1
|
McLaurin DM, Tucker SK, Siddique SJ, Challagundla L, Gibert Y, Hebert MD. A Novel Role for Coilin in Vertebrate Innate Immunity. FASEB J 2025; 39:e70580. [PMID: 40277349 PMCID: PMC12023821 DOI: 10.1096/fj.202403276r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/04/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
Coilin is a protein localized in the nucleus, where it plays a role in the assembly of the Cajal Body and is involved in ribonucleoprotein biogenesis. Our recent research has uncovered new roles for coilin, including its involvement in producing microRNAs and in modifying other proteins through phosphorylation and SUMOylation. We also proposed that coilin could respond to stress signals. In plants, coilin has been shown to help regulate immune genes and activate defense mechanisms, especially in response to stress. In this study, we used two vertebrate models to study coilin function: a human primary foreskin fibroblast cell line deficient in coilin through RNA interference and a newly created zebrafish line with a mutation in the coilin gene generated by CRISPR-Cas9. Transcriptomic analysis in these two models of coilin deficiency revealed dysregulation of immunity-related genes in both species. To phenotypically validate the transcriptomic results, we challenged zebrafish coilin mutants with lipopolysaccharide (LPS), which triggers an innate immune response, and identified an attenuated response to LPS in vivo in the zebrafish coilin mutants. Our results support a vital novel function for coilin in vertebrates in regulating the expression of immunity-related genes. Moreover, these findings could lead to more research on how coilin regulates innate immunity in animals and humans.
Collapse
Affiliation(s)
- Douglas M. McLaurin
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| | - Sara K. Tucker
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| | - Shanzida J. Siddique
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| | - Lavanya Challagundla
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| | - Yann Gibert
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| | - Michael D. Hebert
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
2
|
Chudakova D, Kuzenkova L, Fisenko A, Savostyanov K. In Search of Spinal Muscular Atrophy Disease Modifiers. Int J Mol Sci 2024; 25:11210. [PMID: 39456991 PMCID: PMC11508272 DOI: 10.3390/ijms252011210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
The 5q Spinal Muscular Atrophy (SMA) is a hereditary autosomal recessive disease caused by defects in the survival motor neuron (SMN1) gene encoding survival motor neuron (SMN) protein. Currently, it is the leading cause of infantile mortality worldwide. SMA is a progressive neurodegenerative disease with "continuum of clinical severity", which can be modulated by genetic and epigenetic factors known as disease modifiers (DMs). Individuals (even siblings) with the same defects in SMN1 gene might have strikingly different types of SMA, supposedly due to the impact of DMs. There are several therapeutic options for SMA, all of them focusing on the restoration of the SMN protein levels to normal. Determining DMs and the pathways in which they are involved might aid in enhancing existing curative approaches. Furthermore, DMs might become novel therapeutic targets or prognostic biomarkers of the disease. This narrative review provides a brief overview of the genetics and pathobiology of SMA, and its bona fide modifiers. We describe novel, emerging DMs, approaches and tools used to identify them, as well as their potential mechanisms of action and impact on disease severity. We also propose several disease-modifying molecular mechanisms which could provide a partial explanation of the staggering variability of SMA phenotypes.
Collapse
Affiliation(s)
| | | | | | - Kirill Savostyanov
- National Medical Research Center of Children’s Health of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| |
Collapse
|
3
|
Schüning T, Zeug A, Strienke K, Franz P, Tsiavaliaris G, Hensel N, Viero G, Ponimaskin E, Claus P. The spinal muscular atrophy gene product regulates actin dynamics. FASEB J 2024; 38:e70055. [PMID: 39305126 DOI: 10.1096/fj.202300183r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/31/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024]
Abstract
Spinal Muscular Atrophy (SMA) is a neuromuscular disease caused by low levels of the Survival of Motoneuron (SMN) protein. SMN interacts with and regulates the actin-binding protein profilin2a, thereby influencing actin dynamics. Dysfunctional actin dynamics caused by SMN loss disrupts neurite outgrowth, axonal pathfinding, and formation of functional synapses in neurons. Whether the SMN protein directly interacts with and regulates filamentous (F-) and monomeric globular (G-) actin is still elusive. In a quantitative single cell approach, we show that SMN loss leads to dysregulated F-/G-actin fractions. Furthermore, quantitative assessment of cell morphology suggests an F-actin organizational defect. Interestingly, this is mediated by an interaction of SMN with G- and F-actin. In co-immunoprecipitation, in-vitro pulldown and co-localization assays, we elucidated that this interaction is independent of the SMN-profilin2a interaction. Therefore, we suggest two populations being relevant for functional actin dynamics in healthy neurons: SMN-profilin2a-actin and SMN-actin. Additionally, those two populations may influence each other and therefore regulate binding of SMN to actin. In SMA, we showed a dysregulated co-localization pattern of SMN-actin which could only partially rescued by SMN restoration. However, dysregulation of F-/G-actin fractions was reduced by SMN restoration. Taken together, our results suggest a novel molecular function of SMN in binding to actin independent from SMN-profilin2a interaction.
Collapse
Affiliation(s)
- Tobias Schüning
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Hannover, Germany
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Andre Zeug
- Institute of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Katharina Strienke
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Hannover, Germany
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Peter Franz
- Cellular Biophysics, Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Georgios Tsiavaliaris
- Cellular Biophysics, Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Niko Hensel
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Gabriella Viero
- Institute of Biophysics (IBF), CNR Unit at Trento, Trento, Italy
| | - Evgeni Ponimaskin
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Peter Claus
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
4
|
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
|
5
|
Cottam NC, Bamfo T, Harrington MA, Charvet CJ, Hekmatyar K, Tulin N, Sun J. Cerebellar structural, astrocytic, and neuronal abnormalities in the SMNΔ7 mouse model of spinal muscular atrophy. Brain Pathol 2023; 33:e13162. [PMID: 37218083 PMCID: PMC10467044 DOI: 10.1111/bpa.13162] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Spinalmuscular atrophy (SMA) is a neuromuscular disease that affects as many as 1 in 6000 individuals at birth, making it the leading genetic cause of infant mortality. A growing number of studies indicate that SMA is a multi-system disease. The cerebellum has received little attention even though it plays an important role in motor function and widespread pathology has been reported in the cerebella of SMA patients. In this study, we assessed SMA pathology in the cerebellum using structural and diffusion magnetic resonance imaging, immunohistochemistry, and electrophysiology with the SMNΔ7 mouse model. We found a significant disproportionate loss in cerebellar volume, decrease in afferent cerebellar tracts, selective lobule-specific degeneration of Purkinje cells, abnormal lobule foliation and astrocyte integrity, and a decrease in spontaneous firing of cerebellar output neurons in the SMA mice compared to controls. Our data suggest that defects in cerebellar structure and function due to decreased survival motor neuron (SMN) levels impair the functional cerebellar output affecting motor control, and that cerebellar pathology should be addressed to achieve comprehensive treatment and therapy for SMA patients.
Collapse
Affiliation(s)
- Nicholas C. Cottam
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
| | - Tiffany Bamfo
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
| | | | - Christine J. Charvet
- Delaware Center for Neuroscience ResearchDelaware State UniversityDoverDelawareUSA
- Department of Anatomy, Physiology and PharmacologyAuburn UniversityAuburnAlabamaUSA
- Department of PsychologyDelaware State UniversityDoverDEUnited States
| | - Khan Hekmatyar
- Center for Biomedical and Brain ImagingUniversity of DelawareNewarkDelawareUSA
- Bioimaging Research Center for Biomedical and Brain ImagingUniversity of GeorgiaAthensGeorgiaUSA
| | - Nikita Tulin
- Department of NeuroscienceTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Jianli Sun
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
- Delaware Center for Neuroscience ResearchDelaware State UniversityDoverDelawareUSA
| |
Collapse
|
6
|
Detering NT, Schüning T, Hensel N, Claus P. The phospho-landscape of the survival of motoneuron protein (SMN) protein: relevance for spinal muscular atrophy (SMA). Cell Mol Life Sci 2022; 79:497. [PMID: 36006469 PMCID: PMC11071818 DOI: 10.1007/s00018-022-04522-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/27/2022] [Accepted: 08/09/2022] [Indexed: 11/03/2022]
Abstract
Spinal muscular atrophy (SMA) is caused by low levels of the survival of motoneuron (SMN) Protein leading to preferential degeneration of lower motoneurons in the ventral horn of the spinal cord and brain stem. However, the SMN protein is ubiquitously expressed and there is growing evidence of a multisystem phenotype in SMA. Since a loss of SMN function is critical, it is important to decipher the regulatory mechanisms of SMN function starting on the level of the SMN protein itself. Posttranslational modifications (PTMs) of proteins regulate multiple functions and processes, including activity, cellular trafficking, and stability. Several PTM sites have been identified within the SMN sequence. Here, we map the identified SMN PTMs highlighting phosphorylation as a key regulator affecting localization, stability and functions of SMN. Furthermore, we propose SMN phosphorylation as a crucial factor for intracellular interaction and cellular distribution of SMN. We outline the relevance of phosphorylation of the spinal muscular atrophy (SMA) gene product SMN with regard to basic housekeeping functions of SMN impaired in this neurodegenerative disease. Finally, we compare SMA patient mutations with putative and verified phosphorylation sites. Thus, we emphasize the importance of phosphorylation as a cellular modulator in a clinical perspective as a potential additional target for combinatorial SMA treatment strategies.
Collapse
Affiliation(s)
- Nora Tula Detering
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Tobias Schüning
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Niko Hensel
- Ottawa Hospital Research Institute (OHRI), Ottawa, Canada
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Peter Claus
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Hannover, Germany.
- Center for Systems Neuroscience (ZSN), Hannover, Germany.
| |
Collapse
|
7
|
Brown SJ, Kline RA, Synowsky SA, Shirran SL, Holt I, Sillence KA, Claus P, Wirth B, Wishart TM, Fuller HR. The Proteome Signatures of Fibroblasts from Patients with Severe, Intermediate and Mild Spinal Muscular Atrophy Show Limited Overlap. Cells 2022; 11:cells11172624. [PMID: 36078032 PMCID: PMC9454632 DOI: 10.3390/cells11172624] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 12/04/2022] Open
Abstract
Most research to characterise the molecular consequences of spinal muscular atrophy (SMA) has focused on SMA I. Here, proteomic profiling of skin fibroblasts from severe (SMA I), intermediate (SMA II), and mild (SMA III) patients, alongside age-matched controls, was conducted using SWATH mass spectrometry analysis. Differentially expressed proteomic profiles showed limited overlap across each SMA type, and variability was greatest within SMA II fibroblasts, which was not explained by SMN2 copy number. Despite limited proteomic overlap, enriched canonical pathways common to two of three SMA severities with at least one differentially expressed protein from the third included mTOR signalling, regulation of eIF2 and eIF4 signalling, and protein ubiquitination. Network expression clustering analysis identified protein profiles that may discriminate or correlate with SMA severity. From these clusters, the differential expression of PYGB (SMA I), RAB3B (SMA II), and IMP1 and STAT1 (SMA III) was verified by Western blot. All SMA fibroblasts were transfected with an SMN-enhanced construct, but only RAB3B expression in SMA II fibroblasts demonstrated an SMN-dependent response. The diverse proteomic profiles and pathways identified here pave the way for studies to determine their utility as biomarkers for patient stratification or monitoring treatment efficacy and for the identification of severity-specific treatments.
