1
|
Mohammadi S, Seyedalipour B, Hashemi SZ, Hosseinkhani S, Mohseni M. Implications of ALS-Associated Mutations on Biochemical and Biophysical Features of hSOD1 and Aggregation Formation. Biochem Genet 2024; 62:3658-3680. [PMID: 38196030 DOI: 10.1007/s10528-023-10619-y] [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: 07/20/2022] [Accepted: 12/02/2023] [Indexed: 01/11/2024]
Abstract
One of the recognized motor neuron degenerative disorders is amyotrophic lateral sclerosis (ALS). By now, several mutations have been reported and linked to ALS patients, some of which are induced by mutations in the human superoxide dismutase (hSOD1) gene. The ALS-provoking mutations are located throughout the structure of hSOD1 and promote the propensity to aggregate. Despite numerous investigations, the underlying mechanism related to the toxicity of mutant hSOD1 through the gain of a toxic function is still vague. We surveyed two mutant forms of hSOD1 by removing and adding cysteine at positions 146 and 72, respectively, to investigate the biochemical characterization and amyloid formation. Our findings predicted the harmful and destabilizing impact of two SOD1 mutants using multiple programs. The specific activity of the wild-type form was about 1.42- and 1.92-fold higher than that of C146R and G72C mutants, respectively. Comparative structural studies using CD spectropolarimetry, and intrinsic and ANS fluorescence showed alterations in secondary structure content, exposure of hydrophobic patches, and structural compactness of WT-hSOD1 vs. mutants. We demonstrated that two mutants were able to promote amyloid-like aggregates under amyloid induction circumstances (50-mM Tris-HCl pH 7.4, 0.2-M KSCN, 50-mM DTT, 37 °C, 190 rpm). Monitoring aggregates were done using an enhancement in thioflavin T fluorescence and alterations in Congo red absorption. The mutants accelerated fibrillation with subsequently greater fluorescence amplitude and a shorter lag time compared to WT-SOD1. These findings support the aggregation of ALS-associated SOD1 mutants as an integral part of ALS pathology.
Collapse
Affiliation(s)
- Saeede Mohammadi
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Bagher Seyedalipour
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran.
| | - Seyedeh Zohreh Hashemi
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojtaba Mohseni
- Department of Microbiology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| |
Collapse
|
2
|
Giannakou M, Akrani I, Tsoka A, Myrianthopoulos V, Mikros E, Vorgias C, Hatzinikolaou DG. Discovery of Novel Inhibitors against ALS-Related SOD1(A4V) Aggregation through the Screening of a Chemical Library Using Differential Scanning Fluorimetry (DSF). Pharmaceuticals (Basel) 2024; 17:1286. [PMID: 39458929 PMCID: PMC11510448 DOI: 10.3390/ph17101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Cu/Zn Superoxide Dismutase 1 (SOD1) is a 32 kDa cytosolic dimeric metalloenzyme that neutralizes superoxide anions into oxygen and hydrogen peroxide. Mutations in SOD1 are associated with ALS, a disease causing motor neuron atrophy and subsequent mortality. These mutations exert their harmful effects through a gain of function mechanism, rather than a loss of function. Despite extensive research, the mechanism causing selective motor neuron death still remains unclear. A defining feature of ALS pathogenesis is protein misfolding and aggregation, evidenced by ubiquitinated protein inclusions containing SOD1 in affected motor neurons. This work aims to identify compounds countering SOD1(A4V) misfolding and aggregation, which could potentially aid in ALS treatment. METHODS The approach employed was in vitro screening of a library comprising 1280 pharmacologically active compounds (LOPAC®) in the context of drug repurposing. Using differential scanning fluorimetry (DSF), these compounds were tested for their impact on SOD1(A4V) thermal stability. RESULTS AND CONCLUSIONS Dimer stability was the parameter chosen as the criterion for screening, since the dissociation of the native SOD1 dimer is the step prior to its in vitro aggregation. The screening revealed one compound raising protein-ligand Tm by 6 °C, eleven inducing a higher second Tm, suggesting a stabilization effect, and fourteen reducing Tm from 10 up to 26 °C, suggesting possible interactions or non-specific binding.
Collapse
Affiliation(s)
- Maria Giannakou
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
- Enzyme and Microbial Biotechnology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Ifigeneia Akrani
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Angeliki Tsoka
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Vassilios Myrianthopoulos
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Emmanuel Mikros
- Laboratory of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15784 Zografou, Greece; (I.A.)
| | - Constantinos Vorgias
- Biochemistry and Molecular Biology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| | - Dimitris G. Hatzinikolaou
- Enzyme and Microbial Biotechnology Unit, Department of Biology, National and Kapodistrian University of Athens, 15784 Zografou, Greece
| |
Collapse
|
3
|
Winroth I, Börjesson A, Andersen PM, Karlsson T. Cognitive deficits in ALS patients with SOD1 mutations. J Clin Exp Neuropsychol 2024; 46:669-682. [PMID: 39258714 DOI: 10.1080/13803395.2024.2393366] [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: 02/20/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024]
Abstract
OBJECTIVE Cognitive decline is common in patients with amyotrophic lateral sclerosis (ALS), especially in carriers of the mutation C9ORF72HRE. However, cognitive impairment is poorly understood in carriers of mutations in other genes causing ALS. We performed a comprehensive neuropsychological testing in patients with mutations in the SOD1 (mSOD1) gene. METHODS We examined 5 cognitive domains in 48 symptomatic patients with either hereditary or sporadic ALS. These were compared with 37 matched controls. RESULTS Carriers of SOD1-mutations and sporadic ALS had circumscribed deficits, but in a pattern different from C9ORF72HRE. All groups had deficits in working memory, although mSOD1-carriers significantly outperform sporadic ALS and C9ORF72HRE in an attention-driven visuospatial task involving copying a complex figure. Carriers of the D90A-SOD1 mutation overall performed as well as or better than carriers of other SOD1-mutations, except complex working memory. Bayesian analyses suggest (with evidence of moderate strength) that tasks involving the language domain did not differ between controls, mSOD1 and sporadic ALS. CONCLUSION Distinct cognitive impairments are prevalent in different ALS-syndromes and vary in patients with different pathogenic SOD1 mutations. The type and degree of impairment differed depending on genotype and was significantly least pronounced in patients homozygous for the D90A SOD1 mutation. The presence of cognitive deficits may influence optimal clinical management and intervention. We propose that cognitive assessment should be included in the routine examination of new patients suspected of ALS. Neuropsychological assessment is an under-recognized outcome parameter in clinical drug trials.
