1
|
Mangione CC, Frank A, Dalgard CL, Burnett BG, Flagg TP. Transcriptional reprogramming in SMA mouse hearts reveals signatures of early heart failure and dysregulated calcium signaling. Hum Mol Genet 2025:ddaf060. [PMID: 40287831 DOI: 10.1093/hmg/ddaf060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Spinal muscular atrophy (SMA) is an inherited neurodegenerative disease that leads to loss of motor neurons in the anterior horn of the spinal cord with consequent muscle atrophy. SMA results from the functional deletions of the SMN1 gene, resulting in insufficient production of the survival motor neuron (SMN) protein. It is not known why lower motor neurons are particularly sensitive to the loss of SMN function, but it is increasingly apparent that extraneuronal tissues, such as cardiac and skeletal muscle, are also affected by SMN deficiency. We have previously shown that SMN deficiency in a mouse model of spinal muscular atrophy (SMNΔ7) impairs cardiomyocyte contraction and Ca2+ handling. In this study, we performed a comparative total mRNA sequencing analysis of whole hearts isolated at an early (P5) or late (P10) stage of the disease process to investigate the mechanisms contributing to cardiac pathology in SMA. The results demonstrate transcriptional signatures consistent with heart failure, dysregulation of Ca2+ signaling, and hypoxia induced changes occurring as early as P5 and persisting through P10. Similar transcriptomic changes in skeletal muscle tissue indicate that there are likely common, cell autonomous molecular mechanisms resulting in both cardiac and skeletal muscle due to SMN deficiency. The identification of these common themes suggests a link underlying the mechanism of neuronal and non-neuronal deficits in SMA.
Collapse
Affiliation(s)
- Cecelia C Mangione
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD, 20814, United States
| | - Andrew Frank
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD, 20814, United States
| | - Clifton L Dalgard
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD, 20814, United States
| | - Barrington G Burnett
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD, 20814, United States
| | - Thomas P Flagg
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, MD, 20814, United States
| |
Collapse
|
2
|
Nair NN, Kline RA, Boyd I, Anikumar M, Thomson A, Lamont DJ, Gray GA, Wishart TM, Murray LM. Alterations in cardiac function correlate with a disruption in fatty acid metabolism in a mouse model of SMA. Hum Mol Genet 2025; 34:547-562. [PMID: 39810393 PMCID: PMC11891873 DOI: 10.1093/hmg/ddaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
Spinal Muscular Atrophy is an autosomal dominant disease caused by mutations and deletions within the SMN1 gene, with predominantly childhood onset. Although primarily a motor neuron disease, defects in non-neuronal tissues are described in both patients and mouse models. Here, we have undertaken a detailed study of the heart in the Smn2B/- mouse models of SMA, and reveal a thinning of the ventriclar walls as previously described in more severe mouse models of SMA. However most structural changes are resolved by accounting for the smaller body size of the SMA mouse, as was also confirmed in the SMN∆7 model. Echocardiography revealed increased systolic function, which was particularly pronounced in subsets of mice and an increase in global longitudinal strain, collectively indicative of increased cardiac stress in the Smn2B/- mouse model. We have used TMT proteomics to perform a longitudinal study of the proteome of the hearts of Smn2B/- mice and reveal a progressive dysregulation of LXR/RXR signalling which is a regulator of lipid metabolism. We further show consistent perturbations in lipid metabolism in the Smn2B/-, Smn-/-;SMN2;SmnΔ7and SmnΔ7/Δ7;SMN2 mouse models of SMA on the day of birth. This work indicates that although structural changes in the heart can be overstated by failing to account for body size, there are functional defects which could predispose the heart to subsequent failure. We identify a common molecular signature across mouse models pointing to a dysregulation in lipid metabolism, and suggest that manipulation of LXR/RXR signalling offers an opportunity to impact upon these pathways.
Collapse
Affiliation(s)
- Nithya N Nair
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Euan McDonald Centre for Motor Neuron Disease Research, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Rachel A Kline
- Euan McDonald Centre for Motor Neuron Disease Research, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, United Kingdom
| | - Imogen Boyd
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Meenakshi Anikumar
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Adrian Thomson
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, 47 Little France Crescent, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Douglas J Lamont
- FingerPrints Proteomics Facility, School of Life Sciences, Dow Street, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Gillian A Gray
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, 47 Little France Crescent, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Thomas M Wishart
- Euan McDonald Centre for Motor Neuron Disease Research, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, United Kingdom
- Centre for Systems Health and Integrated Metabolic Research, Clifton Boulevard, Nottingham Trent University, Nottingham NG1 4GG, United Kingdom
| | - Lyndsay M Murray
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
- Euan McDonald Centre for Motor Neuron Disease Research, Hugh Robson Building, George Square, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| |
Collapse
|
3
|
René CA, Parks RJ. Extracellular vesicles efficiently deliver survival motor neuron protein to cells in culture. Sci Rep 2025; 15:5674. [PMID: 39955442 PMCID: PMC11830090 DOI: 10.1038/s41598-025-90083-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
Spinal Muscular Atrophy (SMA) is a genetic neuromuscular disorder caused by homozygous mutation or deletion of the survival motor neuron 1 (SMN1) gene, leading to a low quantity of SMN protein in cells. This depletion of SMN protein preferentially leads to death of motor neurons and, consequently, muscle atrophy, in addition to defects in many other peripheral tissues. SMN protein is naturally loaded into extracellular vesicles (EVs), which are sub-micron-sized, membrane-bound particles released from all cell types. The innate ability of EVs to deliver cargo to recipient cells has caused these vesicles to gain interest as therapeutic delivery vehicles. In this study, we show that adenovirus-mediated overexpression of SMN protein in HepG2 cells leads to the release of EVs loaded with high levels of SMN protein into conditioned medium. Application of this medium to recipient cells in tissue culture led to uptake of the SMN protein, which subsequently transited to the nucleus and co-localized with Gemin2 protein, forming nuclear gem-like structures similar to the native SMN protein. Overall, this work demonstrates that SMN protein can be delivered to cells through EVs, which holds promise as a potential therapy for patients with SMA.
Collapse
Affiliation(s)
- Charlotte A René
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8L1, Canada
- Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON, K1Y 4E9, Canada
| | - Robin J Parks
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8L1, Canada.
- Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON, K1Y 4E9, Canada.
- Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, ON, K1H 8L6, Canada.
| |
Collapse
|
4
|
Fletcher EV, Chalif JI, Rotterman TM, Pagiazitis JG, Alstyne MV, Sivakumar N, Rabinowitz JE, Pellizzoni L, Alvarez FJ, Mentis GZ. Synaptic imbalance and increased inhibition impair motor function in SMA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610545. [PMID: 39257773 PMCID: PMC11383993 DOI: 10.1101/2024.08.30.610545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Movement is executed through the balanced action of excitatory and inhibitory neurotransmission in motor circuits of the spinal cord. Short-term perturbations in one of the two types of transmission are counteracted by homeostatic changes of the opposing type. Prolonged failure to balance excitatory and inhibitory drive results in dysfunction at the single neuron, as well as neuronal network levels. However, whether dysfunction in one or both types of neurotransmission leads to pathogenicity in neurodegenerative diseases characterized by select synaptic deficits is not known. Here, we used mouse genetics, functional assays, morphological methods, and viral-mediated approaches to uncover the pathogenic contribution of unbalanced excitation-inhibition neurotransmission in a mouse model of spinal muscular atrophy (SMA). We show that vulnerable motor circuits in the SMA spinal cord fail to respond homeostatically to the reduction of excitatory drive and instead increase inhibition. This imposes an excessive burden on motor neurons and further restricts their recruitment to activate muscle contraction. Importantly, genetic or pharmacological reduction of inhibitory synaptic drive improves neuronal function and provides behavioural benefit in SMA mice. Our findings identify the lack of excitation-inhibition homeostasis as a major maladaptive mechanism in SMA, by which the combined effects of reduced excitation and increased inhibition diminish the capacity of premotor commands to recruit motor neurons and elicit muscle contractions.
Collapse
Affiliation(s)
- Emily V. Fletcher
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Joshua I. Chalif
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | | | - John G. Pagiazitis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Meaghan Van Alstyne
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Nandhini Sivakumar
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Joseph E. Rabinowitz
- Department of Pharmacology, Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Livio Pellizzoni
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Columbia University, New York, NY, 10032, USA
| | | | - George Z. Mentis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
5
|
Brown SJ, Šoltić D, Synowsky SA, Shirran SL, Chilcott E, Shorrock HK, Gillingwater TH, Yáñez-Muñoz RJ, Schneider B, Bowerman M, Fuller HR. AAV9-mediated SMN gene therapy rescues cardiac desmin but not lamin A/C and elastin dysregulation in Smn2B/- spinal muscular atrophy mice. Hum Mol Genet 2023; 32:2950-2965. [PMID: 37498175 PMCID: PMC10549791 DOI: 10.1093/hmg/ddad121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/27/2023] [Accepted: 07/25/2023] [Indexed: 07/28/2023] Open
Abstract
Structural, functional and molecular cardiac defects have been reported in spinal muscular atrophy (SMA) patients and mouse models. Previous quantitative proteomics analyses demonstrated widespread molecular defects in the severe Taiwanese SMA mouse model. Whether such changes are conserved across different mouse models, including less severe forms of the disease, has yet to be established. Here, using the same high-resolution proteomics approach in the less-severe Smn2B/- SMA mouse model, 277 proteins were found to be differentially abundant at a symptomatic timepoint (post-natal day (P) 18), 50 of which were similarly dysregulated in severe Taiwanese SMA mice. Bioinformatics analysis linked many of the differentially abundant proteins to cardiovascular development and function, with intermediate filaments highlighted as an enriched cellular compartment in both datasets. Lamin A/C was increased in the cardiac tissue, whereas another intermediate filament protein, desmin, was reduced. The extracellular matrix (ECM) protein, elastin, was also robustly decreased in the heart of Smn2B/- mice. AAV9-SMN1-mediated gene therapy rectified low levels of survival motor neuron protein and restored desmin levels in heart tissues of Smn2B/- mice. In contrast, AAV9-SMN1 therapy failed to correct lamin A/C or elastin levels. Intermediate filament proteins and the ECM have key roles in cardiac function and their dysregulation may explain cardiac impairment in SMA, especially since mutations in genes encoding these proteins cause other diseases with cardiac aberration. Cardiac pathology may need to be considered in the long-term care of SMA patients, as it is unclear whether currently available treatments can fully rescue peripheral pathology in SMA.