Collapse
Affiliation(s)
- Sharon J. Brown
- School of Pharmacy and Bioengineering (PhaB), Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Rachel A. Kline
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK
- Euan MacDonald Centre, University of Edinburgh, Edinburgh EH16 4SB, 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
| | - Ian Holt
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | | | - Peter Claus
- SMATHERIA gGmbH—Non-Profit Biomedical Research Institute, 30625 Hannover, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Rare Diseases, 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, 50931 Cologne, Germany
| | - Thomas M. Wishart
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, UK
- Euan MacDonald Centre, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Heidi R. Fuller
- School of Pharmacy and Bioengineering (PhaB), Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
- Correspondence: ; Tel.: +44-(0)1-782-734546
| |
Collapse
|
8
|
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: 19] [Impact Index Per Article: 4.8] [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
|
9
|
Rademacher S, Detering NT, Schüning T, Lindner R, Santonicola P, Wefel IM, Dehus J, Walter LM, Brinkmann H, Niewienda A, Janek K, Varela MA, Bowerman M, Di Schiavi E, Claus P. A Single Amino Acid Residue Regulates PTEN-Binding and Stability of the Spinal Muscular Atrophy Protein SMN. Cells 2020; 9:cells9112405. [PMID: 33153033 PMCID: PMC7692393 DOI: 10.3390/cells9112405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022] Open
Abstract
Spinal Muscular Atrophy (SMA) is a neuromuscular disease caused by decreased levels of the survival of motoneuron (SMN) protein. Post-translational mechanisms for regulation of its stability are still elusive. Thus, we aimed to identify regulatory phosphorylation sites that modulate function and stability. Our results show that SMN residues S290 and S292 are phosphorylated, of which SMN pS290 has a detrimental effect on protein stability and nuclear localization. Furthermore, we propose that phosphatase and tensin homolog (PTEN), a novel phosphatase for SMN, counteracts this effect. In light of recent advancements in SMA therapies, a significant need for additional approaches has become apparent. Our study demonstrates S290 as a novel molecular target site to increase the stability of SMN. Characterization of relevant kinases and phosphatases provides not only a new understanding of SMN function, but also constitutes a novel strategy for combinatorial therapeutic approaches to increase the level of SMN in SMA.
Collapse
Affiliation(s)
- Sebastian Rademacher
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany; (S.R.); (N.T.D.); (T.S.); (R.L.); (I.-M.W.); (J.D.); (L.M.W.); (H.B.)
| | - Nora T. Detering
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany; (S.R.); (N.T.D.); (T.S.); (R.L.); (I.-M.W.); (J.D.); (L.M.W.); (H.B.)
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Tobias Schüning
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany; (S.R.); (N.T.D.); (T.S.); (R.L.); (I.-M.W.); (J.D.); (L.M.W.); (H.B.)
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Robert Lindner
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany; (S.R.); (N.T.D.); (T.S.); (R.L.); (I.-M.W.); (J.D.); (L.M.W.); (H.B.)
| | - Pamela Santonicola
- Institute of Biosciences and Bioresources, National Research Council of Italy, 80131 Naples, Italy; (P.S.); (E.D.S.)
| | - Inga-Maria Wefel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany; (S.R.); (N.T.D.); (T.S.); (R.L.); (I.-M.W.); (J.D.); (L.M.W.); (H.B.)
| | - Janina Dehus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany; (S.R.); (N.T.D.); (T.S.); (R.L.); (I.-M.W.); (J.D.); (L.M.W.); (H.B.)
| | - Lisa M. Walter
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany; (S.R.); (N.T.D.); (T.S.); (R.L.); (I.-M.W.); (J.D.); (L.M.W.); (H.B.)
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Hella Brinkmann
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany; (S.R.); (N.T.D.); (T.S.); (R.L.); (I.-M.W.); (J.D.); (L.M.W.); (H.B.)
| | - Agathe Niewienda
- Shared Facility for Mass Spectrometry, Institute of Biochemistry, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (A.N.); (K.J.)
| | - Katharina Janek
- Shared Facility for Mass Spectrometry, Institute of Biochemistry, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (A.N.); (K.J.)
| | - Miguel A. Varela
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; (M.A.V.); (M.B.)
- Department of Paediatrics, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Melissa Bowerman
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; (M.A.V.); (M.B.)
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Elia Di Schiavi
- Institute of Biosciences and Bioresources, National Research Council of Italy, 80131 Naples, Italy; (P.S.); (E.D.S.)
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany; (S.R.); (N.T.D.); (T.S.); (R.L.); (I.-M.W.); (J.D.); (L.M.W.); (H.B.)
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
- Correspondence:
| |
Collapse
|
10
|
Sluzalska KD, Slawski J, Sochacka M, Lampart A, Otlewski J, Zakrzewska M. Intracellular partners of fibroblast growth factors 1 and 2 - implications for functions. Cytokine Growth Factor Rev 2020; 57:93-111. [PMID: 32475760 DOI: 10.1016/j.cytogfr.2020.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
Fibroblast growth factors 1 and 2 (FGF1 and FGF2) are mainly considered as ligands of surface receptors through which they regulate a broad spectrum of biological processes. They are secreted in non-canonical way and, unlike other growth factors, they are able to translocate from the endosome to the cell interior. These unique features, as well as the role of the intracellular pool of FGF1 and FGF2, are far from being fully understood. An increasing number of reports address this problem, focusing on the intracellular interactions of FGF1 and 2. Here, we summarize the current state of knowledge of the FGF1 and FGF2 binding partners inside the cell and the possible role of these interactions. The partner proteins are grouped according to their function, including proteins involved in secretion, cell signaling, nucleocytoplasmic transport, binding and processing of nucleic acids, ATP binding, and cytoskeleton assembly. An in-depth analysis of the network of these binding partners could indicate novel, non-classical functions of FGF1 and FGF2 and uncover an additional level of a fine control of the well-known FGF-regulated cellular processes.
Collapse
Affiliation(s)
- Katarzyna Dominika Sluzalska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Jakub Slawski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Martyna Sochacka
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Agata Lampart
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Jacek Otlewski
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Malgorzata Zakrzewska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
11
|
Schill Y, Bijata M, Kopach O, Cherkas V, Abdel-Galil D, Böhm K, Schwab MH, Matsuda M, Compan V, Basu S, Bijata K, Wlodarczyk J, Bard L, Cole N, Dityatev A, Zeug A, Rusakov DA, Ponimaskin E. Serotonin 5-HT 4 receptor boosts functional maturation of dendritic spines via RhoA-dependent control of F-actin. Commun Biol 2020; 3:76. [PMID: 32060357 PMCID: PMC7021812 DOI: 10.1038/s42003-020-0791-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/23/2020] [Indexed: 01/24/2023] Open
Abstract
Activity-dependent remodeling of excitatory connections underpins memory formation in the brain. Serotonin receptors are known to contribute to such remodeling, yet the underlying molecular machinery remains poorly understood. Here, we employ high-resolution time-lapse FRET imaging in neuroblastoma cells and neuronal dendrites to establish that activation of serotonin receptor 5-HT4 (5-HT4R) rapidly triggers spatially-restricted RhoA activity and G13-mediated phosphorylation of cofilin, thus locally boosting the filamentous actin fraction. In neuroblastoma cells, this leads to cell rounding and neurite retraction. In hippocampal neurons in situ, 5-HT4R-mediated RhoA activation triggers maturation of dendritic spines. This is paralleled by RhoA-dependent, transient alterations in cell excitability, as reflected by increased spontaneous synaptic activity, apparent shunting of evoked synaptic responses, and enhanced long-term potentiation of excitatory transmission. The 5-HT4R/G13/RhoA signaling thus emerges as a previously unrecognized molecular pathway underpinning use-dependent functional remodeling of excitatory synaptic connections.
Collapse
Affiliation(s)
- Yvonne Schill
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Monika Bijata
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Olga Kopach
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Volodymyr Cherkas
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Dalia Abdel-Galil
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Katrin Böhm
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Markus H Schwab
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Michiyuki Matsuda
- Bioimaging and Cell Signaling, Kyoto University, Kyoto, 606-8501, Japan
| | | | - Subhadip Basu
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
- Computer Science and Engineering, Jadavpur University, Kolkata, 700032, India
| | - Krystian Bijata
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Jakub Wlodarczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093, Warsaw, Poland
| | - Lucie Bard
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Nicholas Cole
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120, Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke-University, Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Dmitri A Rusakov
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
12
|
Hensel N, Kubinski S, Claus P. The Need for SMN-Independent Treatments of Spinal Muscular Atrophy (SMA) to Complement SMN-Enhancing Drugs. Front Neurol 2020; 11:45. [PMID: 32117013 PMCID: PMC7009174 DOI: 10.3389/fneur.2020.00045] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/13/2020] [Indexed: 12/25/2022] Open
Abstract
Spinal Muscular Atrophy (SMA) is monogenic motoneuron disease caused by low levels of the Survival of Motoneuron protein (SMN). Recently, two different drugs were approved for the treatment of the disease. The antisense oligonucleotide Nusinersen/Spinraza® and the gene replacement therapy Onasemnogene Abeparvovec/Zolgensma® both enhance SMN levels. These treatments result in impressive benefits for the patients. However, there is a significant number of non-responders and an intervention delay has a strong negative impact on the efficacy. Obviously, later stages of motoneuron degeneration cannot be reversed by SMN-restoration. Therefore, complementary, SMN-independent strategies are needed which are able to address such SMN-irreversible degenerative processes. Those are defined as pathological alterations which are not reversed by SMN-restoration for a given dose and intervention delay. It is crucial to tailor SMN-independent approaches to the novel clinical situation with SMN-restoring treatments. On the molecular level, such SMN-irreversible changes become manifest in altered signaling modules as described by molecular systems biology. Based on our current knowledge about altered signaling, we introduce a network approach for an informed decision for the most potent SMN-independent treatment targets. Finally, we present recommendations for the identification of novel treatments which can be combined with SMN-restoring drugs.