Collapse
Affiliation(s)
- Ivar Winroth
- Department of Clinical Sciences, Neuroscience, Umeå University, Umea, Sweden
| | | | - Peter M Andersen
- Department of Clinical Sciences, Neuroscience, Umeå University, Umea, Sweden
| | - Thomas Karlsson
- Department of Clinical Sciences, Neuroscience, Umeå University and Department of Behavioral Sciences and Learning, Linköping University, Umeå and Linköping, Sweden
| |
Collapse
|
4
|
Mielke JK, Klingeborn M, Schultz EP, Markham EL, Reese ED, Alam P, Mackenzie IR, Ly CV, Caughey B, Cashman NR, Leavens MJ. Seeding activity of human superoxide dismutase 1 aggregates in familial and sporadic amyotrophic lateral sclerosis postmortem neural tissues by real-time quaking-induced conversion. Acta Neuropathol 2024; 147:100. [PMID: 38884646 PMCID: PMC11182821 DOI: 10.1007/s00401-024-02752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disease with average lifespan of 2-5 years after diagnosis. The identification of novel prognostic and pharmacodynamic biomarkers are needed to facilitate therapeutic development. Metalloprotein human superoxide dismutase 1 (SOD1) is known to accumulate and form aggregates in patient neural tissue with familial ALS linked to mutations in their SOD1 gene. Aggregates of SOD1 have also been detected in other forms of ALS, including the sporadic form and the most common familial form linked to abnormal hexanucleotide repeat expansions in the Chromosome 9 open reading frame 72 (C9ORF72) gene. Here, we report the development of a real-time quaking-induced conversion (RT-QuIC) seed amplification assay using a recombinant human SOD1 substrate to measure SOD1 seeding activity in postmortem spinal cord and motor cortex tissue from persons with different ALS etiologies. Our SOD1 RT-QuIC assay detected SOD1 seeds in motor cortex and spinal cord dilutions down to 10-5. Importantly, we detected SOD1 seeding activity in specimens from both sporadic and familial ALS cases, with the latter having mutations in either their SOD1 or C9ORF72 genes. Analyses of RT-QuIC parameters indicated similar lag phases in spinal cords of sporadic and familial ALS patients, but higher ThT fluorescence maxima by SOD1 familial ALS specimens and sporadic ALS thoracic cord specimens. For a subset of sporadic ALS patients, motor cortex and spinal cords were examined, with seeding activity in both anatomical regions. Our results suggest SOD1 seeds are in ALS patient neural tissues not linked to SOD1 mutation, suggesting that SOD1 seeding activity may be a promising biomarker, particularly in sporadic ALS cases for whom genetic testing is uninformative.
Collapse
Affiliation(s)
- Justin K Mielke
- Department of Biomedical Sciences, McLaughlin Research Institute, 1520 23rd St. South, Great Falls, MT, 59405, USA
| | - Mikael Klingeborn
- Department of Biomedical Sciences, McLaughlin Research Institute, 1520 23rd St. South, Great Falls, MT, 59405, USA
| | - Eric P Schultz
- Center for Biomolecular Structure and Dynamics, University of Montana, 32 Campus Drive ISB #106, Missoula, MT, USA
| | - Erin L Markham
- Department of Biomedical Sciences, McLaughlin Research Institute, 1520 23rd St. South, Great Falls, MT, 59405, USA
| | - Emily D Reese
- Department of Biomedical Sciences, McLaughlin Research Institute, 1520 23rd St. South, Great Falls, MT, 59405, USA
| | - Parvez Alam
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 S. 4th St., Hamilton, MT, 59840, USA
| | - Ian R Mackenzie
- Departments of Pathology and Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Cindy V Ly
- Department of Neurology, Washington University, 660 S. Euclid Ave, Saint Louis, MO, 63110, USA
| | - Byron Caughey
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 S. 4th St., Hamilton, MT, 59840, USA
| | - Neil R Cashman
- Departments of Pathology and Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Moses J Leavens
- Department of Biomedical Sciences, McLaughlin Research Institute, 1520 23rd St. South, Great Falls, MT, 59405, USA.