Collapse
Affiliation(s)
- Sharon J Brown
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Darija Šoltić
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Silvia A Synowsky
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, St Andrews KY16 9ST, UK
| | - Sally L Shirran
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, St Andrews KY16 9ST, UK
| | - Ellie Chilcott
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham, Surrey TW20 0EX, UK
| | - Hannah K Shorrock
- Edinburgh Medical School: Biomedical Sciences, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School: Biomedical Sciences, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Rafael J Yáñez-Muñoz
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham, Surrey TW20 0EX, UK
| | - Bernard Schneider
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Melissa Bowerman
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
- School of Medicine, Keele University, Keele ST5 5BG, UK
| | - Heidi R Fuller
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| |
Collapse
|
6
|
El Khoury M, Biondi O, Bruneteau G, Sapaly D, Bendris S, Bezier C, Clerc Z, Akar EA, Weill L, Eid AA, Charbonnier F. NADPH oxidase 4 inhibition is a complementary therapeutic strategy for spinal muscular atrophy. Front Cell Neurosci 2023; 17:1242828. [PMID: 37780204 PMCID: PMC10536974 DOI: 10.3389/fncel.2023.1242828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Spinal muscular atrophy (SMA) is a fatal neurodegenerative disorder, characterized by motor neuron (MN) degeneration and severe muscular atrophy and caused by Survival of Motor Neuron (SMN) depletion. Therapies aimed at increasing SMN in patients have proven their efficiency in alleviating SMA symptoms but not for all patients. Thus, combinational therapies are warranted. Here, we investigated the involvement of NADPH oxidase 4 (NOX4) in SMA-induced spinal MN death and if the modulation of Nox4 activity could be beneficial for SMA patients. Methods We analysed in the spinal cord of severe type SMA-like mice before and at the disease onset, the level of oxidative stress and Nox4 expression. Then, we tested the effect of Nox4 inhibition by GKT137831/Setanaxib, a drug presently in clinical development, by intrathecal injection on MN survival and motor behaviour. Finally, we tested if GKT137831/Setanaxib could act synergistically with FDA-validated SMN-upregulating treatment (nusinersen). Results We show that NOX4 is overexpressed in SMA and its inhibition by GKT137831/Setanaxib protected spinal MN from SMA-induced degeneration. These improvements were associated with a significant increase in lifespan and motor behaviour of the mice. At the molecular level, GKT137831 activated the pro-survival AKT/CREB signaling pathway, leading to an increase in SMN expression in SMA MNs. Most importantly, we found that the per os administration of GKT137831 acted synergistically with a FDA-validated SMN-upregulating treatment. Conclusion The pharmacological inhibition of NOX4 by GKT137831/Setanaxib is neuroprotector and could represent a complementary therapeutic strategy to fight against SMA.
Collapse
Affiliation(s)
- Mirella El Khoury
- Faculty of Basic and Biomedical Sciences, University Paris Cité & Inserm UMR_S1124, Paris, France
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Olivier Biondi
- Faculty of Basic and Biomedical Sciences, University Paris Cité & Inserm UMR_S1124, Paris, France
| | - Gaelle Bruneteau
- Centre de Recherche en Myologie, UMRS974, Association Institut de Myologie, Sorbonne Université, INSERM, Paris, France
- Département de Neurologie, Centre référent SLA, APHP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Delphine Sapaly
- Faculty of Basic and Biomedical Sciences, University Paris Cité & Inserm UMR_S1124, Paris, France
| | - Sabrina Bendris
- Faculty of Basic and Biomedical Sciences, University Paris Cité & Inserm UMR_S1124, Paris, France
| | - Cynthia Bezier
- Faculty of Basic and Biomedical Sciences, University Paris Cité & Inserm UMR_S1124, Paris, France
| | - Zoé Clerc
- Faculty of Basic and Biomedical Sciences, University Paris Cité & Inserm UMR_S1124, Paris, France
| | - Elias Abi Akar
- Faculty of Basic and Biomedical Sciences, University Paris Cité & Inserm UMR_S1124, Paris, France
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Laure Weill
- Faculty of Basic and Biomedical Sciences, University Paris Cité & Inserm UMR_S1124, Paris, France
| | - Assaad A. Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Frédéric Charbonnier
- Faculty of Basic and Biomedical Sciences, University Paris Cité & Inserm UMR_S1124, Paris, France
| |
Collapse
|
7
|
Signoria I, van der Pol WL, Groen EJN. Innovating spinal muscular atrophy models in the therapeutic era. Dis Model Mech 2023; 16:dmm050352. [PMID: 37787662 PMCID: PMC10565113 DOI: 10.1242/dmm.050352] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a severe, monogenetic, neuromuscular disease. A thorough understanding of its genetic cause and the availability of robust models has led to the development and approval of three gene-targeting therapies. This is a unique and exciting development for the field of neuromuscular diseases, many of which remain untreatable. The development of therapies for SMA not only opens the door to future therapeutic possibilities for other genetic neuromuscular diseases, but also informs us about the limitations of such treatments. For example, treatment response varies widely and, for many patients, significant disability remains. Currently available SMA models best recapitulate the severe types of SMA, and these models are genetically and phenotypically more homogeneous than patients. Furthermore, treating patients is leading to a shift in phenotypes with increased variability in SMA clinical presentation. Therefore, there is a need to generate model systems that better reflect these developments. Here, we will first discuss current animal models of SMA and their limitations. Next, we will discuss the characteristics required to future-proof models to assist the field in the development of additional, novel therapies for SMA.
Collapse
Affiliation(s)
- Ilaria Signoria
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - W. Ludo van der Pol
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Ewout J. N. Groen
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| |
Collapse
|
8
|
Bild W, Vasincu A, Rusu RN, Ababei DC, Stana AB, Stanciu GD, Savu B, Bild V. Impact of the Renin-Angiotensin System on the Pathogeny and Pharmacotherapeutics of Neurodegenerative Diseases. Biomolecules 2022; 12:1429. [PMID: 36291638 PMCID: PMC9599929 DOI: 10.3390/biom12101429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
Brain neurodegenerative diseases (BND) are debilitating conditions that are especially characteristic of a certain period of life and considered major threats to human health. Current treatments are limited, meaning that there is a challenge in developing new options that can efficiently tackle the different components and pathophysiological processes of these conditions. The renin-angiotensin-aldosterone system (RAS) is an endocrine axis with important peripheral physiological functions such as blood pressure and cardiovascular homeostasis, as well as water and sodium balance and systemic vascular resistance-functions which are well-documented. However, recent work has highlighted the paracrine and autocrine functions of RAS in different tissues, including the central nervous system (CNS). It is known that RAS hyperactivation has pro-inflammatory and pro-oxidant effects, thus suggesting that its pharmacological modulation could be used in the management of these conditions. The present paper underlines the involvement of RAS and its components in the pathophysiology of BNDs such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), Huntington's disease (HD), motor neuron disease (MND), and prion disease (PRD), as well as the identification of drugs and pharmacologically active substances that act upon RAS, which could alleviate their symptomatology or evolution, and thus, contribute to novel therapeutic approaches.
Collapse
Affiliation(s)
- Walther Bild
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Center of Biomedical Research of the Romanian Academy, 700506 Iasi, Romania
| | - Alexandru Vasincu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Răzvan-Nicolae Rusu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Daniela-Carmen Ababei
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Aurelian Bogdan Stana
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Gabriela Dumitrița Stanciu
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Bogdan Savu
- Department of Pediatric Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Veronica Bild
- Center of Biomedical Research of the Romanian Academy, 700506 Iasi, Romania
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
9
|
Zilio E, Piano V, Wirth B. Mitochondrial Dysfunction in Spinal Muscular Atrophy. Int J Mol Sci 2022; 23:10878. [PMID: 36142791 PMCID: PMC9503857 DOI: 10.3390/ijms231810878] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a devastating neuromuscular disorder caused by recessive mutations in the SMN1 gene, globally affecting ~8-14 newborns per 100,000. The severity of the disease depends on the residual levels of functional survival of motor neuron protein, SMN. SMN is a ubiquitously expressed RNA binding protein involved in a plethora of cellular processes. In this review, we discuss the effects of SMN loss on mitochondrial functions in the neuronal and muscular systems that are the most affected in patients with spinal muscular atrophy. Our aim is to highlight how mitochondrial defects may contribute to disease progression and how restoring mitochondrial functionality may be a promising approach to develop new therapies. We also collected from previous studies a list of transcripts encoding mitochondrial proteins affected in various SMA models. Moreover, we speculate that in adulthood, when motor neurons require only very low SMN levels, the natural deterioration of mitochondria associated with aging may be a crucial triggering factor for adult spinal muscular atrophy, and this requires particular attention for therapeutic strategies.
Collapse
Affiliation(s)
- Eleonora Zilio
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Valentina Piano
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
10
|
Habets LE, Bartels B, Asselman FL, Hulzebos EHJ, Stegeman DF, Jeneson JAL, van der Pol WL. Motor Unit and Capillary Recruitment During Fatiguing Arm-Cycling Exercise in Spinal Muscular Atrophy Types 3 and 4. J Neuromuscul Dis 2022; 9:397-409. [PMID: 35466947 DOI: 10.3233/jnd-210765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Exercise intolerance is an important impairment in patients with SMA, but little is known about the mechanisms underlying this symptom. OBJECTIVE To investigate if reduced motor unit- and capillary recruitment capacity in patients with SMA contribute to exercise intolerance. METHODS Adolescent and adult patients with SMA types 3 and 4 (n = 15) and age- and gender matched controls (n = 15) performed a maximal upper body exercise test. We applied respiratory gas analyses, non-invasive surface electromyography (sEMG) and continuous wave near-infrared spectroscopy (CW-NIRS) to study oxygen consumption, arm muscle motor unit- and capillary recruitment, respectively. RESULTS Maximal exercise duration was twofold lower (p < 0.001) and work of breathing and ventilation was 1.6- and 1.8-fold higher (p < 0.05) in patients compared to controls, respectively. Regarding motor unit recruitment, we found higher normalized RMS amplitude onset values of sEMG signals from all muscles and the increase in normalized RMS amplitudes was similar in the m. triceps brachii, m. brachioradialis and m. flexor digitorum in SMA compared to controls. Median frequency, onset values were similar in patients and controls. We found a similar decrease in median frequencies of sEMG recordings from the m. biceps brachii, a diminished decrease from the m. brachioradialis and m. flexor digitorum, but a larger decrease from the m. triceps brachii. With respect to capillary recruitment, CW-NIRS recordings in m. biceps brachii revealed dynamics that were both qualitatively and quantitatively similar in patients and controls. CONCLUSION We found no evidence for the contribution of motor unit- and capillary recruitment capacity of the upper arm muscles in adolescent and adult patients with SMA types 3 and 4 as primary limiting factors to premature fatigue during execution of a maximal arm-cycling task.
Collapse
Affiliation(s)
- Laura E Habets
- Center for Child Development, Exercise and Physical Literacy, Wilhelmina Children's Hospital, University Medical Center Utrecht, AB Utrecht, The Netherlands
| | - Bart Bartels
- Center for Child Development, Exercise and Physical Literacy, Wilhelmina Children's Hospital, University Medical Center Utrecht, AB Utrecht, The Netherlands
| | - Fay-Lynn Asselman
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht Brain Center, Utrecht University, GA Utrecht, The Netherlands
| | - Erik H J Hulzebos
- Center for Child Development, Exercise and Physical Literacy, Wilhelmina Children's Hospital, University Medical Center Utrecht, AB Utrecht, The Netherlands
| | - Dick F Stegeman
- Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Jeroen A L Jeneson
- Center for Child Development, Exercise and Physical Literacy, Wilhelmina Children's Hospital, University Medical Center Utrecht, AB Utrecht, The Netherlands
| | - W Ludo van der Pol
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht Brain Center, Utrecht University, GA Utrecht, The Netherlands
| |
Collapse
|
11
|
Chilcott EM, Muiruri EW, Hirst TC, Yáñez-Muñoz RJ. Systematic review and meta-analysis determining the benefits of in vivo genetic therapy in spinal muscular atrophy rodent models. Gene Ther 2022; 29:498-512. [PMID: 34611322 PMCID: PMC9482879 DOI: 10.1038/s41434-021-00292-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/30/2021] [Accepted: 09/12/2021] [Indexed: 01/31/2023]
Abstract
Spinal muscular atrophy (SMA) is a severe childhood neuromuscular disease for which two genetic therapies, Nusinersen (Spinraza, an antisense oligonucleotide), and AVXS-101 (Zolgensma, an adeno-associated viral vector of serotype 9 AAV9), have recently been approved. We investigated the pre-clinical development of SMA genetic therapies in rodent models and whether this can predict clinical efficacy. We have performed a systematic review of relevant publications and extracted median survival and details of experimental design. A random effects meta-analysis was used to estimate and compare efficacy. We stratified by experimental design (type of genetic therapy, mouse model, route and time of administration) and sought any evidence of publication bias. 51 publications were identified containing 155 individual comparisons, comprising 2573 animals in total. Genetic therapies prolonged survival in SMA mouse models by 3.23-fold (95% CI 2.75-3.79) compared to controls. Study design characteristics accounted for significant heterogeneity between studies and greatly affected observed median survival ratios. Some evidence of publication bias was found. These data are consistent with the extended average lifespan of Spinraza- and Zolgensma-treated children in the clinic. Together, these results support that SMA has been particularly amenable to genetic therapy approaches and highlight SMA as a trailblazer for therapeutic development.