Collapse
Affiliation(s)
- Niko Hensel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,Center of Systems Neuroscience (ZSN), Hannover, Germany
| | - Sabrina Kubinski
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,Center of Systems Neuroscience (ZSN), Hannover, Germany
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,Center of Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
13
|
Hoolachan JM, Sutton ER, Bowerman M. Teaching an old drug new tricks: repositioning strategies for spinal muscular atrophy. FUTURE NEUROLOGY 2019. [DOI: 10.2217/fnl-2019-0006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Spinal muscular atrophy (SMA) is a childhood disorder caused by loss of the SMN gene. Pathological hallmarks are spinal cord motor neuron death, neuromuscular junction dysfunction and muscle atrophy. The first SMN genetic therapy was recently approved and other SMN-dependent treatments are not far behind. However, not all SMA patients will reap their maximal benefit due to limited accessibility, high costs and differential effects depending on timing of administration and disease severity. The repurposing of commercially available drugs is an interesting strategy to ensure more rapid and less expensive access to new treatments. In this mini-review, we will discuss the potential and relevance of repositioning drugs currently used for neurodegenerative, neuromuscular and muscle disorders for SMA.
Collapse
Affiliation(s)
- Joseph M Hoolachan
- School of Medicine, Keele University, Staffordshire, ST5 5BG, UK
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, ST5 5BG, UK
| | - Emma R Sutton
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, ST5 5BG, UK
| | - Melissa Bowerman
- School of Medicine, Keele University, Staffordshire, ST5 5BG, UK
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry, SY10 7AG, UK
| |
Collapse
|
14
|
Kounakis K, Tavernarakis N. The Cytoskeleton as a Modulator of Aging and Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1178:227-245. [PMID: 31493230 DOI: 10.1007/978-3-030-25650-0_12] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cytoskeleton consists of filamentous protein polymers that form organized structures, contributing to a multitude of cell life aspects. It includes three types of polymers: the actin microfilaments, the microtubules and the intermediate filaments. Decades of research have implicated the cytoskeleton in processes that regulate cellular and organismal aging, as well as neurodegeneration associated with injury or neurodegenerative disease, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic Lateral Sclerosis, or Charcot Marie Tooth disease. Here, we provide a brief overview of cytoskeletal structure and function, and discuss experimental evidence linking cytoskeletal function and dynamics with aging and neurodegeneration.
Collapse
Affiliation(s)
- Konstantinos Kounakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece.,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece. .,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece.
| |
Collapse
|
15
|
Chaytow H, Huang YT, Gillingwater TH, Faller KME. The role of survival motor neuron protein (SMN) in protein homeostasis. Cell Mol Life Sci 2018; 75:3877-3894. [PMID: 29872871 PMCID: PMC6182345 DOI: 10.1007/s00018-018-2849-1] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 12/11/2022]
Abstract
Ever since loss of survival motor neuron (SMN) protein was identified as the direct cause of the childhood inherited neurodegenerative disorder spinal muscular atrophy, significant efforts have been made to reveal the molecular functions of this ubiquitously expressed protein. Resulting research demonstrated that SMN plays important roles in multiple fundamental cellular homeostatic pathways, including a well-characterised role in the assembly of the spliceosome and biogenesis of ribonucleoproteins. More recent studies have shown that SMN is also involved in other housekeeping processes, including mRNA trafficking and local translation, cytoskeletal dynamics, endocytosis and autophagy. Moreover, SMN has been shown to influence mitochondria and bioenergetic pathways as well as regulate function of the ubiquitin-proteasome system. In this review, we summarise these diverse functions of SMN, confirming its key role in maintenance of the homeostatic environment of the cell.
Collapse
Affiliation(s)
- Helena Chaytow
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Yu-Ting Huang
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Thomas H Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK.
| | - Kiterie M E Faller
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
16
|
de la Fuente S, Sansa A, Periyakaruppiah A, Garcera A, Soler RM. Calpain Inhibition Increases SMN Protein in Spinal Cord Motoneurons and Ameliorates the Spinal Muscular Atrophy Phenotype in Mice. Mol Neurobiol 2018; 56:4414-4427. [PMID: 30327977 PMCID: PMC6505520 DOI: 10.1007/s12035-018-1379-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/01/2018] [Indexed: 01/07/2023]
Abstract
Spinal muscular atrophy (SMA), a leading genetic cause of infant death, is caused by the loss of survival motor neuron 1 (SMN1) gene. SMA is characterized by the degeneration and loss of spinal cord motoneurons (MNs), muscular atrophy, and weakness. SMN2 is the centromeric duplication of the SMN gene, whose numbers of copies determine the intracellular levels of SMN protein and define the disease onset and severity. It has been demonstrated that elevating SMN levels can be an important strategy in treating SMA and can be achieved by several mechanisms, including promotion of protein stability. SMN protein is a direct target of the calcium-dependent protease calpain and induces its proteolytic cleavage in muscle cells. In this study, we examined the involvement of calpain in SMN regulation on MNs. In vitro experiments showed that calpain activation induces SMN cleavage in CD1 and SMA mouse spinal cord MNs. Additionally, calpain 1 knockdown or inhibition increased SMN level and prevent neurite degeneration in these cells. We examined the effects of calpain inhibition on the phenotype of two severe SMA mouse models. Treatment with the calpain inhibitor, calpeptin, significantly improved the lifespan and motor function of these mice. Our observations show that calpain regulates SMN level in MNs and calpeptin administration improves SMA phenotype demonstrating the potential utility of calpain inhibitors in SMA therapy.
Collapse
Affiliation(s)
- Sandra de la Fuente
- Unitat de Senyalització Neuronal, Department Medicina Experimental, Universitat de Lleida-IRBLleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Alba Sansa
- Unitat de Senyalització Neuronal, Department Medicina Experimental, Universitat de Lleida-IRBLleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Ambika Periyakaruppiah
- Unitat de Senyalització Neuronal, Department Medicina Experimental, Universitat de Lleida-IRBLleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Ana Garcera
- Unitat de Senyalització Neuronal, Department Medicina Experimental, Universitat de Lleida-IRBLleida, Rovira Roure 80, 25198, Lleida, Spain
| | - Rosa M Soler
- Unitat de Senyalització Neuronal, Department Medicina Experimental, Universitat de Lleida-IRBLleida, Rovira Roure 80, 25198, Lleida, Spain.
| |
Collapse
|
17
|
Dayangac-Erden D, Gur-Dedeoglu B, Eskici FN, Oztemur-Islakoglu Y, Erdem-Ozdamar S. Do Perineuronal Net Elements Contribute to Pathophysiology of Spinal Muscular Atrophy? In Vitro and Transcriptomics Insights. ACTA ACUST UNITED AC 2018; 22:598-606. [DOI: 10.1089/omi.2018.0106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Didem Dayangac-Erden
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | | | - Fatma Nazli Eskici
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | | | - Sevim Erdem-Ozdamar
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
18
|
Pletto D, Capra S, Finardi A, Colciaghi F, Nobili P, Battaglia GS, Locatelli D, Cagnoli C. Axon outgrowth and neuronal differentiation defects after a-SMN and FL-SMN silencing in primary hippocampal cultures. PLoS One 2018; 13:e0199105. [PMID: 29902268 PMCID: PMC6001960 DOI: 10.1371/journal.pone.0199105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 05/31/2018] [Indexed: 12/30/2022] Open
Abstract
Spinal Muscular Atrophy (SMA) is a severe autosomal recessive disease characterized by selective motor neuron degeneration, caused by disruptions of the Survival of Motor Neuron 1 (Smn1) gene. The main product of SMN1 is the full-length SMN protein (FL-SMN), that plays an established role in mRNA splicing. FL-SMN is also involved in neurite outgrowth and axonal transport. A shorter SMN isoform, axonal-SMN or a-SMN, displays a more specific axonal localization and has remarkable axonogenic properties in NSC-34. Introduction of known SMA mutations into the a-SMN transcript leads to impairment of axon growth and morphological defects similar to those observed in SMA patients and animal models. Although there is increasing evidence for the relevance of SMN axonal functions in SMA pathogenesis, the specific contributions of FL-SMN and a-SMN are not known yet. This work aimed to analyze the differential roles of FL-SMN and a-SMN in axon outgrowth and in neuronal homeostasis during differentiation of neurons into a mature phenotype. We employed primary cultures of hippocampal neurons as a well-defined model of polarization and differentiation. By analyzing subcellular localization, we showed that a-SMN is preferentially localized in the growing axonal compartment. By specifically silencing FL-SMN or a-SMN proteins, we demonstrated that both proteins play a role in axon growth, as their selective down-regulation reduces axon length without affecting dendritic arborization. a-SMN silencing, and in minor extent FL-SMN silencing, resulted in the growth of multi-neuritic neurons, impaired in the differentiation process of selecting a single axon out of multiple neurites. In these neurons, neurites often display mixed axonal and dendritic markers and abnormal distribution of the axonal initial segment protein Ankirin G, suggesting loss of neuronal polarity. Our results indicate that a-SMN and FL-SMN are needed for neuronal polarization and organization of axonal and dendritic compartments, processes that are fundamental for neuronal function and survival.
