Skip to main content

EDITORIAL article

Front. Cell. Neurosci., 17 March 2023
Sec. Cellular Neuropathology
Volume 17 - 2023 | https://doi.org/10.3389/fncel.2023.1178422

Editorial: Advances in spinal muscular atrophy

  • Delaware Center for Neuroscience Research, Department of Biological Sciences, Delaware State University, Dover, DE, United States

Editorial on the Research Topic
Advances in spinal muscular atrophy

Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disorder with an estimated incidence of 1 in 6,000 to 10,000 in live births, and a carrier frequency of 1/40 to 1/60 (Verhaart et al., 2017). Untreated SMA is the leading inherited cause of infant mortality. This disease is characterized by predominate proximal muscle weakness and atrophy. The SMA field has advanced remarkably since the discovery of the causative survival motor neuron 1 (SMN1) gene in 1995 in SMA patients (Lefebvre et al., 1995). However, the mystery of how lower levels of the ubiquitous SMN protein cause the selective motor neuron (MN) degeneration in SMA is still unclear.

SMN is part of a large macromolecular complex consisting of SMN, Gemin 2–8, and Unrip (Pellizzoni et al., 2002). The core member of SMN complex, Gemin3 regulates MN survival through the NF-κB pathway (Arumugam et al., 2018). Miralles et al. in this Frontiers Research Topic presents evidence for an interdependent expression of SMN and Gemin3. They found that Gemin3 knockdown reduced protein expression levels of SMN and NF-κB pathway members, and caused significant neurite degeneration. Interestingly, SMN over-expression did not prevent neurite degeneration caused by the reduction of endogenous Gemin3, which suggested that Germin3 may be involved in cell degeneration independent of SMN.

Accumulating evidence suggests the activation of the tumor suppressor p53 pathway may be a major contributor to MN degeneration in SMA (Murray et al., 2015; Nichterwitz et al., 2020). However, it is not clear which p53 downstream effectors are involved in MN death. A study from the group of Christian M. Simon in this Frontiers Research Topic investigated p53-dependent nuclear upregulation of c-Fos protein in SMA mouse models, and suggested that nuclear c-Fos accumulation may serve as a novel marker for neuronal death in SMA. The necroptotic cell death pathway is another SMN-independent pathological mechanism which may have a role in SMA-associated motor neuron death. Chehade et al. in this Research Topic report on their studies with the receptor-interacting protein kinases (RIPK) RIPK1 and RIPK3, and Caspase-1 necroptosis pathways in the Smn2B/− mouse model of SMA. The triple mutant (TKO) Smn2B/−;Ripk3−/−;Casp1−/− displayed a robust increase in survival and improved motor function compared to Smn2B/− mice. Larger muscle fibers were also observed in the TKO mice. The authors suggest the combination of small-molecule inhibitors of necroptosis and SMN-restoring drugs as a new strategy for SMA treatment. Recent studies suggest that post-translational modifications (PTMs) regulate the pleiotropic functions of SMN complex. In an article in this Research Topic, Riboldi et al. provide a thorough overview of the PTMs that are involved in the regulation of the SMN complex and SMA pathogenesis including phosphorylation, methylation, ubiquitination, acetylation, SUMOylating, and the crosstalk between PTMs of SMN.

The hallmark of SMA is the selective degeneration of alpha motor neurons in the brainstem and spinal cord, therefore it has been considered as a motor neuron-specific disease for decades. However, an increasing body of evidence suggest it involves many other physiological systems indicating it is necessary to consider SMA as a multi-system disorder (Lipnick et al., 2019; Yeo and Darras, 2020). This Research Topic was formally addressed authoritatively for the first time by scientists and clinicians from 11 different country on the 264th ENMC International workshop in Hoofddorp, the Netherlands, November 19–21st 2021. The workshop participants agreed that there is a multi-systemic pathology in SMA. They also summarized scientific evidence for multi-system defects including impaired myogenesis and synaptic development, spleen, immune system and liver involvement, vascularization defects and hypoxia, mitochondrial defects, collagen dysregulation, cardiac dysfunctions, cognitive impairment, metabolic abnormalities, and endocrine alterations (Detering et al., 2022).

