with a homozygous deletion in the SMN1-gene per number of screened patients. To get an idea about the incidence of newly diagnosed SMA in the year prior to screening a survey covering all neuropediatric centers in the state of Bavaria was conducted, identifying all SMA-cases in 2017 and 2018. Following positive NBS and confirmatory diagnostic test, treatment was advised according to the recommendations of the "American SMA NBS Multidisciplinary Working Group". Immediate treatment with Nusinersen was recommended in children with 2 and 3 SMN2 copies and a conservative strict follow-up strategy in children with ≥4 copies. All children underwent regular standardized neuropediatric examination, CHOP INTEND and HINE-2 testing as well as electrophysiological exams every 2-3 months. RESULTS 165,525 children were screened. 22 cases of SMA were identified, meaning an incidence rate of 1:7524. SMN2 copy number analysis showed 2 SMN2 copies in 45% of patients, 3 SMN2 copies in 19 % and 4 SMN2 copies in 36%. These findings are confirmed in the most recent statistical data-cut from 31st August 2019 (incidence 1:7089, 2 SMN2 copies in 44%, 3 in 15% and 4 in 38%). Comparison with up-to-date German data on SMA incidence and the Bavarian survey give evidence that NBS did not lead to a relevant increase in incidence. 10 patients with 2 or 3 SMN2 copies were treated with Nusinersen, starting between 15- 39 days after birth, in 7/10 patients before onset of symptoms. Presymptomatically treated patients (age at last examination: 1- 12 months, median 8 months) showed no muscle weakness by the age of one month to one year. One child with 4 SMN2 copies became symptomatic at the age of 8 months. CONCLUSIONS Newborn screening, resulting in presymptomatic treatment, improves outcome in children with genetically proven SMA. Newborn screening for SMA should be introduced in all countries where therapy is available. An immediate therapy in cases with 4 SMN2 copies should be considered.
Collapse
Affiliation(s)
- Katharina Vill
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children’s Hospital, LMU – University of Munich, Munich, Germany
| | - Heike Kölbel
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, University of Essen, Germany
| | - Oliver Schwartz
- Department of Pediatric Neurology, Muenster University Hospital, Münster, Germany
| | - Astrid Blaschek
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children’s Hospital, LMU – University of Munich, Munich, Germany
| | | | - Erik Harms
- Department of Pediatric Medicine, Previously Muenster University Hospital, Münster, Germany
| | | | | | | | - Dieter Gläser
- Genetikum, Center for Human Genetics, Neu-Ulm, Germany
| | - Uta Nennstiel
- Screening Center of the Bavarian Health and Food Safety Authority, Oberschleissheim, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University of Cologne, Cologne, Germany
| | - Ulrike Schara
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, University of Essen, Germany
| | - Beate Jensen
- Department of Pediatric Neurology, Muenster University Hospital, Münster, Germany
| | | | | | - Wolfgang Müller-Felber
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children’s Hospital, LMU – University of Munich, Munich, Germany
| |
Collapse
|
33
|
Rao VK, Kapp D, Schroth M. Gene Therapy for Spinal Muscular Atrophy: An Emerging Treatment Option for a Devastating Disease. J Manag Care Spec Pharm 2018; 24:S3-S16. [PMID: 30582825 PMCID: PMC10408414 DOI: 10.18553/jmcp.2018.24.12-a.s3] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is an autosomal recessive neurodegenerative disease that, in most cases, involves homozygous deletion of the SMN1 gene. This causes a deficiency in survival motor neuron (SMN) protein, which plays a critical role in motor neuron development. SMA has a range of phenotype expression resulting in variable age of symptom onset, maximum motor strength achieved, and survival. Without intervention, infants with a more severe form of the disease (type 1 SMA) die before 2 years of age. Although it is rare, SMA is the most common fatal inherited disease of infancy, and until recently, treatment was primarily supportive. In 2016, a new agent, nusinersen, was approved by the FDA. Other treatments are in development, including a gene therapy, AVXS-101. These treatments are not only improving the lives of patients with SMA and their families, they are changing the disease phenotype. They have the greatest benefit when given early in the disease course. OBJECTIVES To discuss current knowledge about SMA, provide clinical evidence for available and emerging treatment options, and present approaches for adding new therapies to hospital/health system formularies to ensure timely access to newly approved therapies for SMA. SUMMARY Advances in clinical care have significantly extended the lives of individuals with SMA, and research into the genetic mechanisms leading to disease have revealed strategies for intervention that target the underlying cause of SMA. Nusinersen is now on the market, and other treatment options, such as AVXS-101, may soon be approved. This article provides an overview of SMA and the genetic mechanisms leading to SMN deficiency, then describes how new and emerging treatments work to overcome this deficiency and prevent associated nerve damage and disability. In addition, we discuss steps for incorporating AVXS-101 into hospital/health system formularies, along with barriers and concerns that may delay access, based in part on lessons learned with nusinersen.