Collapse
Affiliation(s)
- Daniela Pletto
- Molecular Neuroanatomy and Pathogenesis Unit, Neurology VII—Clinical and Experimental Epileptology Unit, Foundation IRCCS Neurological Institute “C. Besta”, Milano, Italy
| | - Silvia Capra
- Molecular Neuroanatomy and Pathogenesis Unit, Neurology VII—Clinical and Experimental Epileptology Unit, Foundation IRCCS Neurological Institute “C. Besta”, Milano, Italy
| | - Adele Finardi
- Molecular Neuroanatomy and Pathogenesis Unit, Neurology VII—Clinical and Experimental Epileptology Unit, Foundation IRCCS Neurological Institute “C. Besta”, Milano, Italy
| | - Francesca Colciaghi
- Molecular Neuroanatomy and Pathogenesis Unit, Neurology VII—Clinical and Experimental Epileptology Unit, Foundation IRCCS Neurological Institute “C. Besta”, Milano, Italy
| | - Paola Nobili
- Molecular Neuroanatomy and Pathogenesis Unit, Neurology VII—Clinical and Experimental Epileptology Unit, Foundation IRCCS Neurological Institute “C. Besta”, Milano, Italy
| | - Giorgio Stefano Battaglia
- Molecular Neuroanatomy and Pathogenesis Unit, Neurology VII—Clinical and Experimental Epileptology Unit, Foundation IRCCS Neurological Institute “C. Besta”, Milano, Italy
| | - Denise Locatelli
- Molecular Neuroanatomy and Pathogenesis Unit, Neurology VII—Clinical and Experimental Epileptology Unit, Foundation IRCCS Neurological Institute “C. Besta”, Milano, Italy
| | - Cinzia Cagnoli
- Molecular Neuroanatomy and Pathogenesis Unit, Neurology VII—Clinical and Experimental Epileptology Unit, Foundation IRCCS Neurological Institute “C. Besta”, Milano, Italy
- * E-mail:
| |
Collapse
|
19
|
Rademacher S, Verheijen BM, Hensel N, Peters M, Bora G, Brandes G, Vieira de Sá R, Heidrich N, Fischer S, Brinkmann H, van der Pol WL, Wirth B, Pasterkamp RJ, Claus P. Metalloprotease-mediated cleavage of PlexinD1 and its sequestration to actin rods in the motoneuron disease spinal muscular atrophy (SMA). Hum Mol Genet 2018; 26:3946-3959. [PMID: 29016853 DOI: 10.1093/hmg/ddx282] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/11/2017] [Indexed: 12/12/2022] Open
Abstract
Cytoskeletal rearrangement during axon growth is mediated by guidance receptors and their ligands which act either as repellent, attractant or both. Regulation of the actin cytoskeleton is disturbed in Spinal Muscular Atrophy (SMA), a devastating neurodegenerative disease affecting mainly motoneurons, but receptor-ligand interactions leading to the dysregulation causing SMA are poorly understood. In this study, we analysed the role of the guidance receptor PlexinD1 in SMA pathogenesis. We showed that PlexinD1 is cleaved by metalloproteases in SMA and that this cleavage switches its function from an attractant to repellent. Moreover, we found that the PlexinD1 cleavage product binds to actin rods, pathological aggregate-like structures which had so far been described for age-related neurodegenerative diseases. Our data suggest a novel disease mechanism for SMA involving formation of actin rods as a molecular sink for a cleaved PlexinD1 fragment leading to dysregulation of receptor signaling.
Collapse
Affiliation(s)
- Sebastian Rademacher
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Bert M Verheijen
- Department of Translational Neuroscience & MIND Facility, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands.,Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Niko Hensel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Miriam Peters
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Center for Rare Diseases Cologne, and Institute of Genetics, University of Cologne, 50931 Cologne, Germany
| | - Gamze Bora
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, 06100 Ankara, Turkey
| | - Gudrun Brandes
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Renata Vieira de Sá
- Department of Translational Neuroscience & MIND Facility, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Natascha Heidrich
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Silke Fischer
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Hella Brinkmann
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany
| | - W Ludo van der Pol
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Center for Rare Diseases Cologne, and Institute of Genetics, University of Cologne, 50931 Cologne, Germany
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience & MIND Facility, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Peter Claus
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, 30625 Hannover, Germany.,Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
20
|
Abstract
Spinal muscular atrophy (SMA) is a motor neuron disease caused by mutations/deletions within the survival of motor neuron 1 (SMN1) gene that lead to a pathological reduction of SMN protein levels. SMN is part of a multiprotein complex, functioning as a molecular chaperone that facilitates the assembly of spliceosomal small nuclear ribonucleoproteins (snRNP). In addition to its role in spliceosome formation, SMN has also been found to interact with mRNA-binding proteins (mRBPs), and facilitate their assembly into mRNP transport granules. The association of protein and RNA in RNP complexes plays an important role in an extensive and diverse set of cellular processes that regulate neuronal growth, differentiation, and the maturation and plasticity of synapses. This review discusses the role of SMN in RNP assembly and localization, focusing on molecular defects that affect mRNA processing and may contribute to SMA pathology.
Collapse
|
21
|
Hensel N, Claus P. The Actin Cytoskeleton in SMA and ALS: How Does It Contribute to Motoneuron Degeneration? Neuroscientist 2017; 24:54-72. [PMID: 28459188 DOI: 10.1177/1073858417705059] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA) are neurodegenerative diseases with overlapping clinical phenotypes based on impaired motoneuron function. However, the pathomechanisms of both diseases are largely unknown, and it is still unclear whether they converge on the molecular level. SMA is a monogenic disease caused by low levels of functional Survival of Motoneuron (SMN) protein, whereas ALS involves multiple genes as well as environmental factors. Recent evidence argues for involvement of actin regulation as a causative and dysregulated process in both diseases. ALS-causing mutations in the actin-binding protein profilin-1 as well as the ability of the SMN protein to directly bind to profilins argue in favor of a common molecular mechanism involving the actin cytoskeleton. Profilins are major regulat ors of actin-dynamics being involved in multiple neuronal motility and transport processes as well as modulation of synaptic functions that are impaired in models of both motoneuron diseases. In this article, we review the current literature in SMA and ALS research with a focus on the actin cytoskeleton. We propose a common molecular mechanism that explains the degeneration of motoneurons for SMA and some cases of ALS.
Collapse
Affiliation(s)
- Niko Hensel
- 1 Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,2 Niedersachsen Network on Neuroinfectiology (N-RENNT), Hannover, Germany
| | - Peter Claus
- 1 Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,2 Niedersachsen Network on Neuroinfectiology (N-RENNT), Hannover, Germany.,3 Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
22
|
Wertz MH, Winden K, Neveu P, Ng SY, Ercan E, Sahin M. Cell-type-specific miR-431 dysregulation in a motor neuron model of spinal muscular atrophy. Hum Mol Genet 2016; 25:2168-2181. [PMID: 27005422 DOI: 10.1093/hmg/ddw084] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/11/2016] [Indexed: 12/17/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal-recessive pediatric neurodegenerative disease characterized by selective loss of spinal motor neurons. It is caused by mutation in the survival of motor neuron 1, SMN1, gene and leads to loss of function of the full-length SMN protein. microRNAs (miRNAs) are small RNAs that are involved in post-transcriptional regulation of gene expression. Prior studies have implicated miRNAs in the pathogenesis of motor neuron disease. We hypothesized that motor neuron-specific miRNA expression changes are involved in their selective vulnerability in SMA. Therefore, we sought to determine the effect of SMN loss on miRNAs and their target mRNAs in spinal motor neurons. We used microarray and RNAseq to profile both miRNA and mRNA expression in primary spinal motor neuron cultures after acute SMN knockdown. By integrating the miRNA:mRNA profiles, a number of dysregulated miRNAs were identified with enrichment in differentially expressed putative mRNA targets. miR-431 expression was highly increased, and a number of its putative mRNA targets were significantly downregulated in motor neurons after SMN loss. Further, we found that miR-431 regulates motor neuron neurite length by targeting several molecules previously identified to play a role in motor neuron axon outgrowth, including chondrolectin. Together, our findings indicate that cell-type-specific dysregulation of miR-431 plays a role in the SMA motor neuron phenotype.
Collapse
Affiliation(s)
- Mary H Wertz
- Department of Neurology, The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kellen Winden
- Department of Neurology, The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pierre Neveu
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Shi-Yan Ng
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA Neurotherapeutics Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore
| | - Ebru Ercan
- Department of Neurology, The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mustafa Sahin
- Department of Neurology, The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Theme 10 Molecular Cell Biology. Amyotroph Lateral Scler Frontotemporal Degener 2015; 16 Suppl 1:176-85. [PMID: 26517031 DOI: 10.3109/21678421.2015.1098815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
24
|
Li H, Custer SK, Gilson T, Hao LT, Beattie CE, Androphy EJ. α-COP binding to the survival motor neuron protein SMN is required for neuronal process outgrowth. Hum Mol Genet 2015; 24:7295-307. [PMID: 26464491 DOI: 10.1093/hmg/ddv428] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/06/2015] [Indexed: 01/30/2023] Open
Abstract
Spinal muscular atrophy (SMA), a heritable neurodegenerative disease, results from insufficient levels of the survival motor neuron (SMN) protein. α-COP binds to SMN, linking the COPI vesicular transport pathway to SMA. Reduced levels of α-COP restricted development of neuronal processes in NSC-34 cells and primary cortical neurons. Remarkably, heterologous expression of human α-COP restored normal neurite length and morphology in SMN-depleted NSC-34 cells in vitro and zebrafish motor neurons in vivo. We identified single amino acid mutants of α-COP that selectively abrogate SMN binding, retain COPI-mediated Golgi-ER trafficking functionality, but were unable to support neurite outgrowth in cellular and zebrafish models of SMA. Taken together, these demonstrate the functional role of COPI association with the SMN protein in neuronal development.
Collapse
Affiliation(s)
- Hongxia Li
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA and
| | - Sara K Custer
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA and
| | - Timra Gilson
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA and
| | - Le Thi Hao
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Christine E Beattie
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Elliot J Androphy
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA and
| |
Collapse
|
25
|
Morano M, Wrobel S, Fregnan F, Ziv-Polat O, Shahar A, Ratzka A, Grothe C, Geuna S, Haastert-Talini K. Nanotechnology versus stem cell engineering: in vitro comparison of neurite inductive potentials. Int J Nanomedicine 2014; 9:5289-306. [PMID: 25484582 PMCID: PMC4238897 DOI: 10.2147/ijn.s71951] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Innovative nerve conduits for peripheral nerve reconstruction are needed in order to specifically support peripheral nerve regeneration (PNR) whenever nerve autotransplantation is not an option. Specific support of PNR could be achieved by neurotrophic factor delivery within the nerve conduits via nanotechnology or stem cell engineering and transplantation. Methods Here, we comparatively investigated the bioactivity of selected neurotrophic factors conjugated to iron oxide nanoparticles (np-NTFs) and of bone marrow-derived stem cells genetically engineered to overexpress those neurotrophic factors (NTF-BMSCs). The neurite outgrowth inductive activity was monitored in culture systems of adult and neonatal rat sensory dorsal root ganglion neurons as well as in the cell line from rat pheochromocytoma (PC-12) cell sympathetic culture model system. Results We demonstrate that np-NTFs reliably support numeric neurite outgrowth in all utilized culture models. In some aspects, especially with regard to their long-term bioactivity, np-NTFs are even superior to free NTFs. Engineered NTF-BMSCs proved to be less effective in induction of sensory neurite outgrowth but demonstrated an increased bioactivity in the PC-12 cell culture system. In contrast, primary nontransfected BMSCs were as effective as np-NTFs in sensory neurite induction and demonstrated an impairment of neuronal differentiation in the PC-12 cell system. Conclusion Our results evidence that nanotechnology as used in our setup is superior over stem cell engineering when it comes to in vitro models for PNR. Furthermore, np-NTFs can easily be suspended in regenerative hydrogel matrix and could be delivered that way to nerve conduits for future in vivo studies and medical application.