| |
Collapse
|
5
|
Duan C, Kang M, Pan X, Gan Z, Huang V, Li G, Place RF, Li LC. Intrathecal administration of a novel siRNA modality extends survival and improves motor function in the SOD1 G93A ALS mouse model. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102147. [PMID: 38435120 PMCID: PMC10907209 DOI: 10.1016/j.omtn.2024.102147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 02/12/2024] [Indexed: 03/05/2024]
Abstract
Antisense oligonucleotides (ASOs) were the first modality to pioneer targeted gene knockdown in the treatment of amyotrophic lateral sclerosis (ALS) caused by mutant superoxide dismutase 1 (SOD1). RNA interference (RNAi) is another mechanism of gene silencing in which short interfering RNAs (siRNAs) effectively degrade complementary transcripts. However, delivery to extrahepatic tissues like the CNS has been a bottleneck in the clinical development of RNAi. Herein, we identify potent siRNA duplexes for the knockdown of human SOD1 in which medicinal chemistry and conjugation to an accessory oligonucleotide (ACO) enable activity in CNS tissues. Local delivery via intracerebroventricular or intrathecal injection into SOD1G93A mice delayed disease progression and extended animal survival with superior efficacy compared with an ASO resembling tofersen in sequence and chemistry. Treatment also prevented disease-related declines in motor function, including improvements in animal mobility, muscle strength, and coordination. The ACO itself does not target any specific complementary nucleic acid sequence; rather, it imparts benefits conducive to bioavailability and delivery through its chemistry. The complete conjugate (i.e., siRNA-ACO) represents a novel modality for delivery of duplex RNA (e.g., siRNA) to the CNS that is currently being tested in the clinic for treatment of ALS.
Collapse
Affiliation(s)
- Chunling Duan
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | - Moorim Kang
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | - Xiaojie Pan
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | - Zubao Gan
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | - Vera Huang
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | - Guanlin Li
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | | | - Long-Cheng Li
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| |
Collapse
|
6
|
Ivantsik O, John A, Kydonopoulou K, Mitropoulos K, Gerou S, Ali BR, Patrinos GP. Novel Pathogenic Variants Leading to Sporadic Amyotrophic Lateral Sclerosis in Greek Patients. Genes (Basel) 2024; 15:309. [PMID: 38540370 PMCID: PMC10970271 DOI: 10.3390/genes15030309] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 06/14/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive disease that affects motor neurons, leading to paralysis and death usually 3-5 years after the onset of symptoms. The investigation of both sporadic and familial ALS highlighted four main genes that contribute to the pathogenesis of the disease: SOD1, FUS, TARDBP and C9orf72. This study aims to provide a comprehensive investigation of genetic variants found in SOD1, FUS and TARDBP genes in Greek sporadic ALS (sALS) cases. Our sequencing analysis of the coding regions of the abovementioned genes that include the majority of the variants that lead to ALS in 32 sALS patients and 3 healthy relatives revealed 6 variants in SOD1, 19 variants in FUS and 37 variants in TARDBP, of which the SOD1 p.D90A and the FUS c.*356G>A (rs886051940) variants have been previously associated with ALS, while two novel nonsense pathogenic variants were also identified, namely FUS p.R241* and TDP-43 p.Y214*. Our study contributes to the worldwide effort toward clarifying the genetic basis of sALS to better understand the disease's molecular pathology.
Collapse
Affiliation(s)
- Ouliana Ivantsik
- Laboratory of Pharmacogenomics and Individualized Therapy, Division of Pharmacology and Biosciences, Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Rion, Greece
| | - Anne John
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates
| | | | - Konstantinos Mitropoulos
- Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, 10679 Athens, Greece
| | - Spyridon Gerou
- ANALYSI Biomedical Laboratories S.A., 54623 Thessaloniki, Greece
| | - Bassam R Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates
- ASPIRE Abu Dhabi Precision Medicine Ρesearch Institute, Al-Ain P.O. Box 15551, United Arab Emirates
| | - George P Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, Division of Pharmacology and Biosciences, Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Rion, Greece
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates
- ASPIRE Abu Dhabi Precision Medicine Ρesearch Institute, Al-Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 15551, United Arab Emirates
- Clinical Bioinformatics Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
7
|
Martinelli I, Ghezzi A, Zucchi E, Gianferrari G, Ferri L, Moglia C, Manera U, Solero L, Vasta R, Canosa A, Grassano M, Brunetti M, Mazzini L, De Marchi F, Simonini C, Fini N, Vinceti M, Pinti M, Chiò A, Calvo A, Mandrioli J. Predictors for progression in amyotrophic lateral sclerosis associated to SOD1 mutation: insight from two population-based registries. J Neurol 2023; 270:6081-6092. [PMID: 37668704 DOI: 10.1007/s00415-023-11963-0] [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: 06/07/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND Uncovering distinct features and trajectories of amyotrophic lateral sclerosis (ALS) associated with SOD1 mutations (SOD1-ALS) can provide valuable insights for patient' counseling and stratification for trials, and interventions timing. Our study aims to pinpoint distinct clinical characteristics of SOD1-ALS by delving into genotype-phenotype correlations and factors that potentially impact disease progression. METHODS This is a retrospective observational study of a SOD1-ALS cohort from two Italian registers situated in the regions of Emilia-Romagna, Piedmont and Valle d'Aosta. RESULTS Out of 2204 genotyped ALS patients, 2.5% carried SOD1 mutations, with a M:F ratio of 0.83. SOD1-ALS patients were younger, and more frequently reported a family history of ALS and/or FTD. SOD1-ALS had a longer survival compared to patients without ALS-associated gene mutations. However, here was considerable variability in survival across distinct SOD1 mutations, with an average survival of less than a year for the L39V, G42S, G73S, D91N mutations. Among SOD1-ALS, multivariate analysis showed that, alongside established clinical prognostic factors such as advanced age at onset and high progression rate at diagnosis, mutations located in exon 2 or within highly conserved gene positions predicted worse survival. Conversely, among comorbidities, cancer history was independently associated with longer survival. INTERPRETATION Within the context of an overall slower disease, SOD1-ALS exhibits some degree of heterogeneity linked to the considerable genetic diversity arising from the multitude of potential mutations sites and specific clinical prognostic factors, including cancer history. Revealing the factors that modulate the phenotypic heterogeneity of SOD1-ALS could prove advantageous in improving the efficacy of upcoming therapeutic approaches.