Collapse
Affiliation(s)
- Ellie M. Chilcott
- grid.4970.a0000 0001 2188 881XAGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and Environment, Royal Holloway University of London, TW20 0EX London, UK ,Present Address: Institute for Women’s Health, UCL, 86-96 Chenies Mews, London, WC1E 6HX UK
| | - Evalyne W. Muiruri
- grid.4970.a0000 0001 2188 881XAGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and Environment, Royal Holloway University of London, TW20 0EX London, UK
| | - Theodore C. Hirst
- grid.416232.00000 0004 0399 1866Department of Neurosurgery, Royal Victoria Hospital, Belfast, BT12 6BA UK
| | - Rafael J. Yáñez-Muñoz
- grid.4970.a0000 0001 2188 881XAGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and Environment, Royal Holloway University of London, TW20 0EX London, UK
| |
Collapse
|
12
|
Zhang S, Liu H, Fang Q, He H, Lu X, Wang Y, Fan X. Shexiang Tongxin Dropping Pill Protects Against Chronic Heart Failure in Mice via Inhibiting the ERK/MAPK and TGF-β Signaling Pathways. Front Pharmacol 2021; 12:796354. [PMID: 34925046 PMCID: PMC8682969 DOI: 10.3389/fphar.2021.796354] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Chronic heart failure (CHF) is a major public health problem with high mortality and morbidity worldwide. Shexiang Tongxin Dropping Pill (STDP) is a widely used traditional Chinese medicine preparation for coronary heart disease and growing evidence proves that STDP exerts beneficial effects on CHF in the clinic. However, the molecular mechanism of the therapeutic effects of STDP on CHF remains largely unknown. Objective: This study aimed to elucidate the mechanism of action of STDP against CHF by integrating network pharmacology analysis and whole-transcriptome sequencing. Methods: First, the mouse model of CHF was established by the transverse aortic constriction (TAC) surgery, and the efficacy of STDP against CHF was evaluated by assessing the alterations in cardiac function, myocardial fibrosis, and cardiomyocyte hypertrophy with echocardiography, Masson’s trichrome staining, and wheat germ agglutinin staining. Next, a CHF disease network was constructed by integrating cardiovascular disease-related genes and the transcriptome sequencing data, which was used to explore the underlying mechanism of action of STDP. Then, the key targets involved in the effects of STDP on CHF were determined by network analysis algorithms, and pathway enrichment analysis was performed to these key genes. Finally, important targets in critical pathway were verified in vivo. Results: STDP administration obviously improved cardiac function, relieved cardiomyocyte hypertrophy, and ameliorated myocardial fibrosis in CHF mice. Moreover, STDP significantly reversed the imbalanced genes that belong to the disease network of CHF in mice with TAC, and the number of genes with the reverse effect was 395. Pathway analysis of the crucial genes with recovery efficiency revealed that pathways related to fibrosis and energy metabolism were highly enriched, while TGF-β pathway and ERK/MAPK pathway were predicted to be significantly affected. Consistently, validation experiments confirmed that inhibiting ERK/MAPK and TGF-β signaling pathways via reduction of the phosphorylation level of Smad3 and ERK1/2 is the important mechanism of STDP against CHF. Conclusion: Our data demonstrated that STDP can recover the imbalanced CHF network disturbed by the modeling of TAC through the multi-target and multi-pathway manner in mice, and the mechanisms are mainly related to inhibition of ERK/MAPK and TGF-β signaling pathways.
Collapse
Affiliation(s)
- Shuying Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hanbing Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qianqian Fang
- Inner Mongolia Conba Pharmaceutical Co., Ltd., Hohhot, China
| | - Houhong He
- Zhejiang Conba Pharmaceutical Co., Ltd., Hangzhou, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Yi Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaohui Fan
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| |
Collapse
|
13
|
Metabolic Dysfunction in Spinal Muscular Atrophy. Int J Mol Sci 2021; 22:ijms22115913. [PMID: 34072857 PMCID: PMC8198411 DOI: 10.3390/ijms22115913] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 12/11/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive genetic disorder leading to paralysis, muscle atrophy, and death. Significant advances in antisense oligonucleotide treatment and gene therapy have made it possible for SMA patients to benefit from improvements in many aspects of the once devastating natural history of the disease. How the depletion of survival motor neuron (SMN) protein, the product of the gene implicated in the disease, leads to the consequent pathogenic changes remains unresolved. Over the past few years, evidence toward a potential contribution of gastrointestinal, metabolic, and endocrine defects to disease phenotype has surfaced. These findings ranged from disrupted body composition, gastrointestinal tract, fatty acid, glucose, amino acid, and hormonal regulation. Together, these changes could have a meaningful clinical impact on disease traits. However, it is currently unclear whether these findings are secondary to widespread denervation or unique to the SMA phenotype. This review provides an in-depth account of metabolism-related research available to date, with a discussion of unique features compared to other motor neuron and related disorders.
Collapse
|
14
|
James R, Chaytow H, Ledahawsky LM, Gillingwater TH. Revisiting the role of mitochondria in spinal muscular atrophy. Cell Mol Life Sci 2021; 78:4785-4804. [PMID: 33821292 PMCID: PMC8195803 DOI: 10.1007/s00018-021-03819-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/22/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023]
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disease of variable clinical severity that is caused by mutations in the survival motor neuron 1 (SMN1) gene. Despite its name, SMN is a ubiquitous protein that functions within and outside the nervous system and has multiple cellular roles in transcription, translation, and proteostatic mechanisms. Encouragingly, several SMN-directed therapies have recently reached the clinic, albeit this has highlighted the increasing need to develop combinatorial therapies for SMA to achieve full clinical efficacy. As a subcellular site of dysfunction in SMA, mitochondria represents a relevant target for a combinatorial therapy. Accordingly, we will discuss our current understanding of mitochondrial dysfunction in SMA, highlighting mitochondrial-based pathways that offer further mechanistic insights into the involvement of mitochondria in SMA. This may ultimately facilitate translational development of targeted mitochondrial therapies for SMA. Due to clinical and mechanistic overlaps, such strategies may also benefit other motor neuron diseases and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Rachel James
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Helena Chaytow
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Leire M Ledahawsky
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK.
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, UK.
| |
Collapse
|
15
|
Deguise MO, Beauvais A, Schneider BL, Kothary R. Blood Flow to the Spleen is Altered in a Mouse Model of Spinal Muscular Atrophy. J Neuromuscul Dis 2021; 7:315-322. [PMID: 32333548 DOI: 10.3233/jnd-200493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder affecting young children. While pre-clinical models of SMA show small spleens, the same is not true in humans. Here, we show by doppler ultrasonography decreased splenic blood flow in Smn2B/- mice. Further, AAV9-SMN gene therapy does not rescue the distal ear and tail necrosis nor the spleen size in these mice, suggesting that the latter may be linked to a cardiovascular defect. Absence of smaller spleens in human patients is likely due to differences in presentation of defects in SMA between pre-clinical mouse models and human patients, particularly the susceptibility to cardiovascular issues.
Collapse
Affiliation(s)
- Marc-Olivier Deguise
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada
| | - Ariane Beauvais
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Bernard L Schneider
- Bertarelli Foundation Gene Therapy Platform, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland.,Brain Mind Institute, 27218 Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
16
|
In Search of a Cure: The Development of Therapeutics to Alter the Progression of Spinal Muscular Atrophy. Brain Sci 2021; 11:brainsci11020194. [PMID: 33562482 PMCID: PMC7915832 DOI: 10.3390/brainsci11020194] [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: 12/02/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
Until the recent development of disease-modifying therapeutics, spinal muscular atrophy (SMA) was considered a devastating neuromuscular disease with a poor prognosis for most affected individuals. Symptoms generally present during early childhood and manifest as muscle weakness and progressive paralysis, severely compromising the affected individual’s quality of life, independence, and lifespan. SMA is most commonly caused by the inheritance of homozygously deleted SMN1 alleles with retention of one or more copies of a paralog gene, SMN2, which inversely correlates with disease severity. The recent advent and use of genetically targeted therapies have transformed SMA into a prototype for monogenic disease treatment in the era of genetic medicine. Many SMA-affected individuals receiving these therapies achieve traditionally unobtainable motor milestones and survival rates as medicines drastically alter the natural progression of this disease. This review discusses historical SMA progression and underlying disease mechanisms, highlights advances made in therapeutic research, clinical trials, and FDA-approved medicines, and discusses possible second-generation and complementary medicines as well as optimal temporal intervention windows in order to optimize motor function and improve quality of life for all SMA-affected individuals.
Collapse
|
17
|
Spinal muscular atrophy: Broad disease spectrum and sex-specific phenotypes. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166063. [PMID: 33412266 DOI: 10.1016/j.bbadis.2020.166063] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022]
Abstract
Spinal muscular atrophy (SMA) is one of the major genetic disorders associated with infant mortality. More than 90% of cases of SMA result from deletions of or mutations in the Survival Motor Neuron 1 (SMN1) gene. SMN2, a nearly identical copy of SMN1, does not compensate for the loss of SMN1 due to predominant skipping of exon 7. The spectrum of SMA is broad, ranging from prenatal death to infant mortality to survival into adulthood. All tissues, including brain, spinal cord, bone, skeletal muscle, heart, lung, liver, pancreas, gastrointestinal tract, kidney, spleen, ovary and testis, are directly and/or indirectly affected in SMA. Accumulating evidence on impaired mitochondrial biogenesis and defects in X chromosome-linked modifying factors, coupled with the sexual dimorphic nature of many tissues, point to sex-specific vulnerabilities in SMA. Here we review the role of sex in the pathogenesis of SMA.
Collapse
|
18
|
AAV9-DOK7 gene therapy reduces disease severity in Smn 2B/- SMA model mice. Biochem Biophys Res Commun 2020; 530:107-114. [PMID: 32828271 DOI: 10.1016/j.bbrc.2020.07.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/07/2020] [Indexed: 11/23/2022]
Abstract
Spinal Muscular Atrophy (SMA) is an autosomal recessive neuromuscular disease caused by deletions or mutations in the survival motor neuron (SMN1) gene. An important hallmark of disease progression is the pathology of neuromuscular junctions (NMJs). Affected NMJs in the SMA context exhibit delayed maturation, impaired synaptic transmission, and loss of contact between motor neurons and skeletal muscle. Protection and maintenance of NMJs remains a focal point of therapeutic strategies to treat SMA, and the recent implication of the NMJ-organizer Agrin in SMA pathology suggests additional NMJ organizing molecules may contribute. DOK7 is an NMJ organizer that functions downstream of Agrin. The potential of DOK7 as a putative therapeutic target was demonstrated by adeno-associated virus (AAV)-mediated gene therapy delivery of DOK7 in Amyotrophic Lateral Sclerosis (ALS) and Emery Dreyefuss Muscular Dystrophy (EDMD). To assess the potential of DOK7 as a disease modifier of SMA, we administered AAV-DOK7 to an intermediate mouse model of SMA. AAV9-DOK7 treatment conferred improvements in NMJ architecture and reduced muscle fiber atrophy. Additionally, these improvements resulted in a subtle reduction in phenotypic severity, evidenced by improved grip strength and an extension in survival. These findings reveal DOK7 is a novel modifier of SMA.