Collapse
Affiliation(s)
- Michela Morano
- Department of Clinical and Biological Sciences, Università Degli Studi di Torino, Orbassano, Piemonte, Italy
| | - Sandra Wrobel
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Lower-Saxony, Germany ; Center for Systems Neuroscience (ZSN), Hannover, Lower-Saxony, Germany
| | - Federica Fregnan
- Department of Clinical and Biological Sciences, Università Degli Studi di Torino, Orbassano, Piemonte, Italy
| | | | | | - Andreas Ratzka
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Lower-Saxony, Germany
| | - Claudia Grothe
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Lower-Saxony, Germany ; Center for Systems Neuroscience (ZSN), Hannover, Lower-Saxony, Germany
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, Università Degli Studi di Torino, Orbassano, Piemonte, Italy
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Lower-Saxony, Germany ; Center for Systems Neuroscience (ZSN), Hannover, Lower-Saxony, Germany
| |
Collapse
|
26
|
D'Ambrosi N, Rossi S, Gerbino V, Cozzolino M. Rac1 at the crossroad of actin dynamics and neuroinflammation in Amyotrophic Lateral Sclerosis. Front Cell Neurosci 2014; 8:279. [PMID: 25249940 PMCID: PMC4157560 DOI: 10.3389/fncel.2014.00279] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/22/2014] [Indexed: 12/11/2022] Open
Abstract
Rac1 is a major player of the Rho family of small GTPases that controls multiple cell signaling pathways, such as the organization of cytoskeleton (including adhesion and motility), cell proliferation, apoptosis and activation of immune cells. In the nervous system, in particular, Rac1 GTPase plays a key regulatory function of both actin and microtubule cytoskeletal dynamics and thus it is central to axonal growth and stability, as well as dendrite and spine structural plasticity. Rac1 is also a crucial regulator of NADPH-dependent membrane oxidase (NOX), a prominent source of reactive oxygen species (ROS), thus having a central role in the inflammatory response and neurotoxicity mediated by microglia cells in the nervous system. As such, alterations in Rac1 activity might well be involved in the processes that give rise to Amyotrophic Lateral Sclerosis (ALS), a complex syndrome where cytoskeletal disturbances in motor neurons and redox alterations in the inflammatory compartment play pivotal and synergic roles in the final disease outcomes. Here we will discuss the genetic and mechanistic evidence indicating the relevance of Rac1 dysregulation in the pathogenesis of ALS.
Collapse
Affiliation(s)
- Nadia D'Ambrosi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Simona Rossi
- National Research Council, Institute of Translational Pharmacology Rome, Italy ; Fondazione Santa Lucia IRCCS Rome, Italy
| | - Valeria Gerbino
- Fondazione Santa Lucia IRCCS Rome, Italy ; Department of Biology, Università di Roma Tor Vergata Rome, Italy
| | - Mauro Cozzolino
- National Research Council, Institute of Translational Pharmacology Rome, Italy
| |
Collapse
|
27
|
Investigations of curcumin and resveratrol on neurite outgrowth: perspectives on spinal muscular atrophy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:709108. [PMID: 25105137 PMCID: PMC4101952 DOI: 10.1155/2014/709108] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/08/2014] [Accepted: 06/05/2014] [Indexed: 12/18/2022]
Abstract
Spinal Muscular Atrophy (SMA) is an autosomal recessive neurodegenerative disease with progressive muscle weakness and atrophy. SMA is caused by low levels of the Survival of Motor Neuron (SMN) protein, which also leads to neurite outgrowth defects in neuronal cells. Rescue of the outgrowth defect is thought to be a strategy for SMA treatment. Polyphenolic histone deacetylase (HDAC) inhibitors might be good candidates due to their neuritogenic properties. In the present study, it was investigated whether neurite outgrowth defects could be rescued by curcumin and resveratrol, which are SMN-inducing polyphenols, having HDAC inhibition activity. According to our results, although curcumin and resveratrol failed to restore the neurite outgrowth defects, the SMN protein was found to be necessary for the neurite-promoting activity of curcumin in neuron-like PC12 cells.
Collapse
|
28
|
SMN is required for the maintenance of embryonic stem cells and neuronal differentiation in mice. Brain Struct Funct 2014; 220:1539-53. [PMID: 24633826 DOI: 10.1007/s00429-014-0743-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 02/28/2014] [Indexed: 01/02/2023]
Abstract
Survival motor neuron (SMN) is the determining factor in spinal muscular atrophy, the most common genetic cause of childhood mortality. We have previously found that SMN regulates stem cell division, proliferation and differentiation in Drosophila. However, it is unknown whether a similar effect exists in vertebrates. Here, we show that SMN is enriched in highly proliferative embryonic stem cells (ESCs) in mice and reduction of SMN impairs the pluripotency of ESCs. Moreover, we find that SMN reduction activates ERK signaling and affects neuronal differentiation in vitro. Teratomas with reduced SMN grow more slowly and show weaker signals of neuronal differentiation than those with a normal level of SMN. Finally, we show that over-expression of SMN is protective for ESCs from retinoic acid-induced differentiation. Taken together, our results suggest that SMN plays a role in the maintenance of pluripotent ESCs and neuronal differentiation in mice.
Collapse
|
29
|
Bowerman M, Michalski JP, Beauvais A, Murray LM, DeRepentigny Y, Kothary R. Defects in pancreatic development and glucose metabolism in SMN-depleted mice independent of canonical spinal muscular atrophy neuromuscular pathology. Hum Mol Genet 2014; 23:3432-44. [PMID: 24497575 PMCID: PMC4049303 DOI: 10.1093/hmg/ddu052] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Spinal muscular atrophy (SMA) is characterized by motor neuron loss, caused by mutations or deletions in the ubiquitously expressed survival motor neuron 1 (SMN1) gene. We recently identified a novel role for Smn protein in glucose metabolism and pancreatic development in both an intermediate SMA mouse model (Smn(2B/-)) and type I SMA patients. In the present study, we sought to determine if the observed metabolic and pancreatic defects are SMA-dependent. We employed a line of heterozygous Smn-depleted mice (Smn(+/-)) that lack the hallmark SMA neuromuscular pathology and overt phenotype. At 1 month of age, pancreatic/metabolic function of Smn(+/-)mice is indistinguishable from wild type. However, when metabolically challenged with a high-fat diet, Smn(+/-)mice display abnormal localization of glucagon-producing α-cells within the pancreatic islets and increased hepatic insulin and glucagon sensitivity, through increased p-AKT and p-CREB, respectively. Further, aging results in weight gain, an increased number of insulin-producing β cells, hyperinsulinemia and increased hepatic glucagon sensitivity in Smn(+/-)mice. Our study uncovers and highlights an important function of Smn protein in pancreatic islet development and glucose metabolism, independent of canonical SMA pathology. These findings suggest that carriers of SMN1 mutations and/or deletions may be at an increased risk of developing pancreatic and glucose metabolism defects, as even small depletions in Smn protein may be a risk factor for diet- and age-dependent development of metabolic disorders.
Collapse
Affiliation(s)
- Melissa Bowerman
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada The Neuroscience Institute of Montpellier (INM), Inserm UMR1051, Saint Eloi Hospital, Montpellier, France
| | - John-Paul Michalski
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada Department of Cellular and Molecular Medicine and
| | | | | | | | - Rashmi Kothary
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada Department of Cellular and Molecular Medicine and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
30
|
Förthmann B, van Bergeijk J, Lee YW, Lübben V, Schill Y, Brinkmann H, Ratzka A, Stachowiak MK, Hebert M, Grothe C, Claus P. Regulation of neuronal differentiation by proteins associated with nuclear bodies. PLoS One 2013; 8:e82871. [PMID: 24358231 PMCID: PMC3866168 DOI: 10.1371/journal.pone.0082871] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 11/06/2013] [Indexed: 12/17/2022] Open
Abstract
Nuclear bodies are large sub-nuclear structures composed of RNA and protein molecules. The Survival of Motor Neuron (SMN) protein localizes to Cajal bodies (CBs) and nuclear gems. Diminished cellular concentration of SMN is associated with the neurodegenerative disease Spinal Muscular Atrophy (SMA). How nuclear body architecture and its structural components influence neuronal differentiation remains elusive. In this study, we analyzed the effects of SMN and two of its interaction partners in cellular models of neuronal differentiation. The nuclear 23 kDa isoform of Fibroblast Growth Factor - 2 (FGF-2(23)) is one of these interacting proteins - and was previously observed to influence nuclear bodies by destabilizing nuclear gems and mobilizing SMN from Cajal bodies (CBs). Here we demonstrate that FGF-2(23) blocks SMN-promoted neurite outgrowth, and also show that SMN disrupts FGF-2(23)-dependent transcription. Our results indicate that FGF-2(23) and SMN form an inactive complex that interferes with neuronal differentiation by mutually antagonizing nuclear functions. Coilin is another nuclear SMN binding partner and a marker protein for Cajal bodies (CBs). In addition, coilin is essential for CB function in maturation of small nuclear ribonucleoprotein particles (snRNPs). The role of coilin outside of Cajal bodies and its putative impacts in tissue differentiation are poorly defined. The present study shows that protein levels of nucleoplasmic coilin outside of CBs decrease during neuronal differentiation. Overexpression of coilin has an inhibitory effect on neurite outgrowth. Furthermore, we find that nucleoplasmic coilin inhibits neurite outgrowth independent of SMN binding revealing a new function for coilin in neuronal differentiation.