Collapse
Affiliation(s)
- Ilaria Martinelli
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
- Clinical and Experimental Medicine Ph.D. Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Ghezzi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Zucchi
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy.
- Neuroscience PhD Program, University of Modena and Reggio Emilia, Modena, Italy.
| | - Giulia Gianferrari
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Ferri
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
- Neuroscience PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Moglia
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
| | - Umberto Manera
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
| | - Luca Solero
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
| | - Rosario Vasta
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
| | - Antonio Canosa
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
- SC Neurologia 1U, AOU Città della Salute e della Scienza of Torino, Turin, Italy
| | - Maurizio Grassano
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
- SC Neurologia 1U, AOU Città della Salute e della Scienza of Torino, Turin, Italy
| | - Maura Brunetti
- SC Neurologia 1U, AOU Città della Salute e della Scienza of Torino, Turin, Italy
| | - Letizia Mazzini
- Neurology Unit, ALS Center, AOU Maggiore della Carità and University of Piemonte Orientale, Novara, Italy
| | - Fabiola De Marchi
- Neurology Unit, ALS Center, AOU Maggiore della Carità and University of Piemonte Orientale, Novara, Italy
| | - Cecilia Simonini
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
| | - Nicola Fini
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
| | - Marco Vinceti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Science of Public Health, Research Centre in Environmental, Genetic and Nutritional Epidemiology, University of Modena and Reggio Emilia, Modena, Italy
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Adriano Chiò
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
- SC Neurologia 1U, AOU Città della Salute e della Scienza of Torino, Turin, Italy
| | - Andrea Calvo
- Department of Neuroscience "Rita Levi Montalcini", ALS Centre, University of Torino, Turin, Italy
- SC Neurologia 1U, AOU Città della Salute e della Scienza of Torino, Turin, Italy
| | - Jessica Mandrioli
- Department of Neurosciences, Azienda Ospedaliero Universitaria di Modena, Viale Pietro Giardini, 1355, 41126, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
8
|
Zhao B, Jiang Q, Lin J, Wei Q, Li C, Hou Y, Cao B, Zhang L, Ou R, Liu K, Yang T, Xiao Y, Shang H. TBK1 variants in Chinese patients with amyotrophic lateral sclerosis: Genetic analysis and clinical features. Eur J Neurol 2023; 30:3079-3089. [PMID: 37422901 DOI: 10.1111/ene.15973] [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: 09/29/2022] [Revised: 03/09/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND AND PURPOSE Haploinsufficiency of TANK-binding kinase 1 (TBK1) loss-of-function (LoF) variants has been shown to be pathogenic in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). However, the genetic spectrum of TBK1 and clinical features of ALS patients with TBK1 variants remain largely unknown in Asians. METHODS Genetic analysis was performed on 2011 Chinese ALS patients. Software was used to predict the deleteriousness of missense variants in TBK1. In addition, PubMed, Embase and Web of Science were searched for related literature. RESULTS Twenty-six TBK1 variants were identified in 33 of 2011 ALS patients, including six novel LoF variants (0.3%) and 20 rare missense variants, 12 of which were predicted to be deleterious (0.6%). In addition to TBK1 variants, 11 patients had other ALS-related gene variants. Forty-two previous studies found that the frequency of TBK1 variants was 1.81% in ALS/FTD patients. The frequency of TBK1 LoF variants in ALS was 0.5% (Asians 0.4%; Caucasian 0.6%) and that of missense variants was 0.8% (Asians 1.0%; Caucasian 0.8%). ALS patients with TBK1 LoF variants affecting the kinase domain had a significantly younger age of onset than patients carrying LoF variants affecting the coiled coil domains CCD1 and CCD2. FTD has a frequency of 10% in Caucasian ALS patients with TBK1 LoF variants, which was not found in our cohort. CONCLUSION Our study expanded the genotypic spectrum of ALS patients with TBK1 variants and found that the clinical manifestations of TBK1 carriers are diverse.
Collapse
Affiliation(s)
- Bi Zhao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Qirui Jiang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Junyu Lin
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Qianqian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Yanbing Hou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Bei Cao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Lingyu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Kuncheng Liu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Tianmi Yang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Xiao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Korczowska-Łącka I, Słowikowski B, Piekut T, Hurła M, Banaszek N, Szymanowicz O, Jagodziński PP, Kozubski W, Permoda-Pachuta A, Dorszewska J. Disorders of Endogenous and Exogenous Antioxidants in Neurological Diseases. Antioxidants (Basel) 2023; 12:1811. [PMID: 37891890 PMCID: PMC10604347 DOI: 10.3390/antiox12101811] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
In diseases of the central nervous system, such as Alzheimer's disease (AD), Parkinson's disease (PD), stroke, amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and even epilepsy and migraine, oxidative stress load commonly surpasses endogenous antioxidative capacity. While oxidative processes have been robustly implicated in the pathogenesis of these diseases, the significance of particular antioxidants, both endogenous and especially exogenous, in maintaining redox homeostasis requires further research. Among endogenous antioxidants, enzymes such as catalase, superoxide dismutase, and glutathione peroxidase are central to disabling free radicals, thereby preventing oxidative damage to cellular lipids, proteins, and nucleic acids. Whether supplementation with endogenously occurring antioxidant compounds such as melatonin and glutathione carries any benefit, however, remains equivocal. Similarly, while the health benefits of certain exogenous antioxidants, including ascorbic acid (vitamin C), carotenoids, polyphenols, sulforaphanes, and anthocyanins are commonly touted, their clinical efficacy and effectiveness in particular neurological disease contexts need to be more robustly defined. Here, we review the current literature on the cellular mechanisms mitigating oxidative stress and comment on the possible benefit of the most common exogenous antioxidants in diseases such as AD, PD, ALS, HD, stroke, epilepsy, and migraine. We selected common neurological diseases of a basically neurodegenerative nature.