Collapse
|
34
|
Saffari A, Kölker S, Hoffmann GF, Weiler M, Ziegler A. Novel challenges in spinal muscular atrophy - How to screen and whom to treat? Ann Clin Transl Neurol 2018; 6:197-205. [PMID: 30656198 PMCID: PMC6331314 DOI: 10.1002/acn3.689] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 12/27/2022] Open
Abstract
In recent years, disease‐modifying and life‐prolonging therapies for spinal muscular atrophy (SMA) have been developed. However, patients are currently diagnosed with significant delay and therapies are often administered in advanced stages of motor neuron degeneration, showing limited effects. Methods to identify children in presymptomatic stages are currently evaluated in newborn screening programs. Yet, not all children develop symptoms shortly after birth raising the question whom to treat and when to initiate therapy. Finally, monitoring disease progression becomes essential to individualize management. Here, we review the literature on screening approaches, strategies to predict disease severity, and biomarkers to monitor therapy.
Collapse
Affiliation(s)
- Afshin Saffari
- Division of Child Neurology and Metabolic Medicine Center for Child and Adolescent Medicine University Hospital Heidelberg Heidelberg Germany
| | - Stefan Kölker
- Division of Child Neurology and Metabolic Medicine Center for Child and Adolescent Medicine University Hospital Heidelberg Heidelberg Germany
| | - Georg F Hoffmann
- Division of Child Neurology and Metabolic Medicine Center for Child and Adolescent Medicine University Hospital Heidelberg Heidelberg Germany
| | - Markus Weiler
- Department of Neurology University Hospital Heidelberg Heidelberg Germany
| | - Andreas Ziegler
- Division of Child Neurology and Metabolic Medicine Center for Child and Adolescent Medicine University Hospital Heidelberg Heidelberg Germany
| |
Collapse
|
35
|
Glascock J, Sampson J, Haidet-Phillips A, Connolly A, Darras B, Day J, Finkel R, Howell RR, Klinger K, Kuntz N, Prior T, Shieh PB, Crawford TO, Kerr D, Jarecki J. Treatment Algorithm for Infants Diagnosed with Spinal Muscular Atrophy through Newborn Screening. J Neuromuscul Dis 2018; 5:145-158. [PMID: 29614695 PMCID: PMC6004919 DOI: 10.3233/jnd-180304] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Spinal muscular atrophy (SMA) is an autosomal recessive disease characterized by the degeneration of alpha motor neurons in the spinal cord, leading to muscular atrophy. SMA is caused by deletions or mutations in the survival motor neuron 1 gene (SMN1). In humans, a nearly identical copy gene, SMN2, is present. Because SMN2 has been shown to decrease disease severity in a dose-dependent manner, SMN2 copy number is predictive of disease severity. Objective: To develop a treatment algorithm for SMA-positive infants identified through newborn screening based upon SMN2 copy number. Methods: A working group comprised of 15 SMA experts participated in a modified Delphi process, moderated by a neutral third-party expert, to develop treatment guidelines. Results: The overarching recommendation is that all infants with two or three copies of SMN2 should receive immediate treatment (n = 13). For those infants in which immediate treatment is not recommended, guidelines were developed that outline the timing and appropriate screens and tests to be used to determine the timing of treatment initiation. Conclusions: The identification SMA affected infants via newborn screening presents an unprecedented opportunity for achievement of maximal therapeutic benefit through the administration of treatment pre-symptomatically. The recommendations provided here are intended to help formulate treatment guidelines for infants who test positive during the newborn screening process.
Collapse
Affiliation(s)
| | | | | | - Anne Connolly
- Washington University School of Medicine, St. Louis, MO, USA
| | - Basil Darras
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - John Day
- Stanford University, Stanford, CA, USA
| | - Richard Finkel
- Nemours Children's Hospital, University of Central Florida College of Medicine, Orlando, FL, USA
| | - R Rodney Howell
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Nancy Kuntz
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Thomas Prior
- Department of Molecular Pathology, Ohio State Wexner Medical Center, Columbus, OH, USA
| | - Perry B Shieh
- University of California Los Angeles, Los Angeles, CA, USA
| | - Thomas O Crawford
- Departments of Neurology and Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | | | | |
Collapse
|
36
|
Tizzano EF, Zafeiriou D. Prenatal aspects in spinal muscular atrophy: From early detection to early presymptomatic intervention. Eur J Paediatr Neurol 2018; 22:944-950. [PMID: 30219357 DOI: 10.1016/j.ejpn.2018.08.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/23/2018] [Accepted: 08/29/2018] [Indexed: 01/22/2023]
Abstract
With the recent advances in spinal muscular atrophy therapies, the complete scenario of standard of care and following up is changing not only in the clinical field with new phenotypes emerging but also with new expectations for patients, caregivers and health providers. The actual evidence indicates that early intervention and treatment is crucial for better response and prognosis. Knowledge of the prenatal and pre-symptomatic postnatal stages of the disease are becoming essential to consider the opportunities of timely diagnosis and to decide the earliest therapeutic intervention.