Adding to above evidence, two articles in this Research Topic support the multi-systemic nature of SMA. Bonanno et al. evaluated the innate and adaptive immunity pattern in SMA patients before and after nusinersen treatment. They detected a significant increase in a spectrum of cytokines in serum of pediatric and adult SMA patients at baseline including IL-1b, IL-4, IL-6, IL-10, IFN-g, IL-17A, IL-22, IL-23, IL-31, and IL-33. After 6 months of nusinersen treatment, IL-4, IFN-g, Il-22, IL-23, and IL-33 significantly decreased in serum of pediatric SMA patients while IL-4, IL-6, INF-g, and IL-17A were significantly decreased in serum of adult SMA patients. The authors also reported the presence of inflammatory mediators in cerebrospinal fluid. Another article in this Frontiers Research Topic, Cui et al. reports on left ventricular strain imaging and the serum lipid profile related to cardiovascular disease in 80 later-onset SMA and 80 age-, gender-, and body surface area-matched control children. They reported lower measurement of global longitudinal strain and higher measurement of the time to peak longitudinal strain in SMA patients than control. They also reported higher level of total cholesterol, low-density lipoprotein (LDL)/HDL, and Apo B/Apo A1 levels in SMA patients. This data suggests the potential for SMA patients to be at increased risk for cardiomyopathy.

Recent approved SMN-restoring therapies (Sprinraza, Zolgensma, and Everysdi) have significant clinical impact in patients resulting in incremental improvements in motor function and developmental milestones, and preventing worsening of SMA symptoms. However, they are not curative. A significant number of patients respond poorly to these therapies, and their benefits vary among those that do respond (Day et al., 2022; Reilly et al., 2022). In this Frontiers Research Topic, Qiu et al. reviewed the development of nusinersen (the first SMA treatment), its contribution to the development of other drugs, and the potential of novel drug discovery with emerging strategies and new technology such as the clustered regularly interspaced short palindromic repeats (CRISPER) technology. Gene therapy for SMA represents a significant milestone in the treatment of neurologic disease. Zolgensma has demonstrated improved survival and motor milestone achievements for presymptomatic infants and patients with SMA1 (McMillan et al., 2022). A commentary article in this Research Topic from Rossoll and Singh discusses the challenges associated with SMA gene therapy including preparation, administration and the efficient body-wide distribution of the SMN1 gene delivered with the AAV9 vector. They also provide perspectives for the development of the next generation of gene therapies including alternative or additional routes of delivery, newer generation of AAV capsids, and optimized promoters and other tissue and cell-type specific control elements.

In summary, the insightful articles published in this Frontiers Research Topic provide new findings on the mechanisms of SMN pathology, evidence for multi-systematic defects in SMA, and perspectives for future treatment development. They also advocate for in-depth study of the systematic mechanisms of SMA pathology for identifying novel therapeutic targets for combinatorial approaches with SMN-restoring gene therapies.

Author contributions

The author confirms being the sole contributor of this work and has approved it for publication.

Acknowledgments

All the contributors to this Research Topic, the reviewers for their insightful and prudent reviews, and the Frontiers editorial staffs for the great support are greatly appreciated. The author thanks the comments from Melissa Harrington on the manuscript.