Collapse
|
19
|
SMN-deficiency disrupts SERCA2 expression and intracellular Ca 2+ signaling in cardiomyocytes from SMA mice and patient-derived iPSCs. Skelet Muscle 2020; 10:16. [PMID: 32384912 PMCID: PMC7206821 DOI: 10.1186/s13395-020-00232-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 11/17/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease characterized by loss of alpha motor neurons and skeletal muscle atrophy. The disease is caused by mutations of the SMN1 gene that result in reduced functional expression of survival motor neuron (SMN) protein. SMN is ubiquitously expressed, and there have been reports of cardiovascular dysfunction in the most severe SMA patients and animal models of the disease. In this study, we directly assessed the function of cardiomyocytes isolated from a severe SMA model mouse and cardiomyocytes generated from patient-derived IPSCs. Consistent with impaired cardiovascular function at the very early disease stages in mice, heart failure markers such as brain natriuretic peptide were significantly elevated. Functionally, cardiomyocyte relaxation kinetics were markedly slowed and the T50 for Ca2+ sequestration increased to 146 ± 4 ms in SMN-deficient cardiomyocytes from 126 ± 4 ms in wild type cells. Reducing SMN levels in cardiomyocytes from control patient IPSCs slowed calcium reuptake similar to SMA patent-derived cardiac cells. Importantly, restoring SMN increased calcium reuptake rate. Taken together, these results indicate that SMN deficiency impairs cardiomyocyte function at least partially through intracellular Ca2+ cycling dysregulation.
Collapse
|
20
|
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
|
21
|
Shababi M, Smith CE, Kacher M, Alrawi Z, Villalon E, Davis D, Bryda EC, Lorson CL. Development of a novel severe mouse model of spinal muscular atrophy with respiratory distress type 1: FVB-nmd. Biochem Biophys Res Commun 2019; 520:341-346. [PMID: 31604525 PMCID: PMC6936219 DOI: 10.1016/j.bbrc.2019.10.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 11/29/2022]
Abstract
Spinal Muscular Atrophy with Respiratory Distress type 1 (SMARD1) is an autosomal recessive disease that develops early during infancy. The gene responsible for disease development is immunoglobulin helicase μ-binding protein 2 (IGHMBP2). IGHMBP2 is a ubiquitously expressed gene but its mutation results in the loss of alpha-motor neurons and subsequent muscle atrophy initially of distal muscles. The current SMARD1 mouse model arose from a spontaneous mutation originally referred to as neuromuscular degeneration (nmd). The nmd mice have the C57BL/6 genetic background and contain an A to G mutation in intron 4 of the endogenous Ighmbp2 gene. This mutation causes aberrant splicing, resulting in only 20-25% of full-length functional protein. Several congenital conditions including hydrocephalus are common in the C57BL/6 background, consistent with our previous observations when developing a gene therapy approach for SMARD1. Additionally, a modifier allele exists on chromosome 13 in nmd mice that can partially suppress the phenotype, resulting in some animals that have extended life spans (up to 200 days). To eliminate the intrinsic complications of the C57BL/6 background and the variation in survival due to the genetic modifier effect, we created a new SMARD1 mouse model that contains the same intron 4 mutation in Ighmbp2 as nmd mice but is now on a FVB congenic background. FVB-nmd are consistently more severe than the original nmd mice with respect to survival, weigh and motor function. The relatively short life span (18-21 days) of FVB-nmd mice allows us to monitor therapeutic efficacy for a variety of novel therapeutics in a timely manner without the complication of the small percentage of longer-lived animals that were observed in our colony of nmd mice.
Collapse
Affiliation(s)
- Monir Shababi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Caley E. Smith
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA,Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Mona Kacher
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Zayd Alrawi
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Eric Villalon
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Daniel Davis
- Animal Modeling Core, University of Missouri, Columbia, Missouri, USA
| | - Elizabeth C. Bryda
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA,Animal Modeling Core, University of Missouri, Columbia, Missouri, USA
| | - Christian L. Lorson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA,Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri, USA
| |
Collapse
|
22
|
Šoltić D, Shorrock HK, Allardyce H, Wilson EL, Holt I, Synowsky SA, Shirran SL, Parson SH, Gillingwater TH, Fuller HR. Lamin A/C dysregulation contributes to cardiac pathology in a mouse model of severe spinal muscular atrophy. Hum Mol Genet 2019; 28:3515-3527. [PMID: 31397869 PMCID: PMC6927462 DOI: 10.1093/hmg/ddz195] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiac pathology is emerging as a prominent systemic feature of spinal muscular atrophy (SMA), but little is known about the underlying molecular pathways. Using quantitative proteomics analysis, we demonstrate widespread molecular defects in heart tissue from the Taiwanese mouse model of severe SMA. We identify increased levels of lamin A/C as a robust molecular phenotype in the heart of SMA mice and show that lamin A/C dysregulation is also apparent in SMA patient fibroblast cells and other tissues from SMA mice. Lamin A/C expression was regulated in vitro by knockdown of the E1 ubiquitination factor ubiquitin-like modifier activating enzyme 1, a key downstream mediator of SMN-dependent disease pathways, converging on β-catenin signaling. Increased levels of lamin A are known to increase the rigidity of nuclei, inevitably disrupting contractile activity in cardiomyocytes. The increased lamin A/C levels in the hearts of SMA mice therefore provide a likely mechanism explaining morphological and functional cardiac defects, leading to blood pooling. Therapeutic strategies directed at lamin A/C may therefore offer a new approach to target cardiac pathology in SMA.
Collapse
Affiliation(s)
- Darija Šoltić
- Institute for Science and Technology in Medicine, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Hannah K Shorrock
- Edinburgh Medical School: Biomedical Sciences
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Hazel Allardyce
- Institute of Education for Medical and Dental Science, College of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB24 3FX, UK
| | - Emma L Wilson
- Chester Medical School, University of Chester, Chester CH1 4BJ, UK
| | - Ian Holt
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Silvia A Synowsky
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, St Andrews KY16 9ST, UK
| | - Sally L Shirran
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, St Andrews KY16 9ST, UK
| | - Simon H Parson
- Institute of Education for Medical and Dental Science, College of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB24 3FX, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School: Biomedical Sciences
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Heidi R Fuller
- Institute for Science and Technology in Medicine, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| |
Collapse
|
23
|
Shababi M, Villalón E, Kaifer KA, DeMarco V, Lorson CL. A Direct Comparison of IV and ICV Delivery Methods for Gene Replacement Therapy in a Mouse Model of SMARD1. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 10:348-360. [PMID: 30202772 PMCID: PMC6127875 DOI: 10.1016/j.omtm.2018.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 08/13/2018] [Indexed: 01/22/2023]
Abstract
Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is an infantile autosomal recessive disease caused by the loss of the ubiquitously expressed IGHMBP2 gene. SMARD1 causes degeneration of alpha-motor neurons, resulting in distal muscle weakness, diaphragm paralysis, and respiratory malfunction. We have reported that delivery of a low dose of AAV9-IGHMBP2 to the CNS results in a significant rescue of the SMARD1 mouse model (nmd). To examine how a delivery route can impact efficacy, a direct comparison of intravenous (IV) and intracerebroventricular (ICV) delivery of AAV9-IGHMBP2 was performed. Using a low-dose, both IV and ICV delivery routes led to a significant extension in survival and increased body weight. Conversely, only ICV-treated animals demonstrated improvements in the hindlimb muscle, neuromuscular junction, and motor function. The hindlimb phenotype of IV-treated mice resembled the untreated nmd mice. We investigated whether the increased survival of IV-treated nmd mice was the result of a positive impact on the cardiac function. Our results revealed that cardiac function and pathology were similarly improved in IV- and ICV-treated mice. We concluded that while IV delivery of a low dose does not improve the hindlimb phenotype and motor function, partial restoration of cardiac performance is sufficient to significantly extend survival.
Collapse
Affiliation(s)
- Monir Shababi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.,Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Eric Villalón
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.,Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Kevin A Kaifer
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.,Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Vince DeMarco
- Department of Medicine, Division of Endocrinology, Diabetes and Cardiovascular Center, University of Missouri, Columbia, MO, USA.,Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA.,Research Service, Harry S. Truman Memorial Veterans Hospital, University of Missouri, Columbia, MO, USA
| | - Christian L Lorson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.,Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
24
|
Maxwell GK, Szunyogova E, Shorrock HK, Gillingwater TH, Parson SH. Developmental and degenerative cardiac defects in the Taiwanese mouse model of severe spinal muscular atrophy. J Anat 2018; 232:965-978. [PMID: 29473159 PMCID: PMC5978979 DOI: 10.1111/joa.12793] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2018] [Indexed: 12/31/2022] Open
Abstract
Spinal muscular atrophy (SMA), an autosomal recessive disease caused by a decrease in levels of the survival motor neuron (SMN) protein, is the most common genetic cause of infant mortality. Although neuromuscular pathology is the most severe feature of SMA, other organs and tissues, including the heart, are also known to be affected in both patients and animal models. Here, we provide new insights into changes occurring in the heart, predominantly at pre- and early symptomatic ages, in the Taiwanese mouse model of severe SMA. Thinning of the interventricular septum and dilation of the ventricles occurred at pre- and early symptomatic ages. However, the left ventricular wall was significantly thinner in SMA mice from birth, occurring prior to any overt neuromuscular symptoms. Alterations in collagen IV protein from birth indicated changes to the basement membrane and contributed to the abnormal arrangement of cardiomyocytes in SMA hearts. This raises the possibility that developmental defects, occurring prenatally, may contribute to cardiac pathology in SMA. In addition, cardiomyocytes in SMA hearts exhibited oxidative stress at pre-symptomatic ages and increased apoptosis during early symptomatic stages of disease. Heart microvasculature was similarly decreased at an early symptomatic age, likely contributing to the oxidative stress and apoptosis phenotypes observed. Finally, an increased incidence of blood retention in SMA hearts post-fixation suggests the likelihood of functional defects, resulting in blood pooling. These pathologies mirror dilated cardiomyopathy, with clear consequences for heart function that would likely contribute to potential heart failure. Our findings add significant additional experimental evidence in support of the requirement to develop systemic therapies for SMA capable of treating non-neuromuscular pathologies.
Collapse
Affiliation(s)
| | - Eva Szunyogova
- Institute for Medical ScienceUniversity of AberdeenAberdeenUK
- Euan MacDonald Centre for Motor Neurone Disease ResearchUniversity of EdinburghEdinburghUK
| | - Hannah K. Shorrock
- Euan MacDonald Centre for Motor Neurone Disease ResearchUniversity of EdinburghEdinburghUK
- Edinburgh Medical School: Biomedical SciencesUniversity of EdinburghEdinburghUK
| | - Thomas H. Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease ResearchUniversity of EdinburghEdinburghUK
- Edinburgh Medical School: Biomedical SciencesUniversity of EdinburghEdinburghUK
| | - Simon H. Parson
- Institute for Medical ScienceUniversity of AberdeenAberdeenUK
- Euan MacDonald Centre for Motor Neurone Disease ResearchUniversity of EdinburghEdinburghUK
| |
Collapse
|
25
|
Falsaperla R, Vitaliti G, Collotta AD, Fiorillo C, Pulvirenti A, Alaimo S, Romano C, Ruggieri M. Electrocardiographic Evaluation in Patients With Spinal Muscular Atrophy: A Case-Control Study. J Child Neurol 2018; 33:487-492. [PMID: 29687752 DOI: 10.1177/0883073818767170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND This study aimed to show the impairment of autonomic cardiac conduction causing bradycardia and/or electrocardiographic alterations in children affected by spinal muscular atrophy type 1 and 2 (SMA 1 and 2). METHODS We included 25 spinal muscular atrophy patients, admitted from November 2016 to May 2017. All patients underwent an electrocardiographic examination and we studied PR and QRS intervals, P-waves and QRS amplitudes, and heart rate in spinal muscular atrophy patients compared to a control group. RESULTS In all patients, we found longer PRi and QRSi ( P < .05), lower P-wave and QRS complex amplitudes ( P < .01), and a decreased heart rate ( P < .01) with respect to controls. When we divided our patients into SMA1 and SMA2 subgroups, we found that statistical differences were maintained for P-wave and QRS complex amplitudes and heart rate, but not for PRi and QRSi with respect to controls. CONCLUSION We suggest the hypothesis of SMN expression on cardiac tissue condition and/or autonomic cardiac conduction.