Collapse
Affiliation(s)
- Eduardo F Tizzano
- Department of Clinical and Molecular Genetics and Rare Diseases Unit and Medicine Genetics Group, VHIR, Hospital Valle Hebron, Barcelona, Spain; CIBERER, Barcelona, Spain.
| | - Dimitrios Zafeiriou
- 1st Department of Pediatrics, "Hippokratio" General Hospital, Aristotle University, Thessaloniki, Greece.
| |
Collapse
|
37
|
Maretina MA, Zheleznyakova GY, Lanko KM, Egorova AA, Baranov VS, Kiselev AV. Molecular Factors Involved in Spinal Muscular Atrophy Pathways as Possible Disease-modifying Candidates. Curr Genomics 2018; 19:339-355. [PMID: 30065610 PMCID: PMC6030859 DOI: 10.2174/1389202919666180101154916] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 01/07/2023] Open
Abstract
Spinal Muscular Atrophy (SMA) is a neuromuscular disorder caused by mutations in the SMN1 gene. Being a monogenic disease, it is characterized by high clinical heterogeneity. Variations in penetrance and severity of symptoms, as well as clinical discrepancies between affected family members can result from modifier genes influence on disease manifestation. SMN2 gene copy number is known to be the main phenotype modifier and there is growing evidence of additional factors contributing to SMA severity. Potential modifiers of spinal muscular atrophy can be found among the wide variety of different factors, such as multiple proteins interacting with SMN or promoting motor neuron survival, epigenetic modifications, transcriptional or splicing factors influencing SMN2 expression. Study of these factors enables to reveal mechanisms underlying SMA pathology and can have pronounced clinical application.
Collapse
Affiliation(s)
- Marianna A. Maretina
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya line, 3, Saint Petersburg199034, Russia
- Saint Petersburg State University, Universitetskaya emb. 7/9, 199034Saint Petersburg, Russia
| | - Galina Y. Zheleznyakova
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska Universitetssjukhuset, 171 76 Stockholm, Sweden
| | - Kristina M. Lanko
- Saint Petersburg State Institute of Technology, Moskovsky prospect, 26, Saint Petersburg190013, Russia
| | - Anna A. Egorova
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya line, 3, Saint Petersburg199034, Russia
| | - Vladislav S. Baranov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya line, 3, Saint Petersburg199034, Russia
- Saint Petersburg State University, Universitetskaya emb. 7/9, 199034Saint Petersburg, Russia
| | - Anton V. Kiselev
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya line, 3, Saint Petersburg199034, Russia
| |
Collapse
|
38
|
Lin X, Li JJ, Qian WJ, Zhang QJ, Wang ZF, Lu YQ, Dong EL, He J, Wang N, Ma LX, Chen WJ. Modeling the differential phenotypes of spinal muscular atrophy with high-yield generation of motor neurons from human induced pluripotent stem cells. Oncotarget 2018; 8:42030-42042. [PMID: 28159932 PMCID: PMC5522047 DOI: 10.18632/oncotarget.14925] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/27/2016] [Indexed: 12/14/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a devastating motor neuron disease caused by mutations of the survival motor neuron 1 (SMN1) gene. SMN2, a paralogous gene to SMN1, can partially compensate for the loss of SMN1. On the basis of age at onset, highest motor function and SMN2 copy numbers, childhood-onset SMA can be divided into three types (SMA I-III). An inverse correlation was observed between SMN2 copies and the differential phenotypes of SMA. Interestingly, this correlation is not always absolute. Using SMA induced pluripotent stem cells (iPSCs), we found that the SMN was significantly decreased in both SMA III and SMA I iPSCs derived postmitotic motor neurons (pMNs) and γ-aminobutyric acid (GABA) neurons. Moreover, the significant differences of SMN expression level between SMA III (3 copies of SMN2) and SMA I (2 copies of SMN2) were observed only in pMNs culture, but not in GABA neurons or iPSCs. From these findings, we further discovered that the neurite outgrowth was suppressed in both SMA III and SMA I derived MNs. Meanwhile, the significant difference of neurite outgrowth between SMA III and SMA I group was also found in long-term cultures. However, significant hyperexcitability was showed only in SMA I derived mature MNs, but not in SMA III group. Above all, we propose that SMN protein is a major factor of phenotypic modifier. Our data may provide a new insight into recognition for differential phenotypes of SMA disease.