Collapse
Affiliation(s)
- Izabela Korczowska-Łącka
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Bartosz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Thomas Piekut
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Mikołaj Hurła
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Natalia Banaszek
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Oliwia Szymanowicz
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| | - Paweł P. Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (B.S.); (P.P.J.)
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Agnieszka Permoda-Pachuta
- Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, 20-059 Lublin, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 61-701 Poznan, Poland (M.H.)
| |
Collapse
|
10
|
Sim KS, Inoue T. Structure of a superoxide dismutase from a tardigrade: Ramazzottius varieornatus strain YOKOZUNA-1. Acta Crystallogr F Struct Biol Commun 2023; 79:169-179. [PMID: 37358501 PMCID: PMC10327573 DOI: 10.1107/s2053230x2300523x] [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: 04/02/2023] [Accepted: 06/12/2023] [Indexed: 06/27/2023] Open
Abstract
Superoxide dismutase (SOD) is an essential and ubiquitous antioxidant protein that is widely present in biological systems. The anhydrobiotic tardigrades are some of the toughest micro-animals. They have an expanded set of genes for antioxidant proteins such as SODs. These proteins are thought to play an essential role in oxidative stress resistance in critical situations such as desiccation, although their functions at the molecular level have yet to be explored. Here, crystal structures of a copper/zinc-containing SOD (RvSOD15) from an anhydrobiotic tardigrade, Ramazzottius varieornatus strain YOKOZUNA-1, are reported. In RvSOD15, one of the histidine ligands of the catalytic copper center is replaced by a valine (Val87). The crystal structures of the wild type and the V87H mutant show that even though a histidine is placed at position 87, a nearby flexible loop can destabilize the coordination of His87 to the Cu atom. Model structures of other RvSODs were investigated and it was found that some of them are also unusual SODs, with features such as deletion of the electrostatic loop or β3 sheet and unusual metal-binding residues. These studies show that RvSOD15 and some other RvSODs may have evolved to lose the SOD function, suggesting that gene duplications of antioxidant proteins do not solely explain the high stress tolerance of anhydrobiotic tardigrades.
Collapse
Affiliation(s)
- Kee-Shin Sim
- Graduate School of Pharmaceutical Science, Osaka University, Suita City, Osaka 565-0871, Japan
| | - Tsuyoshi Inoue
- Graduate School of Pharmaceutical Science, Osaka University, Suita City, Osaka 565-0871, Japan
| |
Collapse
|
11
|
Forsberg KM, Graffmo KS, Stenvall E, Tabikh N, Marklund SL, Brännström T, Andersen PM. Widespread CNS pathology in amyotrophic lateral sclerosis homozygous for the D90A SOD1 mutation. Acta Neuropathol 2023; 145:13-28. [PMID: 36385230 PMCID: PMC9807479 DOI: 10.1007/s00401-022-02519-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Mutations in the gene encoding the ubiquitously expressed free radical scavenging enzyme superoxide dismutase-1 (SOD1) are found in 2-6% of amyotrophic lateral sclerosis patients. The most frequent SOD1 mutation worldwide is D90A. Amyotrophic lateral sclerosis caused by this mutation has some unusual features: the heredity is usually recessive, the phenotype is stereotypic with slowly evolving motor symptoms beginning in the legs and may also include sensory, autonomic, and urinary bladder involvement. Furthermore, the mutant protein resembles the wild type, with normal content and enzymatic activity in the central nervous system. Here, we report neuropathological findings in nine patients homozygous for the D90A mutation. All nine had numerous small granular inclusions immunoreactive for misfolded SOD1 in motor neurons and glial nuclei in the spinal cord and brainstem. In addition to degeneration of the corticospinal tracts, all patients had degeneration of the dorsal columns. We also found intense gliosis in circumscribed cortical areas of the frontal and temporal lobes and in the insula. In these areas and in adjacent white matter, there were SOD1 staining neuropil threads. A few SOD1-immunopositive cytoplasmic neuronal inclusions were observed in cortical areas, as were glial nuclear inclusions. As suggested by the symptoms and signs and earlier neurophysiological and imaging investigations, the histopathology in patients homozygous for the D90A SOD1 extends beyond the motor system to include cognitive and sensory cortical areas. However, even in the patients that had a symptomatic disease duration of more than 2 or 3 decades and lived into their 70s or 80s, there were no SOD1-inclusion pathology and no typical dysfunction (apart from the musculature) in non-nervous organs. Thus, only specific parts of the CNS seem to be vulnerable to toxicity provoked by homozygously expressed mutant SOD1.