Collapse
Affiliation(s)
- Raffaele Falsaperla
- 1 General Pediatrics and Pediatric Acute and Emergency Complex Unit, Policlinico-Vittorio-Emanuele University Hospital, University of Catania, Italy
| | - Giovanna Vitaliti
- 1 General Pediatrics and Pediatric Acute and Emergency Complex Unit, Policlinico-Vittorio-Emanuele University Hospital, University of Catania, Italy
| | - Ausilia Desiree Collotta
- 1 General Pediatrics and Pediatric Acute and Emergency Complex Unit, Policlinico-Vittorio-Emanuele University Hospital, University of Catania, Italy
| | - Chiara Fiorillo
- 2 Unit of Paediatric Neurology and Muscle Diseases, G. Gaslini Institute, Genoa, Italy
| | - Alfredo Pulvirenti
- 3 Department of Clinical and Experimental Medicine, Section of Bioinformatics, University of Catania, Italy
| | - Salvatore Alaimo
- 3 Department of Clinical and Experimental Medicine, Section of Bioinformatics, University of Catania, Italy
| | - Catia Romano
- 1 General Pediatrics and Pediatric Acute and Emergency Complex Unit, Policlinico-Vittorio-Emanuele University Hospital, University of Catania, Italy
| | - Martino Ruggieri
- 4 Unit of Rare Diseases of the Nervous System in Childhood, Policlinico-Vittorio-Emanuele University Hospital, University of Catania, Italy
| |
Collapse
|
26
|
Howell MD, Ottesen EW, Singh NN, Anderson RL, Seo J, Sivanesan S, Whitley EM, Singh RN. TIA1 is a gender-specific disease modifier of a mild mouse model of spinal muscular atrophy. Sci Rep 2017; 7:7183. [PMID: 28775379 PMCID: PMC5543135 DOI: 10.1038/s41598-017-07468-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/26/2017] [Indexed: 02/08/2023] Open
Abstract
Spinal muscular atrophy (SMA) is caused by deletions or mutations of Survival Motor Neuron 1 (SMN1) gene. The nearly identical SMN2 cannot compensate for SMN1 loss due to exon 7 skipping. The allele C (C +/+) mouse recapitulates a mild SMA-like phenotype and offers an ideal system to monitor the role of disease-modifying factors over a long time. T-cell-restricted intracellular antigen 1 (TIA1) regulates SMN exon 7 splicing. TIA1 is reported to be downregulated in obese patients, although it is not known if the effect is gender-specific. We show that female Tia1-knockout (Tia1 -/-) mice gain significant body weight (BW) during early postnatal development. We next examined the effect of Tia1 deletion in novel C +/+/Tia1 -/- mice. Underscoring the opposing effects of Tia1 deletion and low SMN level on BW gain, both C +/+ and C +/+/Tia1 -/- females showed similar BW gain trajectory at all time points during our study. We observed early tail necrosis in C +/+/Tia1 -/- females but not in males. We show enhanced impairment of male reproductive organ development and exacerbation of the C +/+/Tia1 -/- testis transcriptome. Our findings implicate a protein factor as a gender-specific modifier of a mild mouse model of SMA.
Collapse
Affiliation(s)
- Matthew D Howell
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Eric W Ottesen
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Natalia N Singh
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Rachel L Anderson
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
| | - Joonbae Seo
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | | | - Elizabeth M Whitley
- Department of Veterinary Pathology, Iowa State University, Ames, IA, 50011-1250, USA
- Pathogenesis, LLC, Gainesville, Florida, 32614, USA
| | - Ravindra N Singh
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, 50011, USA.
| |
Collapse
|
27
|
Deguise MO, De Repentigny Y, McFall E, Auclair N, Sad S, Kothary R. Immune dysregulation may contribute to disease pathogenesis in spinal muscular atrophy mice. Hum Mol Genet 2017; 26:801-819. [PMID: 28108555 PMCID: PMC5409095 DOI: 10.1093/hmg/ddw434] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/16/2016] [Indexed: 01/21/2023] Open
Abstract
Spinal muscular atrophy (SMA) has long been solely considered a neurodegenerative disorder. However, recent work has highlighted defects in many other cell types that could contribute to disease aetiology. Interestingly, the immune system has never been extensively studied in SMA. Defects in lymphoid organs could exacerbate disease progression by neuroinflammation or immunodeficiency. Smn depletion led to severe alterations in the thymus and spleen of two different mouse models of SMA. The spleen from Smn depleted mice was dramatically smaller at a very young age and its histological architecture was marked by mislocalization of immune cells in the Smn2B/- model mice. In comparison, the thymus was relatively spared in gross morphology but showed many histological alterations including cortex thinning in both mouse models at symptomatic ages. Thymocyte development was also impaired as evidenced by abnormal population frequencies in the Smn2B/- thymus. Cytokine profiling revealed major changes in different tissues of both mouse models. Consistent with our observations, we found that survival motor neuron (Smn) protein levels were relatively high in lymphoid organs compared to skeletal muscle and spinal cord during postnatal development in wild type mice. Genetic introduction of one copy of the human SMN2 transgene was enough to rescue splenic and thymic defects in Smn2B/- mice. Thus, Smn is required for the normal development of lymphoid organs, and altered immune function may contribute to SMA disease pathogenesis.
Collapse
Affiliation(s)
- Marc-Olivier Deguise
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Department of Cellular and Molecular Medicine.,Centre for Neuromuscular Disease, University of Ottawa
| | - Yves De Repentigny
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Centre for Neuromuscular Disease, University of Ottawa
| | - Emily McFall
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Centre for Neuromuscular Disease, University of Ottawa
| | - Nicole Auclair
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Faculty of Science, University of Ottawa, Ottawa, Ontario, Canada, K1N 9B4
| | - Subash Sad
- Department of Biochemistry, Microbiology, and Immunology
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Department of Cellular and Molecular Medicine.,Centre for Neuromuscular Disease, University of Ottawa.,Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| |
Collapse
|
28
|
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
|
29
|
Wijngaarde CA, Blank AC, Stam M, Wadman RI, van den Berg LH, van der Pol WL. Cardiac pathology in spinal muscular atrophy: a systematic review. Orphanet J Rare Dis 2017; 12:67. [PMID: 28399889 PMCID: PMC5387385 DOI: 10.1186/s13023-017-0613-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/14/2017] [Indexed: 01/09/2023] Open
Abstract
Background Hereditary proximal spinal muscular atrophy (SMA) is a severe neuromuscular disease of childhood caused by homozygous loss of function of the survival motor neuron (SMN) 1 gene. The presence of a second, nearly identical SMN gene (SMN2) in the human genome ensures production of residual levels of the ubiquitously expressed SMN protein. Alpha-motor neurons in the ventral horns of the spinal cord are most vulnerable to reduced SMN concentrations but the development or function of other tissues may also be affected, and cardiovascular abnormalities have frequently been reported both in patients and SMA mouse models. Methods We systematically reviewed reported cardiac pathology in relation to SMN deficiency. To investigate the relevance of the possible association in more detail, we used clinical classification systems to characterize structural cardiac defects and arrhythmias. Conclusions Seventy-two studies with a total of 264 SMA patients with reported cardiac pathology were identified, along with 14 publications on SMA mouse models with abnormalities of the heart. Structural cardiac pathology, mainly septal defects and abnormalities of the cardiac outflow tract, was reported predominantly in the most severely affected patients (i.e. SMA type 1). Cardiac rhythm disorders were most frequently reported in patients with milder SMA types (e.g. SMA type 3). All included studies lacked control groups and a standardized approach for cardiac evaluation. The convergence to specific abnormalities of cardiac structure and function may indicate vulnerability of specific cell types or developmental processes relevant for cardiogenesis. Future studies would benefit from a controlled and standardized approach for cardiac evaluation in patients with SMA. Electronic supplementary material The online version of this article (doi:10.1186/s13023-017-0613-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- C A Wijngaarde
- Department of Neurology and Neurosurgery, F02.230, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands.
| | - A C Blank
- Department of Pediatric Cardiology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M Stam
- Department of Neurology and Neurosurgery, F02.230, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - R I Wadman
- Department of Neurology and Neurosurgery, F02.230, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - L H van den Berg
- Department of Neurology and Neurosurgery, F02.230, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands
| | - W L van der Pol
- Department of Neurology and Neurosurgery, F02.230, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Heidelberglaan 100, 3508 GA, Utrecht, The Netherlands.
| |
Collapse
|
30
|
Szunyogova E, Zhou H, Maxwell GK, Powis RA, Francesco M, Gillingwater TH, Parson SH. Survival Motor Neuron (SMN) protein is required for normal mouse liver development. Sci Rep 2016; 6:34635. [PMID: 27698380 PMCID: PMC5048144 DOI: 10.1038/srep34635] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/12/2016] [Indexed: 01/15/2023] Open
Abstract
Spinal Muscular Atrophy (SMA) is caused by mutation or deletion of the survival motor neuron 1 (SMN1) gene. Decreased levels of, cell-ubiquitous, SMN protein is associated with a range of systemic pathologies reported in severe patients. Despite high levels of SMN protein in normal liver, there is no comprehensive study of liver pathology in SMA. We describe failed liver development in response to reduced SMN levels, in a mouse model of severe SMA. The SMA liver is dark red, small and has: iron deposition; immature sinusoids congested with blood; persistent erythropoietic elements and increased immature red blood cells; increased and persistent megakaryocytes which release high levels of platelets found as clot-like accumulations in the heart. Myelopoiesis in contrast, was unaffected. Further analysis revealed significant molecular changes in SMA liver, consistent with the morphological findings. Antisense treatment from birth with PMO25, increased lifespan and ameliorated all morphological defects in liver by postnatal day 21. Defects in the liver are evident at birth, prior to motor system pathology, and impair essential liver function in SMA. Liver is a key recipient of SMA therapies, and systemically delivered antisense treatment, completely rescued liver pathology. Liver therefore, represents an important therapeutic target in SMA.
Collapse
Affiliation(s)
- Eva Szunyogova
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Euan MacDonald Center for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Haiyan Zhou
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Gillian K. Maxwell
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| | - Rachael A. Powis
- Euan MacDonald Center for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- Center for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Muntoni Francesco
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Thomas H. Gillingwater
- Euan MacDonald Center for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- Center for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Simon H. Parson
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Euan MacDonald Center for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
31
|
Deguise MO, Boyer JG, McFall ER, Yazdani A, De Repentigny Y, Kothary R. Differential induction of muscle atrophy pathways in two mouse models of spinal muscular atrophy. Sci Rep 2016; 6:28846. [PMID: 27349908 PMCID: PMC4924104 DOI: 10.1038/srep28846] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/08/2016] [Indexed: 12/15/2022] Open
Abstract
Motor neuron loss and neurogenic atrophy are hallmarks of spinal muscular atrophy (SMA), a leading genetic cause of infant deaths. Previous studies have focused on deciphering disease pathogenesis in motor neurons. However, a systematic evaluation of atrophy pathways in muscles is lacking. Here, we show that these pathways are differentially activated depending on severity of disease in two different SMA model mice. Although proteasomal degradation is induced in skeletal muscle of both models, autophagosomal degradation is present only in Smn(2B/-) mice but not in the more severe Smn(-/-); SMN2 mice. Expression of FoxO transcription factors, which regulate both proteasomal and autophagosomal degradation, is elevated in Smn(2B/-) muscle. Remarkably, administration of trichostatin A reversed all molecular changes associated with atrophy. Cardiac muscle also exhibits differential induction of atrophy between Smn(2B/-) and Smn(-/-); SMN2 mice, albeit in the opposite direction to that of skeletal muscle. Altogether, our work highlights the importance of cautious analysis of different mouse models of SMA as distinct patterns of atrophy induction are at play depending on disease severity. We also revealed that one of the beneficial impacts of trichostatin A on SMA model mice is via attenuation of muscle atrophy through reduction of FoxO expression to normal levels.