Collapse
Affiliation(s)
- Xiang Lin
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Jin-Jing Li
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Wen-Jing Qian
- Institutes of Brain Science, Institute of Neurobiology, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Qi-Jie Zhang
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Zhong-Feng Wang
- Institutes of Brain Science, Institute of Neurobiology, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Ying-Qian Lu
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - En-Lin Dong
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Jin He
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Ning Wang
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Li-Xiang Ma
- Department of Anatomy, Histology & Embryology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wan-Jin Chen
- Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China.,Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
39
|
Correlation between SMA type and SMN2 copy number revisited: An analysis of 625 unrelated Spanish patients and a compilation of 2834 reported cases. Neuromuscul Disord 2018; 28:208-215. [PMID: 29433793 DOI: 10.1016/j.nmd.2018.01.003] [Citation(s) in RCA: 302] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 11/30/2017] [Accepted: 01/07/2018] [Indexed: 01/01/2023]
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by loss or mutations in SMN1. According to age of onset, achieved motor abilities, and life span, SMA patients are classified into type I (never sit), II (never walk unaided) or III (achieve independent walking abilities). SMN2, the highly homologous copy of SMN1, is considered the most important phenotypic modifier of the disease. Determination of SMN2 copy number is essential to establish careful genotype-phenotype correlations, predict disease evolution, and to stratify patients for clinical trials. We have determined SMN2 copy numbers in 625 unrelated Spanish SMA patients with loss or mutation of both copies of SMN1 and a clear assignation of the SMA type by clinical criteria. Furthermore, we compiled data from relevant worldwide reports that link SMN2 copy number with SMA severity published from 1999 to date (2834 patients with different ethnic and geographic backgrounds). Altogether, we have assembled a database with a total of 3459 patients to delineate more universal prognostic rules regarding the influence of SMN2 copy number on SMA phenotype. This issue is crucial in the present scenario of therapeutic advances with the perspective of SMA neonatal screening and early diagnosis to initiate treatments.
Collapse
|
40
|
Abstract
Despite significant advances in basic research, the treatment of degenerative diseases of the nervous system remains one of the greatest challenges for translational medicine. The childhood onset motor neuron disorder spinal muscular atrophy (SMA) has been viewed as one of the more tractable targets for molecular therapy due to a detailed understanding of the molecular genetic basis of the disease. In SMA, inactivating mutations in the SMN1 gene can be partially compensated for by limited expression of SMN protein from a variable number of copies of the SMN2 gene, which provides both a molecular explanation for phenotypic severity and a target for therapy. The advent of the first tailored molecular therapy for SMA, based on modulating the splicing behaviour of the SMN2 gene provides, for the first time, a treatment which alters the natural history of motor neuron degeneration. Here we consider how this will change the landscape for diagnosis, clinical management and future therapeutic trials in SMA, as well as the implications for the molecular therapy of other neurological diseases.
Collapse
Affiliation(s)
- K Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - E F Tizzano
- Department of Clinical and Molecular Genetics, Rare Diseases Unit, Hospital Valle Hebron, Barcelona, Spain.,CIBERER, Barcelona, Spain
| |
Collapse
|
41
|
Tizzano EF, Finkel RS. Spinal muscular atrophy: A changing phenotype beyond the clinical trials. Neuromuscul Disord 2017; 27:883-889. [PMID: 28757001 DOI: 10.1016/j.nmd.2017.05.011] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/03/2017] [Accepted: 05/11/2017] [Indexed: 01/14/2023]
Abstract
Spinal muscular atrophy is a monogenic, progressive motor neuron disorder caused by deletion or mutation in the SMN1 gene. A broad range of phenotypic severity, from very weak infants (Type 1) to ambulant children (type 3), is modified mainly by the number of copies of the "backup" SMN2 gene. Since the discovery of the role of both genes, basic research into the pathobiology of SMA, with in vitro and animal model studies, has identified therapeutic targets. Development of clinical outcome measures, natural history studies and standard of care guidelines have contributed to the development of protocols for therapeutic drugs now under clinical investigation. Following regulatory approval of the first drug treatment for SMA in the US (December, 2016) and marketing authorization in Europe (June, 2017), the prospects for care of these patients have changed. The evolution of the phenotype of SMA now needs to be considered beyond the clinical trials. This perspective review discusses potential new trajectories in the phenotype of SMA and the need for multidisciplinary teams to prepare for this changing landscape.