Collapse
Affiliation(s)
- Benjamin Förthmann
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Yu-Wei Lee
- Department of Pathology and Anatomical Sciences, State University of New York, Buffalo, New York, United States of America
| | - Verena Lübben
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
| | - Yvonne Schill
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
| | - Hella Brinkmann
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
| | - Andreas Ratzka
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
| | - Michal K. Stachowiak
- Department of Pathology and Anatomical Sciences, State University of New York, Buffalo, New York, United States of America
| | - Michael Hebert
- Department of Biochemistry, The University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Claudia Grothe
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Peter Claus
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, University of Veterinary Medicine Hannover, Hannover, Germany
- * E-mail:
| |
Collapse
|
31
|
Hensel N, Stockbrügger I, Rademacher S, Broughton N, Brinkmann H, Grothe C, Claus P. Bilateral crosstalk of rho- and extracellular-signal-regulated-kinase (ERK) pathways is confined to an unidirectional mode in spinal muscular atrophy (SMA). Cell Signal 2013; 26:540-8. [PMID: 24316236 DOI: 10.1016/j.cellsig.2013.11.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/15/2013] [Accepted: 11/26/2013] [Indexed: 10/26/2022]
Abstract
Rho-kinase (ROCK) as well as extracellular signal regulated kinase (ERK) control actin cytoskeletal organization thereby regulating dynamic changes of cellular morphology. In neurons, motility processes such as axonal guidance and neurite outgrowth demand a fine regulation of upstream pathways. Here we demonstrate a bilateral ROCK-ERK information flow in neurons. This process is shifted towards an unidirectional crosstalk in a model of the neurodegenerative disease Spinal Muscular Atrophy (SMA), ultimately leading to neurite outgrowth dysregulations. As both pathways are of therapeutic relevance for SMA, our results argue for a combinatorial ROCK/ERK-targeting as a future treatment strategy.
Collapse
Affiliation(s)
- Niko Hensel
- Institute of Neuroanatomy, Hannover Medical School, 30625 Hannover, Germany; Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Inga Stockbrügger
- Institute of Neuroanatomy, Hannover Medical School, 30625 Hannover, Germany
| | - Sebastian Rademacher
- Institute of Neuroanatomy, Hannover Medical School, 30625 Hannover, Germany; Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Natasha Broughton
- Institute of Neuroanatomy, Hannover Medical School, 30625 Hannover, Germany
| | - Hella Brinkmann
- Institute of Neuroanatomy, Hannover Medical School, 30625 Hannover, Germany
| | - Claudia Grothe
- Institute of Neuroanatomy, Hannover Medical School, 30625 Hannover, Germany; Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Peter Claus
- Institute of Neuroanatomy, Hannover Medical School, 30625 Hannover, Germany; Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany; Niedersachsen Research Network on Neuroinfection (N-RENNT), Germany.
| |
Collapse
|
32
|
Lunke S, El-Osta A. Applicability of histone deacetylase inhibition for the treatment of spinal muscular atrophy. Neurotherapeutics 2013; 10:677-87. [PMID: 23996601 PMCID: PMC3805858 DOI: 10.1007/s13311-013-0209-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Spinal muscular atrophy (SMA), a neurodegenerative disease with potentially devastating and even deadly effects on affected individuals, was first described in the late nineteenth century. Although the survival of motor neuron (SMN) gene was identified nearly 2 decades ago to be causative of the disease, neither an effective treatment nor a cure are currently available. Yet efforts are on-going to test a multitude of treatment strategies with the potential to alleviate disease symptoms in human and clinical trials. Among the most studied compounds for the treatment of SMA are histone deacetylase inhibitors. Several of these epigenetic modifiers have been shown to increase expression of the crucial SMN gene in vitro and in vivo, an effect linked to increased histone acetylation and remodeling of the chromatin landscape surrounding the SMN gene promoter. Here, we review the history and current state of use of histone deacetylase inhibitors in SMA, as well as the success of clinical trials investigating the clinical applicability of these epigenetic modifiers in SMA treatment.
Collapse
Affiliation(s)
- Sebastian Lunke
- />Epigenetics in Human Health and Disease Laboratory, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004 Australia
- />Translational Genomics Laboratory, Centre for Translational Pathology, Department of Pathology, University of Melbourne, Melbourne, VIC 3010 Australia
| | - Assam El-Osta
- />Epigenetics in Human Health and Disease Laboratory, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC 3004 Australia
- />Epigenomics Profiling Facility, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, VIC Australia
- />Department of Pathology, The University of Melbourne, Melbourne, VIC Australia
- />Faculty of Medicine, Monash University, Monash, VIC Australia
| |
Collapse
|
33
|
Sleigh JN, Barreiro-Iglesias A, Oliver PL, Biba A, Becker T, Davies KE, Becker CG, Talbot K. Chondrolectin affects cell survival and neuronal outgrowth in in vitro and in vivo models of spinal muscular atrophy. Hum Mol Genet 2013; 23:855-69. [PMID: 24067532 DOI: 10.1093/hmg/ddt477] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Spinal muscular atrophy (SMA) is characterized by the selective loss of spinal motor neurons owing to reduced levels of survival motor neuron (Smn) protein. In addition to its well-established role in assembling constituents of the spliceosome, diverse cellular functions have been proposed for Smn, but the reason why low levels of this widely expressed protein result in selective motor neuron pathology is still debated. In longitudinal studies of exon-level changes in SMA mouse model tissues, designed to determine the contribution of splicing dysfunction to the disease, we have previously shown that a generalized defect in splicing is unlikely to play a causative role in SMA. Nevertheless, we identified a small subset of genes that were alternatively spliced in the spinal cord compared with control mice before symptom onset, indicating a possible mechanistic role in disease. Here, we have performed functional studies of one of these genes, chondrolectin (Chodl), known to be highly expressed in motor neurons and important for correct motor axon outgrowth in zebrafish. Using in vitro and in vivo models of SMA, we demonstrate altered expression of Chodl in SMA mouse spinal motor neurons, show that Chodl has distinct effects on cell survival and neurite outgrowth and that increasing the expression of chodl can rescue motor neuron outgrowth defects in Smn-depleted zebrafish. Our findings thus link the dysregulation of Chodl to the pathophysiology of motor neuron degeneration in SMA.
Collapse
Affiliation(s)
- James N Sleigh
- Department of Physiology, Anatomy and Genetics, MRC Functional Genomics Unit, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Förthmann B, Brinkmann H, Ratzka A, Stachowiak MK, Grothe C, Claus P. Immobile survival of motoneuron (SMN) protein stored in Cajal bodies can be mobilized by protein interactions. Cell Mol Life Sci 2013; 70:2555-68. [PMID: 23334184 PMCID: PMC11113639 DOI: 10.1007/s00018-012-1242-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 11/29/2012] [Accepted: 12/10/2012] [Indexed: 12/25/2022]
Abstract
Reduced levels of survival of motoneuron (SMN) protein lead to spinal muscular atrophy, but it is still unknown how SMN protects motoneurons in the spinal cord against degeneration. In the nucleus, SMN is associated with two types of nuclear bodies denoted as gems and Cajal bodies (CBs). The 23 kDa isoform of fibroblast growth factor-2 (FGF-2(23)) is a nuclear protein that binds to SMN and destabilizes the SMN-Gemin2 complex. In the present study, we show that FGF-2(23) depletes SMN from CBs without affecting their general structure. FRAP analysis of SMN-EGFP in CBs demonstrated that the majority of SMN in CBs remained mobile and allowed quantification of fast, slow and immobile nuclear SMN populations. The potential for SMN release was confirmed by in vivo photoconversion of SMN-Dendra2, indicating that CBs concentrate immobile SMN that could have a specialized function in CBs. FGF-2(23) accelerated SMN release from CBs, accompanied by a conversion of immobile SMN into a mobile population. Furthermore, FGF-2(23) caused snRNP accumulation in CBs. We propose a model in which Cajal bodies store immobile SMN that can be mobilized by its nuclear interaction partner FGF-2(23), leading to U4 snRNP accumulation in CBs, indicating a role for immobile SMN in tri-snRNP assembly.
Collapse
Affiliation(s)
- Benjamin Förthmann
- Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
- Center for Systems Neuroscience, 30625 Hannover, Germany
| | - Hella Brinkmann
- Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| | - Andreas Ratzka
- Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
| | - Michal K. Stachowiak
- Department of Pathology and Anatomical Sciences, State University of New York, Buffalo, NY 14214 USA
| | - Claudia Grothe
- Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
- Center for Systems Neuroscience, 30625 Hannover, Germany
| | - Peter Claus
- Institute of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany
- Center for Systems Neuroscience, 30625 Hannover, Germany
| |
Collapse
|
35
|
Groen EJN, Fumoto K, Blokhuis AM, Engelen-Lee J, Zhou Y, van den Heuvel DMA, Koppers M, van Diggelen F, van Heest J, Demmers JAA, Kirby J, Shaw PJ, Aronica E, Spliet WGM, Veldink JH, van den Berg LH, Pasterkamp RJ. ALS-associated mutations in FUS disrupt the axonal distribution and function of SMN. Hum Mol Genet 2013; 22:3690-704. [PMID: 23681068 DOI: 10.1093/hmg/ddt222] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mutations in the RNA binding protein fused in sarcoma/translated in liposarcoma (FUS/TLS) cause amyotrophic lateral sclerosis (ALS). Although ALS-linked mutations in FUS often lead to a cytosolic mislocalization of the protein, the pathogenic mechanisms underlying these mutations remain poorly understood. To gain insight into these mechanisms, we examined the biochemical, cell biological and functional properties of mutant FUS in neurons. Expression of different FUS mutants (R521C, R521H, P525L) in neurons caused axonal defects. A protein interaction screen performed to explain these phenotypes identified numerous FUS interactors including the spinal muscular atrophy (SMA) causing protein survival motor neuron (SMN). Biochemical experiments showed that FUS and SMN interact directly and endogenously, and that this interaction can be regulated by FUS mutations. Immunostaining revealed co-localization of mutant FUS aggregates and SMN in primary neurons. This redistribution of SMN to cytosolic FUS accumulations led to a decrease in axonal SMN. Finally, cell biological experiments showed that overexpression of SMN rescued the axonal defects induced by mutant FUS, suggesting that FUS mutations cause axonal defects through SMN. This study shows that neuronal aggregates formed by mutant FUS protein may aberrantly sequester SMN and concomitantly cause a reduction of SMN levels in the axon, leading to axonal defects. These data provide a functional link between ALS-linked FUS mutations, SMN and neuronal connectivity and support the idea that different motor neuron disorders such as SMA and ALS may be caused, in part, by defects in shared molecular pathways.
Collapse
Affiliation(s)
- Ewout J N Groen
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Actin isoforms in neuronal development and function. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:157-213. [PMID: 23317819 DOI: 10.1016/b978-0-12-407704-1.00004-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The actin cytoskeleton contributes directly or indirectly to nearly every aspect of neuronal development and function. This diversity of functions is often attributed to actin regulatory proteins, although how the composition of the actin cytoskeleton itself may influence its function is often overlooked. In neurons, the actin cytoskeleton is composed of two distinct isoforms, β- and γ-actin. Functions for β-actin have been investigated in axon guidance, synaptogenesis, and disease. Insight from loss-of-function in vivo studies has also revealed novel roles for β-actin in select brain structures and behaviors. Conversely, very little is known regarding functions of γ-actin in neurons. The dysregulation or mutation of both β- and γ-actin has been implicated in multiple human neurological disorders, however, demonstrating the critical importance of these still poorly understood proteins. This chapter highlights what is currently known regarding potential distinct functions for β- and γ-actin in neurons as well as the significant areas that remain unexplored.