Collapse
Affiliation(s)
- Marc-Olivier Deguise
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 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
| | - Justin G Boyer
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, K1H 8L6 Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5 Canada
| | - Emily R McFall
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, K1H 8L6 Canada
| | - Armin Yazdani
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, K1H 8L6 Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5 Canada
| | - Yves De Repentigny
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, K1H 8L6 Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 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 Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5 Canada
| |
Collapse
|
32
|
Burns JK, Kothary R, Parks RJ. Opening the window: The case for carrier and perinatal screening for spinal muscular atrophy. Neuromuscul Disord 2016; 26:551-9. [PMID: 27460292 DOI: 10.1016/j.nmd.2016.06.459] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 11/26/2022]
Abstract
Spinal muscular atrophy (SMA) is the most common genetically inherited neurodegenerative disease that leads to infant mortality worldwide. SMA is caused by genetic deletion or mutation in the survival of motor neuron 1 (SMN1) gene, which results in a deficiency in SMN protein. For reasons that are still unclear, SMN protein deficiency predominantly affects α-motor neurons, resulting in their degeneration and subsequent paralysis of limb and trunk muscles, progressing to death in severe cases. Emerging evidence suggests that SMN protein deficiency also affects the heart, autonomic nervous system, skeletal muscle, liver, pancreas and perhaps many other organs. Currently, there is no cure for SMA. Patient treatment includes respiratory care, physiotherapy, and nutritional management, which can somewhat ameliorate disease symptoms and increase life span. Fortunately, several novel therapies have advanced to human clinical trials. However, data from studies in animal models of SMA indicate that the greatest therapeutic benefit is achieved through initiating treatment as early as possible, before widespread loss of motor neurons has occurred. In this review, we discuss the merit of carrier and perinatal patient screening for SMA considering the efficacy of emerging therapeutics and the physical, emotional and financial burden of the disease on affected families and society.
Collapse
Affiliation(s)
- Joseph K Burns
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada; University of Ottawa Centre for Neuromuscular Disease, Ottawa, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada; University of Ottawa Centre for Neuromuscular Disease, Ottawa, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada; Department of Medicine, University of Ottawa, Ottawa, Canada
| | - Robin J Parks
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada; University of Ottawa Centre for Neuromuscular Disease, Ottawa, Canada; Department of Medicine, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
33
|
Butchbach MER, Lumpkin CJ, Harris AW, Saieva L, Edwards JD, Workman E, Simard LR, Pellizzoni L, Burghes AHM. Protective effects of butyrate-based compounds on a mouse model for spinal muscular atrophy. Exp Neurol 2016; 279:13-26. [PMID: 26892876 PMCID: PMC4834225 DOI: 10.1016/j.expneurol.2016.02.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/11/2016] [Accepted: 02/13/2016] [Indexed: 11/17/2022]
Abstract
Proximal spinal muscular atrophy (SMA) is a childhood-onset degenerative disease resulting from the selective loss of motor neurons in the spinal cord. SMA is caused by the loss of SMN1 (survival motor neuron 1) but retention of SMN2. The number of copies of SMN2 modifies disease severity in SMA patients as well as in mouse models, making SMN2 a target for therapeutics development. Sodium butyrate (BA) and its analog (4PBA) have been shown to increase SMN2 expression in SMA cultured cells. In this study, we examined the effects of BA, 4PBA as well as two BA prodrugs-glyceryl tributyrate (BA3G) and VX563-on the phenotype of SMNΔ7 SMA mice. Treatment with 4PBA, BA3G and VX563 but not BA beginning at PND04 significantly improved the lifespan and delayed disease end stage, with administration of VX563 also improving the growth rate of these mice. 4PBA and VX563 improved the motor phenotype of SMNΔ7 SMA mice and prevented spinal motor neuron loss. Interestingly, neither 4PBA nor VX563 had an effect on SMN expression in the spinal cords of treated SMNΔ7 SMA mice; however, they inhibited histone deacetylase (HDAC) activity and restored the normal phosphorylation states of Akt and glycogen synthase kinase 3β, both of which are altered by SMN deficiency in vivo. These observations show that BA-based compounds with favorable pharmacokinetics ameliorate SMA pathology possibly by modulating HDAC and Akt signaling.
Collapse
Affiliation(s)
- Matthew E R Butchbach
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Center for Pediatric Research, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, USA; Department of Biological Sciences, University of Delaware, Newark, DE, USA.
| | - Casey J Lumpkin
- Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Ashlee W Harris
- Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, USA
| | - Luciano Saieva
- Center for Motor Neuron Biology and Disease, Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Jonathan D Edwards
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eileen Workman
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Louise R Simard
- Department of Biochemistry and Medical Genetics, University of Manitoba Faculty of Health Sciences, Winnipeg, Manitoba, Canada
| | - Livio Pellizzoni
- Center for Motor Neuron Biology and Disease, Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Arthur H M Burghes
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
34
|
Simone C, Ramirez A, Bucchia M, Rinchetti P, Rideout H, Papadimitriou D, Re DB, Corti S. Is spinal muscular atrophy a disease of the motor neurons only: pathogenesis and therapeutic implications? Cell Mol Life Sci 2016; 73:1003-20. [PMID: 26681261 PMCID: PMC4756905 DOI: 10.1007/s00018-015-2106-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 01/16/2023]
Abstract
Spinal muscular atrophy (SMA) is a genetic neurological disease that causes infant mortality; no effective therapies are currently available. SMA is due to homozygous mutations and/or deletions in the survival motor neuron 1 gene and subsequent reduction of the SMN protein, leading to the death of motor neurons. However, there is increasing evidence that in addition to motor neurons, other cell types are contributing to SMA pathology. In this review, we will discuss the involvement of non-motor neuronal cells, located both inside and outside the central nervous system, in disease onset and progression. Even if SMN restoration in motor neurons is needed, it has been shown that optimal phenotypic amelioration in animal models of SMA requires a more widespread SMN correction. It has been demonstrated that non-motor neuronal cells are also involved in disease pathogenesis and could have important therapeutic implications. For these reasons it will be crucial to take this evidence into account for the clinical translation of the novel therapeutic approaches.
Collapse
Affiliation(s)
- Chiara Simone
- Neuroscience Section, Neurology Unit, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Agnese Ramirez
- Neuroscience Section, Neurology Unit, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Monica Bucchia
- Neuroscience Section, Neurology Unit, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Paola Rinchetti
- Neuroscience Section, Neurology Unit, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Hardy Rideout
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens (BRFAA), Soranou Efesiou 4, 115 27, Athens, Greece
| | - Dimitra Papadimitriou
- Division of Basic Neurosciences, Biomedical Research Foundation of the Academy of Athens (BRFAA), Soranou Efesiou 4, 115 27, Athens, Greece
| | - Diane B Re
- Department of Environmental Health Sciences, Columbia University, New York, NY, 10032, USA
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
| | - Stefania Corti
- Neuroscience Section, Neurology Unit, Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, University of Milan, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy.
| |
Collapse
|
35
|
Rescue of a Mouse Model of Spinal Muscular Atrophy With Respiratory Distress Type 1 by AAV9-IGHMBP2 Is Dose Dependent. Mol Ther 2016; 24:855-66. [PMID: 26860981 DOI: 10.1038/mt.2016.33] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 01/17/2016] [Indexed: 01/07/2023] Open
Abstract
Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is an autosomal recessive disease occurring during childhood. The gene responsible for disease development is a ubiquitously expressed protein, IGHMBP2. Mutations in IGHMBP2 result in the loss of α-motor neurons leading to muscle atrophy in the distal limbs accompanied by respiratory complications. Although genetically and clinically distinct, proximal SMA is also caused by the loss of a ubiquitously expressed gene (SMN). Significant preclinical success has been achieved in proximal SMA using viral-based gene replacement strategies. We leveraged the technologies employed in SMA to demonstrate gene replacement efficacy in an SMARD1 animal model. Intracerebroventricular (ICV) injection of single-stranded AAV9 expressing the full-length cDNA of IGHMBP2 in a low dose led to a significant level of rescue in treated SMARD1 animals. Consistent with drastically increased survival, weight gain, and strength, the rescued animals demonstrated a significant improvement in muscle, NMJ, motor neurons, and axonal pathology. In addition, increased levels of IGHMBP2 in lumbar motor neurons verified the efficacy of the virus to transduce the target tissues. Our results indicate that AAV9-based gene replacement is a viable strategy for SMARD1, although dosing effects and potential negative impacts of high dose and ICV injection should be thoroughly investigated.
Collapse
|
36
|
Arnold W, McGovern VL, Sanchez B, Li J, Corlett KM, Kolb SJ, Rutkove SB, Burghes AH. The neuromuscular impact of symptomatic SMN restoration in a mouse model of spinal muscular atrophy. Neurobiol Dis 2015; 87:116-23. [PMID: 26733414 DOI: 10.1016/j.nbd.2015.12.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/20/2015] [Accepted: 12/25/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Significant advances in the development of SMN-restoring therapeutics have occurred since 2010 when very effective biological treatments were reported in mouse models of spinal muscular atrophy. As these treatments are applied in human clinical trials, there is pressing need to define quantitative assessments of disease progression, treatment stratification, and therapeutic efficacy. The electrophysiological measures Compound Muscle Action Potential and Motor Unit Number Estimation are reliable measures of nerve function. In both the SMN∆7 mouse and a pig model of spinal muscular atrophy, early SMN restoration results in preservation of electrophysiological measures. Currently, clinical trials are underway in patients at post-symptomatic stages of disease progression. In this study, we present results from both early and delayed SMN restoration using clinically-relevant measures including electrical impedance myography, compound muscle action potential, and motor unit number estimation to quantify the efficacy and time-sensitivity of SMN-restoring therapy. METHODS SMA∆7 mice were treated via intracerebroventricular injection with antisense oligonucleotides targeting ISS-N1 to increase SMN protein from the SMN2 gene on postnatal day 2, 4, or 6 and compared with sham-treated spinal muscular atrophy and control mice. Compound muscle action potential and motor unit number estimation of the triceps surae muscles were performed at day 12, 21, and 30 by a single evaluator blinded to genotype and treatment. Similarly, electrical impedance myography was measured on the biceps femoris muscle at 12days for comparison. RESULTS Electrophysiological measures and electrical impedance myography detected significant differences at 12days between control and late-treated (4 or 6days) and sham-treated spinal muscular atrophy mice, but not in mice treated at 2days (p<0.01). EIM findings paralleled and correlated with compound muscle action potential and motor unit number estimation (r=0.61 and r=0.50, respectively, p<0.01). Longitudinal measures at 21 and 30days show that symptomatic therapy results in reduced motor unit number estimation associated with delayed normalization of compound muscle action potential. CONCLUSIONS The incomplete effect of symptomatic treatment is accurately identified by both electrophysiological measures and electrical impedance myography. There is strong correlation between these measures and with weight and righting reflex. This study predicts that measures of compound muscle action potential, motor unit number estimation, and electrical impedance myography are promising biomarkers of treatment stratification and effect for future spinal muscular atrophy trials. The ease of application and simplicity of electrical impedance myography compared with standard electrophysiological measures may be particularly valuable in future pediatric clinical trials.