Collapse
Affiliation(s)
- Eduardo F Tizzano
- Department of Clinical and Molecular Genetics and Rare Disease Unit, CIBERER, Hospital Valle Hebron, Barcelona, Spain
| | - Richard S Finkel
- Nemours Children's Hospital, Orlando, FL, USA; University of Central Florida College of Medicine, Orlando, FL, USA.
| |
Collapse
|
42
|
Gender-Specific Amelioration of SMA Phenotype upon Disruption of a Deep Intronic Structure by an Oligonucleotide. Mol Ther 2017; 25:1328-1341. [PMID: 28412171 DOI: 10.1016/j.ymthe.2017.03.036] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/18/2017] [Accepted: 03/28/2017] [Indexed: 01/01/2023] Open
Abstract
Spinal muscular atrophy (SMA), the leading genetic disease of children, is caused by low levels of survival motor neuron (SMN) protein. Here, we employ A15/283, an antisense oligonucleotide targeting a deep intronic sequence/structure, to examine the impact of restoration of SMN in a mild SMA mouse model. We show gender-specific amelioration of tail necrosis upon subcutaneous administrations of A15/283 into SMA mice at postnatal days 1 and 3. We also demonstrate that a modest increase in SMN due to early administrations of A15/283 dramatically improves testicular development and spermatogenesis. Our results reveal near total correction of expression of several genes in adult testis upon temporary increase in SMN during early postnatal development. This is the first demonstration of in vivo efficacy of an antisense oligonucleotide targeting a deep intronic sequence/structure. This is also the first report of gender-specific amelioration of SMA pathology upon a modest peripheral increase of SMN.
Collapse
|
43
|
Medrano S, Monges S, Gravina LP, Alías L, Mozzoni J, Aráoz HV, Bernal S, Moresco A, Chertkoff L, Tizzano E. Genotype-phenotype correlation of SMN locus genes in spinal muscular atrophy children from Argentina. Eur J Paediatr Neurol 2016; 20:910-917. [PMID: 27510309 DOI: 10.1016/j.ejpn.2016.07.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 06/08/2016] [Accepted: 07/19/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND/PURPOSE Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder, considered one of the leading causes of infant mortality. It is caused by mutations in the SMN1 gene. A highly homologous copy of this gene named SMN2 and other neighbouring genes, SERF1A and NAIP, are considered phenotypic modifiers of the disease. In recent years, notable advances have been made in SMA research regarding evaluation, prognosis, and therapeutic options. Thus, genotype-phenotype studies in SMA are important to stratify patients for motor function tests and for envisaged clinical trials. The aim of this study was to provide clinical and molecular data of a series of Argentinean children with SMA to establish a comprehensive genotype-phenotype correlation. METHODS 144 Argentinean children with SMA (56 children with type I, 58 with type II, and 30 with type III) were evaluated. The copy number of SMN2, SERF1A, and NAIP genes was established using MLPA (Multiplex Ligation-dependent Probe Amplification) and then correlated with the patients clinical subtypes. To improve clinical characterization we considered the initial symptoms that prompted the consultation, age of acquisition of motor abilities to independent walking and age at loss of gait. We also evaluated clinical and molecular features of sibling pairs in seven families. RESULTS A strong correlation was observed between the SMN2 copy number and SMA phenotype while SERF1A and NAIP copy number showed a moderate correlation. We observed intra- and inter-family differences among the SMA types. CONCLUSION This first genotype-phenotype correlation study in Argentinean SMA children provides data to improve patient stratification and define more adequate follow-up parameters.
Collapse
Affiliation(s)
- Sofía Medrano
- Laboratorio de Biología Molecular, Servicio de Genética, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | - Soledad Monges
- Servicio de Neurología, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | - Luis Pablo Gravina
- Laboratorio de Biología Molecular, Servicio de Genética, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | - Laura Alías
- Servicio de Genética, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; CIBERER U-705, Barcelona, Spain
| | - Julieta Mozzoni
- Servicio de Kinesiología, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | - Hilda Verónica Aráoz
- Laboratorio de Biología Molecular, Servicio de Genética, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | - Sara Bernal
- Servicio de Genética, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; CIBERER U-705, Barcelona, Spain
| | - Angélica Moresco
- Servicio de Genética, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | - Lilien Chertkoff
- Laboratorio de Biología Molecular, Servicio de Genética, Hospital de Pediatría Garrahan, Buenos Aires, Argentina
| | - Eduardo Tizzano
- Department of Clinical and Molecular Genetics, Hospital Valle Hebron, Barcelona, Spain; CIBERER U-705, Barcelona, Spain.