Collapse
Affiliation(s)
- Karin M Forsberg
- Department of Clinical Sciences, Neurosciences, Umeå University, 90185, Umeå, Sweden.,Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Karin S Graffmo
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Erica Stenvall
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Naima Tabikh
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Stefan L Marklund
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Thomas Brännström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Peter M Andersen
- Department of Clinical Sciences, Neurosciences, Umeå University, 90185, Umeå, Sweden.
| |
Collapse
|
12
|
Acosta-Uribe J, Aguillón D, Cochran JN, Giraldo M, Madrigal L, Killingsworth BW, Singhal R, Labib S, Alzate D, Velilla L, Moreno S, García GP, Saldarriaga A, Piedrahita F, Hincapié L, López HE, Perumal N, Morelo L, Vallejo D, Solano JM, Reiman EM, Surace EI, Itzcovich T, Allegri R, Sánchez-Valle R, Villegas-Lanau A, White CL, Matallana D, Myers RM, Browning SR, Lopera F, Kosik KS. A neurodegenerative disease landscape of rare mutations in Colombia due to founder effects. Genome Med 2022; 14:27. [PMID: 35260199 PMCID: PMC8902761 DOI: 10.1186/s13073-022-01035-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 02/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Colombian population, as well as those in other Latin American regions, arose from a recent tri-continental admixture among Native Americans, Spanish invaders, and enslaved Africans, all of whom passed through a population bottleneck due to widespread infectious diseases that left small isolated local settlements. As a result, the current population reflects multiple founder effects derived from diverse ancestries. METHODS We characterized the role of admixture and founder effects on the origination of the mutational landscape that led to neurodegenerative disorders under these historical circumstances. Genomes from 900 Colombian individuals with Alzheimer's disease (AD) [n = 376], frontotemporal lobar degeneration-motor neuron disease continuum (FTLD-MND) [n = 197], early-onset dementia not otherwise specified (EOD) [n = 73], and healthy participants [n = 254] were analyzed. We examined their global and local ancestry proportions and screened this cohort for deleterious variants in disease-causing and risk-conferring genes. RESULTS We identified 21 pathogenic variants in AD-FTLD related genes, and PSEN1 harbored the majority (11 pathogenic variants). Variants were identified from all three continental ancestries. TREM2 heterozygous and homozygous variants were the most common among AD risk genes (102 carriers), a point of interest because the disease risk conferred by these variants differed according to ancestry. Several gene variants that have a known association with MND in European populations had FTLD phenotypes on a Native American haplotype. Consistent with founder effects, identity by descent among carriers of the same variant was frequent. CONCLUSIONS Colombian demography with multiple mini-bottlenecks probably enhanced the detection of founder events and left a proportionally higher frequency of rare variants derived from the ancestral populations. These findings demonstrate the role of genomically defined ancestry in phenotypic disease expression, a phenotypic range of different rare mutations in the same gene, and further emphasize the importance of inclusiveness in genetic studies.
Collapse
Affiliation(s)
- Juliana Acosta-Uribe
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, CA, USA
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - David Aguillón
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | | | - Margarita Giraldo
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
- Instituto Neurológico de Colombia (INDEC), Medellín, Colombia
| | - Lucía Madrigal
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Bradley W Killingsworth
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Rijul Singhal
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Sarah Labib
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Diana Alzate
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Lina Velilla
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Sonia Moreno
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Gloria P García
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Amanda Saldarriaga
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Francisco Piedrahita
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Liliana Hincapié
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Hugo E López
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Nithesh Perumal
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Leonilde Morelo
- Department of Internal Medicine, School of Medicine, Universidad del Sinú, Montería, Colombia
| | - Dionis Vallejo
- Department of Neurology, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Juan Marcos Solano
- Department of Neurology, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | | | - Ezequiel I Surace
- Laboratorio de Enfermedades Neurodegenerativas (Fleni-CONICET), Buenos Aires, Argentina
| | - Tatiana Itzcovich
- Laboratorio de Enfermedades Neurodegenerativas (Fleni-CONICET), Buenos Aires, Argentina
| | - Ricardo Allegri
- Centro de Memoria y Envejecimiento (Fleni-CONICET), Buenos Aires, Argentina
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clínic de Barcelona, IDIBAPS and University of Barcelona, Barcelona, Spain
| | - Andrés Villegas-Lanau
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Charles L White
- Neuropathology Section, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Diana Matallana
- Instituto de Envejecimiento, Department of Psychiatry, School of Medicine, Pontifical Xaverian University, Bogotá, Colombia
- Department of Mental Health, Hospital Universitario Santa Fe de Bogotá, Bogotá, Colombia
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Sharon R Browning
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, School of Medicine, Universidad de Antioquia, Medellín, Colombia.
| | - Kenneth S Kosik
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, CA, USA.
| |
Collapse
|
13
|
Müller K, Oh KW, Nordin A, Panthi S, Kim SH, Nordin F, Freischmidt A, Ludolph AC, Ki CS, Forsberg K, Weishaupt J, Kim YE, Andersen PM. De novo mutations in SOD1 are a cause of ALS. J Neurol Neurosurg Psychiatry 2022; 93:201-206. [PMID: 34518333 PMCID: PMC8784989 DOI: 10.1136/jnnp-2021-327520] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/05/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The only identified cause of amyotrophic lateral sclerosis (ALS) are mutations in a number of genes found in familial cases but also in sporadic cases. De novo mutations occurring in a parental gonadal cell, in the zygote or postzygotic during embryonal development can result in an apparently sporadic/isolated case of ALS later in life. We searched for de novo mutations in SOD1 as a cause of ALS. METHODS We analysed peripheral-blood exome, genome and Sanger sequencing to identify deleterious mutations in SOD1 in 4000 ALS patients from Germany, South Korea and Sweden. Parental kinship was confirmed using highly polymorphic microsatellite markers across the genome. Medical genealogical and clinical data were reviewed and compared with the literature. RESULTS We identified four sporadic ALS cases with de novo mutations in SOD1. They aggregate in hot-spot codons earlier found mutated in familial cases. Their phenotypes match closely what has earlier been reported in familial cases with pathogenic mutations in SOD1. We also encountered familial cases where de novo mutational events in recent generations may have been involved. CONCLUSIONS De novo mutations are a cause of sporadic ALS and may also be underpinning smaller families with few affected ALS cases. It was not possible to ascertain if the origin of the de novo mutations was parental germline, zygotic or postzygotic during embryonal development. All ALS patients should be offered genetic counselling and genetic screening, the challenges of variant interpretation do not outweigh the potential benefits including earlier confirmed diagnosis and possible bespoken therapy.