Conflict of interest

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Arumugam, S., Mincheva-Tasheva, S., Periyakaruppiah, A., de la Fuente, S., Soler, R. M., and Garcera, A. (2018). Regulation of survival motor neuron protein by the nuclear factor-kappa B pathway in mouse spinal cord motoneurons. Mol. Neurobiol. 55, 5019–5030. doi: 10.1007/s12035-017-0710-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Day, J. W., Howell, K., Place, A., Long, K., Rossello, J., Kertesz, N., et al. (2022). Advances and limitations for the treatment of spinal muscular atrophy. BMC Pediatr. 22, 632. doi: 10.1186/s12887-022-03671-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Detering, N. T., Zambon, A., Hensel, N., Kothary, R., Swoboda, K., Gillingwater, T. H., et al. (2022). 264th ENMC International Workshop: Multi-system involvement in spinal muscular atrophy Hoofddorp, the Netherlands, November 19th – 21st 2021. Neuromuscul. Disord. 32, 697–705. doi: 10.1016/j.nmd.2022.06.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Lefebvre, S., Bürglen, L., Reboullet, S., Clermont, O., Burlet, P., Viollet, L., et al. (1995). Identification and characterization of a spinal muscular atrophy-determining gene. Cell 80, 155–165. doi: 10.1016/0092-8674(95)90460-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Lipnick, S. L., Agniel, D. M., Aggarwal, R., Makhortova, N. R., Finlayson, S. G., Brocato, A., et al. (2019). Systemic nature of spinal muscular atrophy revealed by studying insurance claims. PLoS ONE 14, e0213680. doi: 10.1371/journal.pone.0213680

PubMed Abstract | CrossRef Full Text | Google Scholar

McMillan, H. J., Proud, C. M., Farrar, M. A., Alexander, I. E., Muntoni, F., and Servais, L. (2022). Onasemnogene abeparvovec for the treatment of spinal muscular atrophy. Expert Opin. Biol. Ther. 22, 1075–1090. doi: 10.1080/14712598.2022.2066471

PubMed Abstract | CrossRef Full Text | Google Scholar

Murray, L. M., Beauvais, A., Gibeault, S., Courtney, N. L., and Kothary, R. (2015). Transcriptional profiling of differentially vulnerable motor neurons at pre-symptomatic stage in the Smn2b/-mouse model of spinal muscular atrophy. Acta Neuropathol. Commun. 3, 55. doi: 10.1186/s40478-015-0231-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Nichterwitz, S., Nijssen, J., Storvall, H., Schweingruber, C., Comley, L. H., Allodi, I., et al. (2020). LCM-seq reveals unique transcriptional adaptation mechanisms of resistant neurons and identifies protective pathways in spinal muscular atrophy. Genome Res. 30, 1083–1096. doi: 10.1101/gr.265017.120

PubMed Abstract | CrossRef Full Text | Google Scholar

Pellizzoni, L., Yong, J., and Dreyfuss, G. (2002). Essential role for the SMN complex in the specificity of snRNP assembly. Science 298, 1775–1779. doi: 10.1126/science.1074962

PubMed Abstract | CrossRef Full Text | Google Scholar

Reilly, A., Chehade, L., and Kothary, R. (2022). Curing SMA: Are we there yet? Gene Therapy 22, 349. doi: 10.1038/s41434-022-00349-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Verhaart, I. E. C., Robertson, A., Leary, R., McMacken, G., König, K., Kirschner, J., et al. (2017). A multi-source approach to determine SMA incidence and research ready population. J. Neurol. 264, 1465–1473. doi: 10.1007/s00415-017-8549-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Yeo, C. J. J., and Darras, B. T. (2020). Overturning the paradigm of spinal muscular atrophy as just a motor neuron disease. Pediatr. Neurol. 109, 12–19. doi: 10.1016/j.pediatrneurol.2020.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: spinal muscular atrophy, survival motor neuron protein (SMN), pathology, multi-system, SMN-restoring therapies

Citation: Sun J (2023) Editorial: Advances in spinal muscular atrophy. Front. Cell. Neurosci. 17:1178422. doi: 10.3389/fncel.2023.1178422

Received: 02 March 2023; Accepted: 07 March 2023;
Published: 17 March 2023.

Edited and reviewed by: Bradley J. Turner, University of Melbourne, Australia

Copyright © 2023 Sun. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jianli Sun, jianlisun@desu.edu

Download