| |
Collapse
|
44
|
Qu YJ, Bai JL, Cao YY, Wang H, Jin YW, Du J, Ge XS, Zhang WH, Li Y, He SX, Song F. Mutation Spectrum of the Survival of Motor Neuron 1 and Functional Analysis of Variants in Chinese Spinal Muscular Atrophy. J Mol Diagn 2016; 18:741-752. [PMID: 27425821 DOI: 10.1016/j.jmoldx.2016.05.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 04/22/2016] [Accepted: 05/10/2016] [Indexed: 11/28/2022] Open
Abstract
Proximal spinal muscular atrophy (SMA) is a common fatal autosomal recessive disorder caused by deletion or mutation of the survival of motor neuron 1 (SMN1). Here, we studied SMA molecular pathology in 653 Chinese patients and found approximately 88.2% with homozygous SMN1 exon 7 deletion and 6.3% with heterozygous exon 7 loss using multiplex ligation-dependent probe amplification. SMN1 variants were detected in 34 patients with heterozygous SMN1 loss by clone sequencing. In 27 of them, 15 variants were identified: five were unreported novel variants [c.-7_9del(p.0), p.Tyr109Cys, p.Ile249Tyrfs*16, p.Tyr272Trpfs*35, and c.835-5T>G], five were previously found only in Chinese patients (p.Ser8Lysfs*23, p.Gln14*, p.Val19Glyfs*21, p.Leu228*, and p.Tyr277Cys), and five were reported in other populations [p.Ala2Gly, p.Gln15*, p.Glu134Lys, p.Ser230Leu, and c.863G>T (r.835_*3del, p.Gly279Glufs*5)]. Variants p.Ser8Lysfs*23 and p.Leu228* were the most common in Chinese SMA. Five variants (p.Ser8Lysfs*23, p.Gln14*, p.Gln15*, p.Val19Glyfs*21, and p.Leu228*) resulted in premature stop codons, likely causing SMN1 mRNA nonsense-mediated decay. The novel variant c.-7_9del (p.0) caused deletion of the translation start codon (AUG), resulting in full-length SMN protein loss. The novel variant c.835-5T>G, located in a splice site, resulted in 90% exon 7 skipping. Our study could facilitate early diagnosis for SMA patients in mutation detection and revealed the specific mutation spectrum of SMN1 in Chinese SMA and high genetic heterogeneity in subtle variants observed between patients from China and Caucasians.
Collapse
Affiliation(s)
- Yu-Jin Qu
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China
| | - Jin-Li Bai
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China
| | - Yan-Yan Cao
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China
| | - Hong Wang
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China
| | - Yu-Wei Jin
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China
| | - Juan Du
- Children's Hospital Affiliated Capital Institute of Pediatrics, Beijing, China
| | - Xiu-Shan Ge
- Children's Hospital Affiliated Capital Institute of Pediatrics, Beijing, China
| | - Wen-Hui Zhang
- Children's Hospital Affiliated Capital Institute of Pediatrics, Beijing, China
| | - Yan Li
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China
| | - Sheng-Xi He
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China
| | - Fang Song
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, China.
| |
Collapse
|
45
|
Qu YJ, Bai JL, Cao YY, Zhang WH, Wang H, Jin YW, Song F. A rare variant (c.863G>T) in exon 7 of SMN1 disrupts mRNA splicing and is responsible for spinal muscular atrophy. Eur J Hum Genet 2016; 24:864-70. [PMID: 26419278 PMCID: PMC4867452 DOI: 10.1038/ejhg.2015.213] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 08/10/2015] [Accepted: 08/25/2015] [Indexed: 11/08/2022] Open
Abstract
Proximal spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder caused by deletion or mutation of SMN1 (survival motor neuron 1). SMN exon 7 splicing is regulated by a number of exonic and intronic regulatory sequences and the trans-factors that bind them. Variants located in or near these regulated regions should be evaluated to determine their effect on splicing. We identified the rare variant c.863G>T (r.835_*3del, p.Gly279Glufs*5) in exon 7 of SMN1 in three patients affected with type I or type II SMA. Most of the SMN1 transcripts exhibited complete loss of exon 7 in vivo. The ex vivo splicing assay demonstrated that the variant disrupts inclusion of exon 7 (~85%) in the SMN1 mRNA; replacement with various bases yielded a variety of splicing effects in SMN1 and SMN2 pre-mRNA. The c.863G>T (r.835_*3del, p.Gly279Glufs*5) variant is located in a region that includes binding sites for multiple splicing factors including Tra2β1. Thus, the variant disrupts Tra2β1 binding, but does not affect binding of hnRNP A1. These findings demonstrate how rare variants influence pre-mRNA splicing of SMN and reveal the functional influence of c.863G>T (r.835_*3del, p.Gly279Glufs*5) variant in patients with SMA.