Collapse
Affiliation(s)
| | - Ki-Wook Oh
- Department of Neurology, Hanyang University Seoul Hospital, Seongdong-gu, Seoul, Republic of Korea
- Cell Therapy Center, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Angelica Nordin
- Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Sudhan Panthi
- Department of Neurology, Ulm University, Ulm, Germany
| | - Seung Hyun Kim
- Department of Neurology, Hanyang University Seoul Hospital, Seongdong-gu, Seoul, Republic of Korea
- Cell Therapy Center, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Frida Nordin
- Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | | | | | - Chang Seok Ki
- Genome Research Centre, GC Genome, Yongin, Republic of Korea
| | - Karin Forsberg
- Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
- Medical Biosciences, Umeå University, Umeå, Sweden
| | - Jochen Weishaupt
- Department for Neurodegeneration, Universitätsmedizin Mannheim, Mannheim, Germany
| | - Young-Eun Kim
- Department of Laboratory Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | | |
Collapse
|
14
|
Genetic architecture of motor neuron diseases. J Neurol Sci 2021; 434:120099. [PMID: 34965490 DOI: 10.1016/j.jns.2021.120099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/26/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022]
Abstract
Motor neuron diseases (MNDs) are rare and frequently fatal neurological disorders in which motor neurons within the brainstem and spinal cord regions slowly die. MNDs are primarily caused by genetic mutations, and > 100 different mutant genes in humans have been discovered thus far. Given the fact that many more MND-related genes have yet to be discovered, the growing body of genetic evidence has offered new insights into the diverse cellular and molecular mechanisms involved in the aetiology and pathogenesis of MNDs. This search may aid in the selection of potential candidate genes for future investigation and, eventually, may open the door to novel interventions to slow down disease progression. In this review paper, we have summarized detailed existing research findings of different MNDs, such as amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), spinal bulbar muscle atrophy (SBMA) and hereditary spastic paraplegia (HSP) in relation to their complex genetic architecture.
Collapse
|
15
|
Wen X, Zhu W, Xia NL, Li Q, Di L, Zhang S, Chen H, Lu Y, Wang M, Xu M, Wang S, Shen XM, Lu J, Da Y. Missense Mutations of Codon 116 in the SOD1 Gene Cause Rapid Progressive Familial ALS and Predict Short Viability With PMA Phenotype. Front Genet 2021; 12:776831. [PMID: 34868265 PMCID: PMC8638083 DOI: 10.3389/fgene.2021.776831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/14/2021] [Indexed: 11/25/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common form of motor neuron disease, characterized by a great variety of both clinical presentations and genetic causes. Previous studies had identified two different missense mutations in SOD1 (p.R116C and p.R116G) causing familial ALS. In this study, we report a novel heterozygous missense mutation in the SOD1 gene (p.R116S) in a family with inherited ALS manifested as fast-deteriorating pure lower motor neuron symptoms. The patient displayed similar clinical picture and prognostic value to previous reported cases with different R116 substitution mutations. Modeling of all R116 substitutions in the resolved SOD1 protein structure revealed a shared mechanism with destroyed hydrogen bonds between R116 and other two residues, which might lead to protein unfolding and oligomer formation, ultimately conferring neurotoxicity.
Collapse
Affiliation(s)
- Xinmei Wen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenjia Zhu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Nan L Xia
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qianwen Li
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Li Di
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shu Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hai Chen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yan Lu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Min Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Min Xu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Suobin Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xin-Ming Shen
- Department of Neurology and Neuromuscular Research Laboratory, Mayo Clinic, Rochester, MN, United States
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Yuwei Da
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Ricci C, Giannini F, Riolo G, Bocci S, Casali S, Battistini S. A Novel Variant in Superoxide Dismutase 1 Gene ( p.V119M) in Als Patients with Pure Lower Motor Neuron Presentation. Genes (Basel) 2021; 12:genes12101544. [PMID: 34680939 PMCID: PMC8535540 DOI: 10.3390/genes12101544] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 11/30/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and fatal disorder characterized by degeneration of motor neurons in the cerebral cortex, brain stem, and spinal cord. Most cases of ALS appear sporadically, but 5–10% of patients have a family history of disease. Mutations in the superoxide dismutase 1 gene (SOD1) have been found in 12–23% of familial cases and in 1–2% of sporadic cases. Currently, more than 180 different SOD1 gene variants have been identified in ALS patients. Here, we describe two apparently sporadic ALS patients carrying the same SOD1 c.355G>A variant, leading to the p.V119M substitution, not previously described. Both the patients showed pure lower motor neuron phenotype. The former presented with the flail leg syndrome, a rare ALS variant, characterized by progressive distal onset weakness and atrophy of lower limbs, slow progression and better survival than typical ALS. The latter exhibited rapidly progressive weakness of upper and lower limbs, neither upper motor neuron nor bulbar involvement, and shorter survival than typical ALS. We provide an accurate description of the phenotype, and a bioinformatics analysis of the p.V119M variant on protein structure. This study may increase the knowledge about genotype-phenotype correlations in ALS and improve the approach to ALS patients.