Collapse
Affiliation(s)
- W Arnold
- Department of Neurology, The Ohio State University Wexner Medical Center, 395 W. 12th Ave, Columbus, OH 43210, United States; Department of Physical Medicine and Rehabilitation, The Ohio State University Wexner Medical Center, 480 Medical Center Drive, Columbus, OH 43210, United States; Department of Neuroscience, The Ohio State University Wexner Medical Center, 480 Medical Center Drive, Columbus, OH 43210, United States
| | - Vicki L McGovern
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, 363 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, United States
| | - Benjamin Sanchez
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Jia Li
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Kaitlyn M Corlett
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, 363 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, United States
| | - Stephen J Kolb
- Department of Neurology, The Ohio State University Wexner Medical Center, 395 W. 12th Ave, Columbus, OH 43210, United States; Department of Neuroscience, The Ohio State University Wexner Medical Center, 480 Medical Center Drive, Columbus, OH 43210, United States; Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, 363 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, United States
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States
| | - Arthur H Burghes
- Department of Neurology, The Ohio State University Wexner Medical Center, 395 W. 12th Ave, Columbus, OH 43210, United States; Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, 363 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, United States
| |
Collapse
|
37
|
Patitucci TN, Ebert AD. SMN deficiency does not induce oxidative stress in SMA iPSC-derived astrocytes or motor neurons. Hum Mol Genet 2015; 25:514-23. [PMID: 26643950 DOI: 10.1093/hmg/ddv489] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/23/2015] [Indexed: 12/18/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a genetic disorder characterized by loss of motor neurons in the spinal cord leading to muscle atrophy and death. Although motor neurons (MNs) are the most obviously affected cells in SMA, recent evidence suggest dysfunction in multiple cell types. Astrocytes are a crucial component of the motor circuit and are intimately involved with MN health and maintenance. We have previously shown that SMA astrocytes are altered both morphologically and functionally early in disease progression, though it is unclear what causes astrocytes to become reactive. Oxidative stress is a common feature among neurodegenerative diseases. Oxidative stress can both induce apoptosis in neurons and can cause astrocytes to become reactive, which are features observed in the SMA induced pluripotent stem cell (iPSC) cultures. Therefore, we asked if oxidative stress contributes to SMA astrocyte pathology. We examined mitochondrial bioenergetics, transcript and protein levels of oxidative and anti-oxidant factors, and reactive oxygen species (ROS) production and found little evidence of oxidative stress. We did observe a significant increase in endogenous catalase expression in SMA iPSCs. While catalase knockdown in SMA iPSCs increased ROS production above basal levels, levels of ROS remained lower than in controls, further arguing against robust oxidative stress in this system. Viral delivery of survival motor neuron (SMN) reversed astrocyte activation and restored catalase levels to normal, without changing mitochondrial respiration or expression of oxidative stress markers. Taken together, these data indicate that SMN deficiency induces astrocyte reactivity, but does not do so through an oxidative stress-mediated process.
Collapse
Affiliation(s)
- Teresa N Patitucci
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, USA
| | - Allison D Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, USA
| |
Collapse
|
38
|
Systemic, postsymptomatic antisense oligonucleotide rescues motor unit maturation delay in a new mouse model for type II/III spinal muscular atrophy. Proc Natl Acad Sci U S A 2015; 112:E5863-72. [PMID: 26460027 DOI: 10.1073/pnas.1509758112] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Clinical presentation of spinal muscular atrophy (SMA) ranges from a neonatal-onset, very severe disease to an adult-onset, milder form. SMA is caused by the mutation of the Survival Motor Neuron 1 (SMN1) gene, and prognosis inversely correlates with the number of copies of the SMN2 gene, a human-specific homolog of SMN1. Despite progress in identifying potential therapies for the treatment of SMA, many questions remain including how late after onset treatments can still be effective and what the target tissues should be. These questions can be addressed in part with preclinical animal models; however, modeling the array of SMA severities in the mouse, which lacks SMN2, has proven challenging. We created a new mouse model for the intermediate forms of SMA presenting with a delay in neuromuscular junction maturation and a decrease in the number of functional motor units, all relevant to the clinical presentation of the disease. Using this new model, in combination with clinical electrophysiology methods, we found that administering systemically SMN-restoring antisense oligonucleotides (ASOs) at the age of onset can extend survival and rescue the neurological phenotypes. Furthermore, these effects were also achieved by administration of the ASOs late after onset, independent of the restoration of SMN in the spinal cord. Thus, by adding to the limited repertoire of existing mouse models for type II/III SMA, we demonstrate that ASO therapy can be effective even when administered after onset of the neurological symptoms, in young adult mice, and without being delivered into the central nervous system.
Collapse
|
39
|
Abstract
Spinal muscular atrophy (SMA) is a leading genetic cause of infant mortality. The disease originates from low levels of SMN protein due to deletion and/or mutations of SMN1 coupled with the inability of SMN2 to compensate for the loss of SMN1. While SMN1 and SMN2 are nearly identical, SMN2 predominantly generates a truncated protein (SMNΔ7) due to skipping of exon 7, the last coding exon. Several avenues for SMA therapy are being explored, including means to enhance SMN2 transcription, correct SMN2 exon 7 splicing, stabilize SMN/SMNΔ7 protein, manipulate SMN-regulated pathways and SMN1 gene delivery by viral vectors. This review focuses on the aspects of target discovery, validations and outcome measures for a promising therapy of SMA.
Collapse
|
40
|
Heier CR, DiDonato CJ. ECG in neonate mice with spinal muscular atrophy allows assessment of drug efficacy. Front Biosci (Elite Ed) 2015; 7:107-16. [PMID: 25553367 DOI: 10.2741/e721] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Molecular technologies have produced diverse arrays of animal models for studying genetic diseases and potential therapeutics. Many have neonatal phenotypes. Spinal muscular atrophy (SMA) is a neuromuscular disorder primarily affecting children, and is of great interest in translational medicine. The most widely used SMA mouse models require all phenotyping to be performed in neonates since they do not survive much past weaning. Pre-clinical studies in neonate mice can be hindered by toxicity and a lack of quality phenotyping assays, since many assays are invalid in pups or require subjective scoring with poor inter-rater variability. We find, however, that passive electrocardiography (ECG) recording in conscious 11-day old SMA mice provides sensitive outcome measures, detecting large differences in heart rate, cardiac conduction, and autonomic control resulting from disease. We find significant drug benefits upon treatment with G418, an aminoglycoside targeting the underlying protein deficiency, even in the absence of overt effects on growth and survival. These findings provide several quantitative physiological biomarkers for SMA preclinical studies, and will be of utility to diverse disease models featuring neonatal cardiac arrhythmias.
Collapse
Affiliation(s)
- Christopher R Heier
- Center for Genetic Medicine Research, Childrens National Medical Center, Washington, DC
| | - Christine J DiDonato
- Center for Genetic Medicine Research, Childrens National Medical Center, Washington, DC
| |
Collapse
|
41
|
Coque E, Raoul C, Bowerman M. ROCK inhibition as a therapy for spinal muscular atrophy: understanding the repercussions on multiple cellular targets. Front Neurosci 2014; 8:271. [PMID: 25221469 PMCID: PMC4148024 DOI: 10.3389/fnins.2014.00271] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/11/2014] [Indexed: 12/28/2022] Open
Abstract
Spinal muscular atrophy (SMA) is the most common genetic disease causing infant death, due to an extended loss of motoneurons. This neuromuscular disorder results from deletions and/or mutations within the Survival Motor Neuron 1 (SMN1) gene, leading to a pathological decreased expression of functional full-length SMN protein. Emerging studies suggest that the small GTPase RhoA and its major downstream effector Rho kinase (ROCK), which both play an instrumental role in cytoskeleton organization, contribute to the pathology of motoneuron diseases. Indeed, an enhanced activation of RhoA and ROCK has been reported in the spinal cord of an SMA mouse model. Moreover, the treatment of SMA mice with ROCK inhibitors leads to an increased lifespan as well as improved skeletal muscle and neuromuscular junction pathology, without preventing motoneuron degeneration. Although motoneurons are the primary target in SMA, an increasing number of reports show that other cell types inside and outside the central nervous system contribute to SMA pathogenesis. As administration of ROCK inhibitors to SMA mice was systemic, the improvement in survival and phenotype could therefore be attributed to specific effects on motoneurons and/or on other non-neuronal cell types. In the present review, we will present the various roles of the RhoA/ROCK pathway in several SMA cellular targets including neurons, myoblasts, glial cells, cardiomyocytes and pancreatic cells as well as discuss how ROCK inhibition may ameliorate their health and function. It is most likely a concerted influence of ROCK modulation on all these cell types that ultimately lead to the observed benefits of pharmacological ROCK inhibition in SMA mice.
Collapse
Affiliation(s)
- Emmanuelle Coque
- The Institute for Neurosciences of Montpellier, Saint Eloi Hospital, Institut National de la Santé et de la Recherche Médicale UMR1051 Montpellier, France ; Université de Montpellier 1 and 2 Montpellier, France
| | - Cédric Raoul
- The Institute for Neurosciences of Montpellier, Saint Eloi Hospital, Institut National de la Santé et de la Recherche Médicale UMR1051 Montpellier, France ; Université de Montpellier 1 and 2 Montpellier, France
| | - Mélissa Bowerman
- The Institute for Neurosciences of Montpellier, Saint Eloi Hospital, Institut National de la Santé et de la Recherche Médicale UMR1051 Montpellier, France ; Université de Montpellier 1 and 2 Montpellier, France
| |
Collapse
|
42
|
Ishigami T, Kino T, Chen L, Minegishi S, Araki N, Umemura M, Abe K, Sasaki R, Yamana H, Umemura S. Identification of bona fide alternative renin transcripts expressed along cortical tubules and potential roles in promoting insulin resistance in vivo without significant plasma renin activity elevation. Hypertension 2014; 64:125-33. [PMID: 24777979 DOI: 10.1161/hypertensionaha.114.03394] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Renin belongs to a family of aspartyl proteases and is the rate-limiting enzyme in the synthesis of the potent vasoactive peptide angiotensin II. Processing of renal renin has been extensively investigated in juxtaglomerular granular cells, in which prorenin and active renin are present in secretory condensed granules. Previous studies demonstrated alternative renin transcription in rat adrenal glands. Different studies reported novel intracellular forms of renin deduced from novel 5' variants derived from renin mRNA in both mice and humans. Comprehensive detailed studies in genetically engineered mice showed that both a secreted and an intracellular form of renin plays divergent mechanism regulating fluid intake and metabolism by the brain renin-angiotensin system; however, the presence, regulation, and functions of these renin isoforms in kidney and adrenal gland are not fully understood in mice. To investigate the characteristics of renin isoforms in mice, we performed a systematic inventory of renin transcripts of mice with and without a duplication of the renin gene alternatively from previous studies. We discovered a novel isoform of renin of the Ren2 gene, which conserved functionally important residues of the prosegment and incomplete isoforms of the Ren1C/D gene lacking a pre-pro segment. In situ hybridization assays revealed alternative renin isoforms expressed along cortical tubules. Newly generated transgenic mice with systemic overexpression of alternative renin transcript showed enhanced local angiotensin II generation without elevation of plasma renin activity and systemic insulin resistance in vivo, providing new pathophysiological insights into insulin resistance exaggerated by bona fide renin isoform.