Collapse
Affiliation(s)
- Yu-jin Qu
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P.R. China
| | - Jin-li Bai
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P.R. China
| | - Yan-yan Cao
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P.R. China
| | - Wen-hui Zhang
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P.R. China
| | - Hong Wang
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P.R. China
| | - Yu-wei Jin
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P.R. China
| | - Fang Song
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P.R. China
| |
Collapse
|
46
|
Ahmad S, Bhatia K, Kannan A, Gangwani L. Molecular Mechanisms of Neurodegeneration in Spinal Muscular Atrophy. J Exp Neurosci 2016; 10:39-49. [PMID: 27042141 PMCID: PMC4807884 DOI: 10.4137/jen.s33122] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 02/07/2023] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disease with a high incidence and is the most common genetic cause of infant mortality. SMA is primarily characterized by degeneration of the spinal motor neurons that leads to skeletal muscle atrophy followed by symmetric limb paralysis, respiratory failure, and death. In humans, mutation of the Survival Motor Neuron 1 (SMN1) gene shifts the load of expression of SMN protein to the SMN2 gene that produces low levels of full-length SMN protein because of alternative splicing, which are sufficient for embryonic development and survival but result in SMA. The molecular mechanisms of the (a) regulation of SMN gene expression and (b) degeneration of motor neurons caused by low levels of SMN are unclear. However, some progress has been made in recent years that have provided new insights into understanding of the cellular and molecular basis of SMA pathogenesis. In this review, we have briefly summarized recent advances toward understanding of the molecular mechanisms of regulation of SMN levels and signaling mechanisms that mediate neurodegeneration in SMA.
Collapse
Affiliation(s)
- Saif Ahmad
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.; Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Kanchan Bhatia
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.; Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Annapoorna Kannan
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.; Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Laxman Gangwani
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center, El Paso, Texas, USA.; Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| |
Collapse
|
47
|
Butchbach MER. Copy Number Variations in the Survival Motor Neuron Genes: Implications for Spinal Muscular Atrophy and Other Neurodegenerative Diseases. Front Mol Biosci 2016; 3:7. [PMID: 27014701 PMCID: PMC4785180 DOI: 10.3389/fmolb.2016.00007] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/25/2016] [Indexed: 12/11/2022] Open
Abstract
Proximal spinal muscular atrophy (SMA), a leading genetic cause of infant death worldwide, is an early-onset, autosomal recessive neurodegenerative disease characterized by the loss of spinal α-motor neurons. This loss of α-motor neurons is associated with muscle weakness and atrophy. SMA can be classified into five clinical grades based on age of onset and severity of the disease. Regardless of clinical grade, proximal SMA results from the loss or mutation of SMN1 (survival motor neuron 1) on chromosome 5q13. In humans a large tandem chromosomal duplication has lead to a second copy of the SMN gene locus known as SMN2. SMN2 is distinguishable from SMN1 by a single nucleotide difference that disrupts an exonic splice enhancer in exon 7. As a result, most of SMN2 mRNAs lack exon 7 (SMNΔ7) and produce a protein that is both unstable and less than fully functional. Although only 10–20% of the SMN2 gene product is fully functional, increased genomic copies of SMN2 inversely correlates with disease severity among individuals with SMA. Because SMN2 copy number influences disease severity in SMA, there is prognostic value in accurate measurement of SMN2 copy number from patients being evaluated for SMA. This prognostic value is especially important given that SMN2 copy number is now being used as an inclusion criterion for SMA clinical trials. In addition to SMA, copy number variations (CNVs) in the SMN genes can affect the clinical severity of other neurological disorders including amyotrophic lateral sclerosis (ALS) and progressive muscular atrophy (PMA). This review will discuss how SMN1 and SMN2 CNVs are detected and why accurate measurement of SMN1 and SMN2 copy numbers is relevant for SMA and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Matthew E R Butchbach
- Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for ChildrenWilmington, DE, USA; Center for Pediatric Research, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for ChildrenWilmington, DE, USA; Department of Biological Sciences, University of DelawareNewark, DE, USA; Department of Pediatrics, Thomas Jefferson UniversityPhiladelphia, PA, USA
| |
Collapse
|
48
|
Decay in survival motor neuron and plastin 3 levels during differentiation of iPSC-derived human motor neurons. Sci Rep 2015; 5:11696. [PMID: 26114395 PMCID: PMC4650562 DOI: 10.1038/srep11696] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/30/2015] [Indexed: 11/08/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disease caused by mutations in Survival Motor Neuron 1 (SMN1), leading to degeneration of alpha motor neurons (MNs) but also affecting other cell types. Induced pluripotent stem cell (iPSC)-derived human MN models from severe SMA patients have shown relevant phenotypes. We have produced and fully characterized iPSCs from members of a discordant consanguineous family with chronic SMA. We differentiated the iPSC clones into ISL-1+/ChAT+ MNs and performed a comparative study during the differentiation process, observing significant differences in neurite length and number between family members. Analyses of samples from wild-type, severe SMA type I and the type IIIa/IV family showed a progressive decay in SMN protein levels during iPSC-MN differentiation, recapitulating previous observations in developmental studies. PLS3 underwent parallel reductions at both the transcriptional and translational levels. The underlying, progressive developmental decay in SMN and PLS3 levels may lead to the increased vulnerability of MNs in SMA disease. Measurements of SMN and PLS3 transcript and protein levels in iPSC-derived MNs show limited value as SMA biomarkers.