Collapse
|
37
|
Bosio Y, Berto G, Camera P, Bianchi F, Ambrogio C, Claus P, Di Cunto F. PPP4R2 regulates neuronal cell differentiation and survival, functionally cooperating with SMN. Eur J Cell Biol 2012; 91:662-74. [PMID: 22559936 DOI: 10.1016/j.ejcb.2012.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Revised: 02/08/2012] [Accepted: 03/12/2012] [Indexed: 01/28/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a human disease caused by reduced levels of the Survival of Motor Neuron (SMN) protein, leading to progressive loss of motor neurons and muscular paralysis. However, it is still not very clear why these cells are specifically sensitive to SMN levels. Therefore, understanding which proteins may functionally interact with SMN in a neuronal context is a very important issue. PPP4R2, a regulatory subunit of the protein phosphatase 4 (PPP4C), was previously identified as a functional interactor of the SMN complex, but has never been studied in neuronal cells. In this report, we show that PPP4R2 displays a very dynamic intracellular localization in mouse and rat neuronal cell lines and in rat primary hippocampal neurons, strongly correlating with differentiation. More importantly, we found that PPP4R2 loss of function impairs the differentiation of the mouse motor-neuronal cell line NSC-34, an effect that can be counteracted by SMN overexpression. In addition, we show that PPP4R2 may specifically protect NSC-34 cells from DNA damage-induced apoptosis and that it is capable to functionally cooperate with SMN in this activity. Our data indicate that PPP4R2 is a SMN partner that may modulate the differentiation and survival of neuronal cells.
Collapse
Affiliation(s)
- Ylenia Bosio
- Molecular Biotechnology Center, Department of Genetics, Biology and Biochemistry, University of Turin, Italy
| | | | | | | | | | | | | |
Collapse
|
38
|
Hensel N, Ratzka A, Brinkmann H, Klimaschewski L, Grothe C, Claus P. Analysis of the fibroblast growth factor system reveals alterations in a mouse model of spinal muscular atrophy. PLoS One 2012; 7:e31202. [PMID: 22348054 PMCID: PMC3278439 DOI: 10.1371/journal.pone.0031202] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 01/04/2012] [Indexed: 12/02/2022] Open
Abstract
The monogenetic disease Spinal Muscular Atrophy (SMA) is characterized by a progressive loss of motoneurons leading to muscle weakness and atrophy due to severe reduction of the Survival of Motoneuron (SMN) protein. Several models of SMA show deficits in neurite outgrowth and maintenance of neuromuscular junction (NMJ) structure. Survival of motoneurons, axonal outgrowth and formation of NMJ is controlled by neurotrophic factors such as the Fibroblast Growth Factor (FGF) system. Besides their classical role as extracellular ligands, some FGFs exert also intracellular functions controlling neuronal differentiation. We have previously shown that intracellular FGF-2 binds to SMN and regulates the number of a subtype of nuclear bodies which are reduced in SMA patients. In the light of these findings, we systematically analyzed the FGF-system comprising five canonical receptors and 22 ligands in a severe mouse model of SMA. In this study, we demonstrate widespread alterations of the FGF-system in both muscle and spinal cord. Importantly, FGF-receptor 1 is upregulated in spinal cord at a pre-symptomatic stage as well as in a mouse motoneuron-like cell-line NSC34 based model of SMA. Consistent with that, phosphorylations of FGFR-downstream targets Akt and ERK are increased. Moreover, ERK hyper-phosphorylation is functionally linked to FGFR-1 as revealed by receptor inhibition experiments. Our study shows that the FGF system is dysregulated at an early stage in SMA and may contribute to the SMA pathogenesis.
Collapse
Affiliation(s)
- Niko Hensel
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Andreas Ratzka
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
| | - Hella Brinkmann
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
| | - Lars Klimaschewski
- Division of Neuroanatomy, Innsbruck Medical University, Innsbruck, Austria
| | - Claudia Grothe
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Peter Claus
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
- * E-mail:
| |
Collapse
|
39
|
Bricceno KV, Fischbeck KH, Burnett BG. Neurogenic and myogenic contributions to hereditary motor neuron disease. NEURODEGENER DIS 2012; 9:199-209. [PMID: 22327341 DOI: 10.1159/000335311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 11/23/2011] [Indexed: 12/21/2022] Open
Abstract
Spinal muscular atrophy and spinal and bulbar muscular atrophy are characterized by lower motor neuron loss and muscle atrophy. Although it is accepted that motor neuron loss is a primary event in disease pathogenesis, inherent defects in muscle may also contribute to the disease progression and severity. In this review, we discuss the relative contributions of primary pathological processes in the motor axons, neuromuscular junctions and muscle to disease manifestations. Characterizing these contributions helps us to better understand the disease mechanisms and to better target therapeutic intervention.
Collapse
Affiliation(s)
- Katherine V Bricceno
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | | |
Collapse
|
40
|
SMN deficiency attenuates migration of U87MG astroglioma cells through the activation of RhoA. Mol Cell Neurosci 2011; 49:282-9. [PMID: 22197680 DOI: 10.1016/j.mcn.2011.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 11/23/2011] [Accepted: 12/05/2011] [Indexed: 12/13/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease that affects alpha motoneurons in the spinal cord caused by homozygous deletion or specific mutations in the survival motoneuron-1 (SMN1) gene. Cell migration is critical at many stages of nervous system development; to investigate the role of SMN in cell migration, U87MG astroglioma cells were transduced with shSMN lentivectors and about 60% reduction in SMN expression was achieved. In a monolayer wound-healing assay, U87MG SMN-depleted cells exhibit reduced cell migration. In these cells, RhoA was activated and phosphorylated levels of myosin regulatory light chain (MLC), a substrate of the Rho kinase (ROCK), were found increased. The decrease in cell motility was related to activation of RhoA/Rho kinase (ROCK) signaling pathway as treatment with the ROCK inhibitor Y-27632 abrogated both the motility defects and MLC phosphorylation in SMN-depleted cells. As cell migration is regulated by continuous remodeling of the actin cytoskeleton, the actin distribution was studied in SMN-depleted cells. A shift from filamentous to monomeric (globular) actin, involving the disappearance of stress fibers, was observed. In addition, profilin I, an actin-sequestering protein showed an increased expression in SMN-depleted cells. SMN is known to physically interact with profilin, reducing its actin-sequestering activity. The present results suggest that in SMN-depleted cells, the increase in profilin I expression and the reduction in SMN inhibitory action on profilin could lead to reduced filamentous actin polymerization, thus decreasing cell motility. We propose that the alterations reported here in migratory activity in SMN-depleted cells, related to abnormal activation of RhoA/ROCK pathway and increased profilin I expression could have a role in developing nervous system by impairing normal neuron and glial cell migration and thus contributing to disease pathogenesis in SMA.
Collapse
|
41
|
Nölle A, Zeug A, van Bergeijk J, Tönges L, Gerhard R, Brinkmann H, Al Rayes S, Hensel N, Schill Y, Apkhazava D, Jablonka S, O'mer J, Srivastav RK, Baasner A, Lingor P, Wirth B, Ponimaskin E, Niedenthal R, Grothe C, Claus P. The spinal muscular atrophy disease protein SMN is linked to the Rho-kinase pathway via profilin. Hum Mol Genet 2011; 20:4865-78. [PMID: 21920940 DOI: 10.1093/hmg/ddr425] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Spinal muscular atrophy (SMA), a frequent neurodegenerative disease, is caused by reduced levels of functional survival of motoneuron (SMN) protein. SMN is involved in multiple pathways, including RNA metabolism and splicing as well as motoneuron development and function. Here we provide evidence for a major contribution of the Rho-kinase (ROCK) pathway in SMA pathogenesis. Using an in vivo protein interaction system based on SUMOylation of proteins, we found that SMN is directly interacting with profilin2a. Profilin2a binds to a stretch of proline residues in SMN, which is heavily impaired by a novel SMN2 missense mutation (S230L) derived from a SMA patient. In different SMA models, we identified differential phosphorylation of the ROCK-downstream targets cofilin, myosin-light chain phosphatase and profilin2a. We suggest that hyper-phosphorylation of profilin2a is the molecular link between SMN and the ROCK pathway repressing neurite outgrowth in neuronal cells. Finally, we found a neuron-specific increase in the F-/G-actin ratio that further support the role of actin dynamics in SMA pathogenesis.
Collapse
Affiliation(s)
- Anna Nölle
- Institute for Neuroanatomy, Hannover Medical School, Hannover 30623, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
The role of molecular microtubule motors and the microtubule cytoskeleton in stress granule dynamics. Int J Cell Biol 2011; 2011:939848. [PMID: 21760798 PMCID: PMC3132543 DOI: 10.1155/2011/939848] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 04/20/2011] [Indexed: 11/18/2022] Open
Abstract
Stress granules (SGs) are cytoplasmic foci that appear in cells exposed to stress-induced translational inhibition. SGs function as a triage center, where mRNAs are sorted for storage, degradation, and translation reinitiation. The underlying mechanisms of SGs dynamics are still being characterized, although many key players have been identified. The main components of SGs are stalled 48S preinitiation complexes. To date, many other proteins have also been found to localize in SGs and are hypothesized to function in SG dynamics. Most recently, the microtubule cytoskeleton and associated motor proteins have been demonstrated to function in SG dynamics. In this paper, we will discuss current literature examining the function of microtubules and the molecular microtubule motors in SG assembly, coalescence, movement, composition, organization, and disassembly.
Collapse
|
43
|
Anderton RS, Meloni BP, Mastaglia FL, Greene WK, Boulos S. Survival of motor neuron protein over-expression prevents calpain-mediated cleavage and activation of procaspase-3 in differentiated human SH-SY5Y cells. Neuroscience 2011; 181:226-33. [PMID: 21333717 DOI: 10.1016/j.neuroscience.2011.02.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 01/11/2011] [Accepted: 02/14/2011] [Indexed: 01/25/2023]
Abstract
Spinal muscular atrophy (SMA), a neurodegenerative disorder primarily affecting motor neurons, is the most common genetic cause of infant death. This incurable disease is caused by the absence of a functional SMN1 gene and a reduction in full length survival of motor neuron (SMN) protein. In this study, a neuroprotective function of SMN was investigated in differentiated human SH-SY5Y cells using an adenoviral vector to over-express SMN protein. The pro-survival capacity of SMN was assessed in an Akt/PI3-kinase inhibition (LY294002) model, as well as an oxidative stress (hydrogen peroxide) and excitotoxic (glutamate) model. SMN over-expression in SH-SY5Y cells protected against Akt/phosphatidylinositol 3-kinase (PI3-kinase) inhibition, but not oxidative stress, nor against excitotoxicity in rat cortical neurons. Western analysis of cell homogenates from SH-SY5Y cultures over-expressing SMN harvested pre- and post-Akt/PI3-kinase inhibition indicated that SMN protein inhibited caspase-3 activation via blockade of calpain-mediated procaspase-3 cleavage. This study has revealed a novel anti-apoptotic function for the SMN protein in differentiated SH-SY5Y cells. Finally, the cell death model described herein will allow the assessment of future therapeutic agents or strategies aimed at increasing SMN protein levels.