Collapse
Affiliation(s)
- Tomoaki Ishigami
- From the Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan.
| | - Tabito Kino
- From the Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Lin Chen
- From the Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Shintaro Minegishi
- From the Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Naomi Araki
- From the Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Masanari Umemura
- From the Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Kaito Abe
- From the Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Rie Sasaki
- From the Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Hisako Yamana
- From the Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Satoshi Umemura
- From the Department of Medical Science and Cardio-Renal Medicine, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
43
|
Butchbach MER, Singh J, Gurney ME, Burghes AHM. The effect of diet on the protective action of D156844 observed in spinal muscular atrophy mice. Exp Neurol 2014; 256:1-6. [PMID: 24681157 DOI: 10.1016/j.expneurol.2014.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 03/17/2014] [Indexed: 12/26/2022]
Abstract
Spinal muscular atrophy (SMA) is an early-onset motor neuron disease characterized by loss of spinal motor neurons which leads to skeletal muscle atrophy. Proximal SMA results from the loss or mutation of the survival motor neuron (SMN) gene. In humans, the SMN gene is duplicated to produce two nearly identical genes, SMN1 and SMN2. SMN1 is lost in SMA but SMN2 is retained; in fact, the number of SMN2 copies correlates with disease severity. The SMN2 inducer D156844 increases the survival and improves phenotype of SMN∆7 SMA mice. Maternal diet also modifies the survival and phenotype of these mice. In this study, we show the effect of maternal diet on the protective effects of D156844 in SMN∆7 SMA mice. SMA mice maintained on the PicoLab20 Mouse diet survived longer when treated with D156844; the effect of diet was additive to the effect of D156844 on these mice. Brain levels of D156844 were higher in neonatal mice maintained on the PicoLab20 diet than those on the Harlan-Teklad 22/5 diet. SMN protein levels in the spinal cord were modestly elevated in D156844-treated, PicoLab20-maintained SMA mice. These data show that maternal diet does influence the responsiveness of D156844 in neonatal SMN∆7 SMA mice.
Collapse
Affiliation(s)
- Matthew E R Butchbach
- Department of Molecular and Cellular Biochemistry, College of Medicine, The Ohio State University, Columbus, OH USA; Center for Applied Clinical Genomics, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Center for Pediatric Research, Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE, USA; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, USA.
| | | | | | - Arthur H M Burghes
- Department of Molecular and Cellular Biochemistry, College of Medicine, The Ohio State University, Columbus, OH USA; Department of Neurology, College of Medicine, The Ohio State University, Columbus, OH USA; Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, OH USA
| |
Collapse
|
44
|
Porensky PN, Burghes AHM. Antisense oligonucleotides for the treatment of spinal muscular atrophy. Hum Gene Ther 2013; 24:489-98. [PMID: 23544870 DOI: 10.1089/hum.2012.225] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive disease affecting ∼1 in 10,000 live births. The most striking component is the loss of α-motor neurons in the ventral horn of the spinal cord, resulting in progressive paralysis and eventually premature death. There is no current treatment paradigm other than supportive care, though the past 15 years has seen a striking advancement in understanding of both SMA genetics and molecular mechanisms. A variety of disease-modifying interventions are rapidly bridging the translational gap from the laboratory to clinical trials, including the application of antisense oligonucleotide (ASO) therapy for the correction of aberrant RNA splicing characteristic of SMA. Survival motor neuron (SMN) is a ubiquitously expressed 38-kD protein. Humans have two genes that produce SMN, SMN1 and SMN2, the former of which is deleted or nonfunctional in the majority of patients with SMA. These two genes are nearly identical with one exception, a C to T transition (C6T) within exon 7 of SMN2. C6T disrupts a modulator of splicing, leading to the exclusion of exon 7 from ∼90% of the mRNA transcript. The resultant truncated Δ7SMN protein does not oligomerize efficiently and is rapidly degraded. SMA can therefore be considered a disease of too little SMN protein. A number of cis-acting splice modifiers have been identified in the region of exon 7, the steric block of which enhances the retention of the exon and a resultant full-length mRNA sequence. ASOs targeted to these splice motifs have shown impressive phenotype rescue in multiple SMA mouse models.
Collapse
Affiliation(s)
- Paul N Porensky
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | | |
Collapse
|
45
|
Lutz CM, Osborne MA. Optimizing mouse models of neurodegenerative disorders: are therapeutics in sight? FUTURE NEUROLOGY 2013; 9:67-75. [PMID: 29479291 DOI: 10.2217/fnl.13.66] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The genomic and biologic conservation between mice and humans, along with our increasing ability to manipulate the mouse genome, places the mouse as a premier model for deciphering disease mechanisms and testing potential new therapies. Despite these advantages, mouse models of neurodegenerative disease are sometimes difficult to generate and can present challenges that must be carefully addressed when used for preclinical studies. For those models that do exist, the standardization and optimization of the models is a critical step in ensuring success in both basic research and preclinical use. This review looks back on the history of model development for neurodegenerative diseases and highlights the key strategies that have been learned in order to improve the design, development and use of mouse models in the study of neurodegenerative disease.
Collapse
Affiliation(s)
- Cathleen M Lutz
- The Jackson Laboratory, Genetic Resource Sciences, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Melissa A Osborne
- The Jackson Laboratory, Genetic Resource Sciences, 600 Main Street, Bar Harbor, ME 04609, USA
| |
Collapse
|
46
|
Arnold WD, Burghes AHM. Spinal muscular atrophy: development and implementation of potential treatments. Ann Neurol 2013; 74:348-62. [PMID: 23939659 DOI: 10.1002/ana.23995] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 07/13/2013] [Accepted: 08/01/2013] [Indexed: 12/13/2022]
Abstract
In neurodegenerative disorders, effective treatments are urgently needed, along with methods to determine whether treatment worked. In this review, we discuss the rapid progress in the understanding of recessive proximal spinal muscular atrophy and how this is leading to exciting potential treatments of the disease. Spinal muscular atrophy is caused by loss of the survival motor neuron 1 (SMN1) gene and reduced levels of SMN protein. The critical downstream targets of SMN deficiency that result in motor neuron loss are not known. However, increasing SMN levels has a marked impact in mouse models, and these therapeutics are rapidly moving toward clinical trials. Promising preclinical therapies, the varying degree of impact on the mouse models, and potential measures of treatment effect are reviewed. One key issue discussed is the variable outcome of increasing SMN at different stages of disease progression.
Collapse
Affiliation(s)
- W David Arnold
- Neuromuscular Division, Department of Neurology, Wexner Medical Center, the Ohio State University, Columbus, OH; Department of Physical Medicine and Rehabilitation, Wexner Medical Center, the Ohio State University, Columbus, OH
| | | |
Collapse
|
47
|
Goulet BB, McFall ER, Wong CM, Kothary R, Parks RJ. Supraphysiological expression of survival motor neuron protein from an adenovirus vector does not adversely affect cell function. Biochem Cell Biol 2013; 91:252-64. [DOI: 10.1139/bcb-2012-0094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Spinal muscular atrophy (SMA) is the most common inherited neurodegenerative disease that leads to infant mortality. It is caused by mutations in the survival motor neuron (SMN) protein resulting in death of alpha motor neurons. Increasing evidence suggests that several other tissues are also affected in SMA, including skeletal and cardiac muscle, liver, and pancreas, indicating that systemic delivery of therapeutics may be necessary for true disease correction. Due to the natural biodistribution of therapeutics, a level of SMN several-fold above physiological levels can be achieved in some tissues. In this study, we address whether supraphysiological levels of SMN adversely affects cell function. Infection of a variety of cell types with an adenovirus (Ad) vector encoding SMN leads to very high expression, but the resulting protein correctly localizes within the cell, and associates with normal cellular partners. Although SMN affects transcription of certain target genes and can alter the splicing pattern of others, we did not observe any difference in select target gene splicing or expression in cells overexpressing SMN. However, normal human fibroblasts treated with Ad-SMN showed a slight reduction in growth rate, suggesting that certain cell types may be differently impacted by high levels of SMN.
Collapse
Affiliation(s)
- Benoit B. Goulet
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Emily R. McFall
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Carmen M. Wong
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Robin J. Parks
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
48
|
Shababi M, Lorson CL, Rudnik-Schöneborn SS. Spinal muscular atrophy: a motor neuron disorder or a multi-organ disease? J Anat 2013; 224:15-28. [PMID: 23876144 DOI: 10.1111/joa.12083] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2013] [Indexed: 12/13/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive disorder that is the leading genetic cause of infantile death. SMA is characterized by loss of motor neurons in the ventral horn of the spinal cord, leading to weakness and muscle atrophy. SMA occurs as a result of homozygous deletion or mutations in Survival Motor Neuron-1 (SMN1). Loss of SMN1 leads to a dramatic reduction in SMN protein, which is essential for motor neuron survival. SMA disease severity ranges from extremely severe to a relatively mild adult onset form of proximal muscle atrophy. Severe SMA patients typically die mostly within months or a few years as a consequence of respiratory insufficiency and bulbar paralysis. SMA is widely known as a motor neuron disease; however, there are numerous clinical reports indicating the involvement of additional peripheral organs contributing to the complete picture of the disease in severe cases. In this review, we have compiled clinical and experimental reports that demonstrate the association between the loss of SMN and peripheral organ deficiency and malfunction. Whether defective peripheral organs are a consequence of neuronal damage/muscle atrophy or a direct result of SMN loss will be discussed.
Collapse
Affiliation(s)
- Monir Shababi
- Department of Veterinary Pathobiology, Life Sciences Center, University of Missouri, Columbia, MO, USA; Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| | | | | |
Collapse
|
49
|
Increasing SMN levels using the histone deacetylase inhibitor SAHA ameliorates defects in skeletal muscle microvasculature in a mouse model of severe spinal muscular atrophy. Neurosci Lett 2013; 544:100-4. [DOI: 10.1016/j.neulet.2013.03.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 03/01/2013] [Accepted: 03/25/2013] [Indexed: 01/16/2023]
|
50
|
Cobb MS, Rose FF, Rindt H, Glascock JJ, Shababi M, Miller MR, Osman EY, Yen PF, Garcia ML, Martin BR, Wetz MJ, Mazzasette C, Feng Z, Ko CP, Lorson CL. Development and characterization of an SMN2-based intermediate mouse model of Spinal Muscular Atrophy. Hum Mol Genet 2013; 22:1843-55. [PMID: 23390132 DOI: 10.1093/hmg/ddt037] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Spinal Muscular Atrophy (SMA) is due to the loss of the survival motor neuron gene 1 (SMN1), resulting in motor neuron (MN) degeneration, muscle atrophy and loss of motor function. While SMN2 encodes a protein identical to SMN1, a single nucleotide difference in exon 7 causes most of the SMN2-derived transcripts to be alternatively spliced resulting in a truncated and unstable protein (SMNΔ7). SMA patients retain at least one SMN2 copy, making it an important target for therapeutics. Many of the existing SMA models are very severe, with animals typically living less than 2 weeks. Here, we present a novel intermediate mouse model of SMA based upon the human genomic SMN2 gene. Genetically, this model is similar to the well-characterized SMNΔ7 model; however, we have manipulated the SMNΔ7 transgene to encode a modestly more functional protein referred to as SMN read-through (SMN(RT)). By introducing the SMN(RT) transgene onto the background of a severe mouse model of SMA (SMN2(+/+);Smn(-/-)), disease severity was significantly decreased based upon a battery of phenotypic parameters, including MN pathology and a significant extension in survival. Importantly, there is not a full phenotypic correction, allowing for the examination of a broad range of therapeutics, including SMN2-dependent and SMN-independent pathways. This novel animal model serves as an important biological and therapeutic model for less severe forms of SMA and provides an in vivo validation of the SMN(RT) protein.
Collapse
Affiliation(s)
- Melissa S Cobb
- Department of Veterinary Pathobiology, Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|