Collapse
|
49
|
Zheleznyakova GY, Nilsson EK, Kiselev AV, Maretina MA, Tishchenko LI, Fredriksson R, Baranov VS, Schiöth HB. Methylation levels of SLC23A2 and NCOR2 genes correlate with spinal muscular atrophy severity. PLoS One 2015; 10:e0121964. [PMID: 25821969 PMCID: PMC4378931 DOI: 10.1371/journal.pone.0121964] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 02/09/2015] [Indexed: 11/19/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a monogenic neurodegenerative disorder subdivided into four different types. Whole genome methylation analysis revealed 40 CpG sites associated with genes that are significantly differentially methylated between SMA patients and healthy individuals of the same age. To investigate the contribution of methylation changes to SMA severity, we compared the methylation level of found CpG sites, designed as "targets", as well as the nearest CpG sites in regulatory regions of ARHGAP22, CDK2AP1, CHML, NCOR2, SLC23A2 and RPL9 in three groups of SMA patients. Of notable interest, compared to type I SMA male patients, the methylation level of a target CpG site and one nearby CpG site belonging to the 5'UTR of SLC23A2 were significantly hypomethylated 19-22% in type III-IV patients. In contrast to type I SMA male patients, type III-IV patients demonstrated a 16% decrease in the methylation levels of a target CpG site, belonging to the 5'UTR of NCOR2. To conclude, this study validates the data of our previous study and confirms significant methylation changes in the SLC23A2 and NCOR2 regulatory regions correlates with SMA severity.
Collapse
Affiliation(s)
- Galina Yu. Zheleznyakova
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
- Faculty of Biology, Saint-Petersburg State University, Saint-Petersburg, Russia
- Laboratory for Prenatal Diagnostics of Inherited Diseases, D.O. Ott Research Institute of Obstetrics and Gynecology RAMS, Saint-Petersburg, Russia
- * E-mail:
| | - Emil K. Nilsson
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Anton V. Kiselev
- Laboratory for Prenatal Diagnostics of Inherited Diseases, D.O. Ott Research Institute of Obstetrics and Gynecology RAMS, Saint-Petersburg, Russia
| | - Marianna A. Maretina
- Faculty of Biology, Saint-Petersburg State University, Saint-Petersburg, Russia
- Laboratory for Prenatal Diagnostics of Inherited Diseases, D.O. Ott Research Institute of Obstetrics and Gynecology RAMS, Saint-Petersburg, Russia
| | | | | | - Vladislav S. Baranov
- Faculty of Biology, Saint-Petersburg State University, Saint-Petersburg, Russia
- Laboratory for Prenatal Diagnostics of Inherited Diseases, D.O. Ott Research Institute of Obstetrics and Gynecology RAMS, Saint-Petersburg, Russia
| | | |
Collapse
|
50
|
Qu YJ, Ge XS, Bai JL, Wang LW, Cao YY, Lu YY, Jin YW, Wang H, Song F. Association of copy numbers of survival motor neuron gene 2 and neuronal apoptosis inhibitory protein gene with the natural history in a Chinese spinal muscular atrophy cohort. J Child Neurol 2015; 30:429-36. [PMID: 25330799 DOI: 10.1177/0883073814553271] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 09/07/2014] [Indexed: 11/16/2022]
Abstract
We evaluated survival motor neuron 2 (SMN2) and neuronal apoptosis inhibitory protein (NAIP) gene copy distribution and the association of copy number with survival in 232 Chinese spinal muscular atrophy (SMA) patients. The SMN2 and NAIP copy numbers correlated positively with the median onset age (r = 0.72 and 0.377). The risk of death for patients with fewer copies of SMN2 or NAIP was much higher than for those with more copies (P < .01). The survival probabilities at 5 years were 5.1%, 90.7%, and 100% for 2, 3, and 4 SMN2 copies and 27.9%, 66.7%, and 87.2% for 0, 1, and 2 NAIP copies, respectively. Our results indicated that combined SMN1-SMN2-NAIP genotypes with fewer copies were associated with earlier onset age and poorer survival probability. Better survival status for Chinese type I SMA might due to a higher proportion of 3 SMN2 and a lower rate of zero NAIP.
Collapse
Affiliation(s)
- Yu-jin Qu
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P. R. China
| | - Xiu-shan Ge
- Department of Neurology, Children's Hospital Affiliated Capital Institute of Pediatrics, Beijing, P. R. China
| | - Jin-li Bai
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P. R. China
| | - Li-wen Wang
- Department of Neurology, Children's Hospital Affiliated Capital Institute of Pediatrics, Beijing, P. R. China
| | - Yan-yan Cao
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P. R. China
| | - Yan-yu Lu
- Department of Health Development, Capital Institute of Pediatrics, Beijing, P. R. China
| | - Yu-wei Jin
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P. R. China
| | - Hong Wang
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P. R. China
| | - Fang Song
- Department of Medical Genetics, Capital Institute of Pediatrics, Beijing, P. R. China
| |
Collapse
|