Collapse
Affiliation(s)
- R S Anderton
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia, WA, Australia.
| | | | | | | | | |
Collapse
|
44
|
Fuentes JL, Strayer MS, Matera AG. Molecular determinants of survival motor neuron (SMN) protein cleavage by the calcium-activated protease, calpain. PLoS One 2010; 5:e15769. [PMID: 21209906 PMCID: PMC3012718 DOI: 10.1371/journal.pone.0015769] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 11/28/2010] [Indexed: 01/13/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a leading genetic cause of childhood mortality, caused by reduced levels of survival motor neuron (SMN) protein. SMN functions as part of a large complex in the biogenesis of small nuclear ribonucleoproteins (snRNPs). It is not clear if defects in snRNP biogenesis cause SMA or if loss of some tissue-specific function causes disease. We recently demonstrated that the SMN complex localizes to the Z-discs of skeletal and cardiac muscle sarcomeres, and that SMN is a proteolytic target of calpain. Calpains are implicated in muscle and neurodegenerative disorders, although their relationship to SMA is unclear. Using mass spectrometry, we identified two adjacent calpain cleavage sites in SMN, S192 and F193. Deletion of small motifs in the region surrounding these sites inhibited cleavage. Patient-derived SMA mutations within SMN reduced calpain cleavage. SMN(D44V), reported to impair Gemin2 binding and amino-terminal SMN association, drastically inhibited cleavage, suggesting a role for these interactions in regulating calpain cleavage. Deletion of A188, a residue mutated in SMA type I (A188S), abrogated calpain cleavage, highlighting the importance of this region. Conversely, SMA mutations that interfere with self-oligomerization of SMN, Y272C and SMNΔ7, had no effect on cleavage. Removal of the recently-identified SMN degron (Δ268-294) resulted in increased calpain sensitivity, suggesting that the C-terminus of SMN is important in dictating availability of the cleavage site. Investigation into the spatial determinants of SMN cleavage revealed that endogenous calpains can cleave cytosolic, but not nuclear, SMN. Collectively, the results provide insight into a novel aspect of the post-translation regulation of SMN.
Collapse
Affiliation(s)
- Jennifer L. Fuentes
- Program in Molecular Biology and Biotechnology, Departments of Biology and Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Molly S. Strayer
- Program in Molecular Biology and Biotechnology, Departments of Biology and Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - A. Gregory Matera
- Program in Molecular Biology and Biotechnology, Departments of Biology and Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
45
|
Dominguez E, Marais T, Chatauret N, Benkhelifa-Ziyyat S, Duque S, Ravassard P, Carcenac R, Astord S, Pereira de Moura A, Voit T, Barkats M. Intravenous scAAV9 delivery of a codon-optimized SMN1 sequence rescues SMA mice. Hum Mol Genet 2010; 20:681-93. [PMID: 21118896 DOI: 10.1093/hmg/ddq514] [Citation(s) in RCA: 230] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Spinal muscular atrophy (SMA) is the most common genetic disease leading to infant mortality. This neuromuscular disorder is caused by the loss or mutation of the telomeric copy of the 'survival of motor neuron' (Smn) gene, termed SMN1. Loss of SMN1 leads to reduced SMN protein levels, inducing degeneration of motor neurons (MN) and progressive muscle weakness and atrophy. To date, SMA remains incurable due to the lack of a method to deliver therapeutically active molecules to the spinal cord. Gene therapy, consisting of reintroducing SMN1 in MNs, is an attractive approach for SMA. Here we used postnatal day 1 systemic injection of self-complementary adeno-associated virus (scAAV9) vectors carrying a codon-optimized SMN1 sequence and a chimeric intron placed downstream of the strong phosphoglycerate kinase (PGK) promoter (SMNopti) to overexpress the human SMN protein in a mouse model of severe SMA. Survival analysis showed that this treatment rescued 100% of the mice, increasing life expectancy from 27 to over 340 days (median survival of 199 days) in mice that normally survive about 13 days. The systemic scAAV9 therapy mediated complete correction of motor function, prevented MN death and rescued the weight loss phenotype close to normal. This study reports the most efficient rescue of SMA mice to date after a single intravenous injection of an optimized SMN-encoding scAAV9, highlighting the considerable potential of this method for the treatment of human SMA.
Collapse
Affiliation(s)
- Elisa Dominguez
- UPMC-AIM UMR S974, INSERM U 974, CNRS UMR 7215, Institut de Myologie, Université Pierre et Marie Curie Paris 6, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Boyer JG, Bowerman M, Kothary R. The many faces of SMN: deciphering the function critical to spinal muscular atrophy pathogenesis. FUTURE NEUROLOGY 2010. [DOI: 10.2217/fnl.10.57] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Spinal muscular atrophy (SMA) is the leading genetic cause of infant death, affecting 1 in 6000–10,000 live births. SMA is an autosomal recessive disorder characterized by the degeneration of α-motor neurons, and lower limb and proximal muscle weakness and wasting. SMA is the result of the deletion of or mutations in the survival motor neuron (SMN)1 gene. Currently, our understanding of how loss of the widely expressed SMN leads to the selective pathogenesis observed in SMA is limited. Here, we discuss the known nuclear and cytoplasmic functions of the SMN protein and how they relate to the SMA pathology reported in motor neurons, striated muscle and at neuromuscular junctions. While a vast amount of work in various cell and animal models has increased our knowledge of the many functions of the SMN protein, we have yet to come to a full understanding of which role(s) are central to SMA pathogenesis.
Collapse
Affiliation(s)
- Justin G Boyer
- Ottawa Hospital Research Institute, Regenerative Medicine Program, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, ON, Canada
| | - Mélissa Bowerman
- Ottawa Hospital Research Institute, Regenerative Medicine Program, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, ON, Canada
| | - Rashmi Kothary
- Department of Medicine, University of Ottawa, ON, Canada
| |
Collapse
|
47
|
SMN, profilin IIa and plastin 3: A link between the deregulation of actin dynamics and SMA pathogenesis. Mol Cell Neurosci 2009; 42:66-74. [DOI: 10.1016/j.mcn.2009.05.009] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Revised: 05/20/2009] [Accepted: 05/26/2009] [Indexed: 11/21/2022] Open
|
48
|
Fibroblast growth factor-2 regulates the stability of nuclear bodies. Proc Natl Acad Sci U S A 2009; 106:12747-52. [PMID: 19617559 DOI: 10.1073/pnas.0900122106] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Nuclear bodies are distinct subnuclear structures. The survival of motoneuron (SMN) gene is mutated or deleted in patients with the neurodegenerative disease spinal muscular atrophy (SMA). The gene product SMN is a marker protein for one class of nuclear bodies denoted as nuclear gems. SMN has also been found in Cajal bodies, which co-localize with gems in many cell types. Interestingly, SMA patients display a reduced number of gems. Little is known about the regulation of nuclear body formation and stabilization. We have previously shown that a nuclear isoform of the fibroblast growth factor-2 (FGF-2(23)) binds directly to SMN. In this study, we analyzed the consequences of FGF-2(23) binding to SMN with regard to nuclear body formation. On a molecular level, we showed that FGF-2(23) competed with Gemin2 (a component of the SMN complex that is necessary for gem stabilization) for binding to SMN. Down-regulation of Gemin2 by siRNA caused destabilization of SMN-positive nuclear bodies. This process is reflected in both cellular and in vivo systems by a negative regulatory function of FGF-2 in nuclear body formation: in HEK293 cells, FGF-2(23) decreased the number of SMN-positive nuclear bodies. The same effect could be observed in motoneurons of FGF-2 transgenic mice. This study demonstrates the functional role of a growth factor in the regulation of structural entities of the nucleus.
Collapse
|
49
|
Lunke S, El-Osta A. The emerging role of epigenetic modifications and chromatin remodeling in spinal muscular atrophy. J Neurochem 2009; 109:1557-69. [DOI: 10.1111/j.1471-4159.2009.06084.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
50
|
Heier CR, DiDonato CJ. Translational readthrough by the aminoglycoside geneticin (G418) modulates SMN stability in vitro and improves motor function in SMA mice in vivo. Hum Mol Genet 2009; 18:1310-22. [PMID: 19150990 DOI: 10.1093/hmg/ddp030] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Proximal spinal muscular atrophy (SMA) is a neuromuscular disorder for which there is no available therapy. SMA is caused by loss or mutation of the survival motor neuron 1 gene, SMN1, with retention of a nearly identical copy gene, SMN2. In contrast to SMN1, most SMN2 transcripts lack exon 7. This alternatively spliced transcript, Delta7-SMN, encodes a truncated protein that is rapidly degraded. Inhibiting this degradation may be of therapeutic value for the treatment of SMA. Recently aminoglycosides, which decrease translational fidelity to promote readthrough of termination codons, were shown to increase SMN levels in patient cell lines. Amid uncertainty concerning the role of SMN's C-terminus, the potential of translational readthrough as a therapeutic mechanism for SMA is unclear. Here, we used stable cell lines to demonstrate the SMN C-terminus modulates protein stability in a sequence-independent manner that is reproducible by translational readthrough. Geneticin (G418) was then identified as a potent inducer of the Delta7-SMN target sequence in vitro through a novel quantitative assay amenable to high throughput screens. Subsequent treatment of patient cell lines demonstrated that G418 increases SMN levels and is a potential lead compound. Furthermore, treatment of SMA mice with G418 increased both SMN protein and mouse motor function. Chronic administration, however, was associated with toxicity that may have prevented the detection of a survival benefit. Collectively, these results substantiate a sequence independent role of SMN's C-terminus in protein stability and provide the first in vivo evidence supporting translational readthrough as a therapeutic strategy for the treatment of SMA.
Collapse
Affiliation(s)
- Christopher R Heier
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | |
Collapse
|