Collapse
|
17
|
Bourbouli M, Paraskevas GP, Rentzos M, Mathioudakis L, Zouvelou V, Bougea A, Tychalas A, Kimiskidis VK, Constantinides V, Zafeiris S, Tzagournissakis M, Papadimas G, Karadima G, Koutsis G, Kroupis C, Kartanou C, Kapaki E, Zaganas I. Genotyping and Plasma/Cerebrospinal Fluid Profiling of a Cohort of Frontotemporal Dementia-Amyotrophic Lateral Sclerosis Patients. Brain Sci 2021; 11:brainsci11091239. [PMID: 34573259 PMCID: PMC8472580 DOI: 10.3390/brainsci11091239] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 09/05/2021] [Accepted: 09/14/2021] [Indexed: 11/16/2022] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are part of the same pathophysiological spectrum and have common genetic and cerebrospinal fluid (CSF) biomarkers. Our aim here was to identify causative gene variants in a cohort of Greek patients with FTD, ALS and FTD-ALS, to measure levels of CSF biomarkers and to investigate genotype-phenotype/CSF biomarker associations. In this cohort of 130 patients (56 FTD, 58 ALS and 16 FTD-ALS), we performed C9orf72 hexanucleotide repeat expansion analysis, whole exome sequencing and measurement of “classical” (Aβ42, total tau and phospho-tau) and novel (TDP-43) CSF biomarkers and plasma progranulin. Through these analyses, we identified 14 patients with C9orf72 repeat expansion and 11 patients with causative variants in other genes (three in TARDBP, three in GRN, three in VCP, one in FUS, one in SOD1). In ALS patients, we found that levels of phospho-tau were lower in C9orf72 repeat expansion and MAPT c.855C>T (p.Asp285Asp) carriers compared to non-carriers. Additionally, carriers of rare C9orf72 and APP variants had lower levels of total tau and Aβ42, respectively. Plasma progranulin levels were decreased in patients carrying GRN pathogenic variants. This study expands the genotypic and phenotypic spectrum of FTD/ALS and offers insights in possible genotypic/CSF biomarker associations.
Collapse
Affiliation(s)
- Mara Bourbouli
- Neurogenetics Laboratory, Neurology Department, Medical School, University of Crete, 71003 Heraklion, Greece; (M.B.); (L.M.); (S.Z.); (M.T.)
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
| | - George P. Paraskevas
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Attikon University General Hospital, 12462 Athens, Greece
| | - Mihail Rentzos
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
| | - Lambros Mathioudakis
- Neurogenetics Laboratory, Neurology Department, Medical School, University of Crete, 71003 Heraklion, Greece; (M.B.); (L.M.); (S.Z.); (M.T.)
| | - Vasiliki Zouvelou
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
| | - Anastasia Bougea
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
| | - Athanasios Tychalas
- Department of Neurology, Papageorgiou General Hospital, 56403 Thessaloniki, Greece;
| | - Vasilios K. Kimiskidis
- 1st Department of Neurology, AHEPA Hospital, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece;
| | - Vasilios Constantinides
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
| | - Spiros Zafeiris
- Neurogenetics Laboratory, Neurology Department, Medical School, University of Crete, 71003 Heraklion, Greece; (M.B.); (L.M.); (S.Z.); (M.T.)
| | - Minas Tzagournissakis
- Neurogenetics Laboratory, Neurology Department, Medical School, University of Crete, 71003 Heraklion, Greece; (M.B.); (L.M.); (S.Z.); (M.T.)
| | - Georgios Papadimas
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
| | - Georgia Karadima
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
| | - Georgios Koutsis
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
| | - Christos Kroupis
- Department of Clinical Biochemistry, Attikon University General Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Chrisoula Kartanou
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
| | - Elisabeth Kapaki
- 1st Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, Eginition Hospital, 11528 Athens, Greece; (G.P.P.); (M.R.); (V.Z.); (A.B.); (V.C.); (G.P.); (G.K.); (G.K.); (C.K.); (E.K.)
| | - Ioannis Zaganas
- Neurogenetics Laboratory, Neurology Department, Medical School, University of Crete, 71003 Heraklion, Greece; (M.B.); (L.M.); (S.Z.); (M.T.)
- Correspondence: ; Tel.: +30-2810-394643
| |
Collapse
|
18
|
Muratet F, Teyssou E, Chiot A, Boillée S, Lobsiger CS, Bohl D, Gyorgy B, Guegan J, Marie Y, Amador MDM, Salachas F, Meininger V, Bernard E, Antoine JC, Camdessanché JP, Camu W, Cazeneuve C, Fauret-Amsellem AL, Leguern E, Mouzat K, Guissart C, Lumbroso S, Corcia P, Vourc'h P, Grapperon AM, Attarian S, Verschueren A, Seilhean D, Millecamps S. Impact of a frequent nearsplice SOD1 variant in amyotrophic lateral sclerosis: optimising SOD1 genetic screening for gene therapy opportunities. J Neurol Neurosurg Psychiatry 2021; 92:942-949. [PMID: 33785574 DOI: 10.1136/jnnp-2020-325921] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/22/2021] [Accepted: 03/07/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Mutations in superoxide dismutase 1 gene (SOD1), encoding copper/zinc superoxide dismutase protein, are the second most frequent high penetrant genetic cause for amyotrophic lateral sclerosis (ALS) motor neuron disease in populations of European descent. More than 200 missense variants are reported along the SOD1 protein. To limit the production of these aberrant and deleterious SOD1 species, antisense oligonucleotide approaches have recently emerged and showed promising effects in clinical trials. To offer the possibility to any patient with SOD1-ALS to benefit of such a gene therapy, it is necessary to ascertain whether any variant of unknown significance (VUS), detected for example in SOD1 non-coding sequences, is pathogenic. METHODS We analysed SOD1 mutation distribution after SOD1 sequencing in a large cohort of 470 French familial ALS (fALS) index cases. RESULTS We identified a total of 27 SOD1 variants in 38 families including two SOD1 variants located in nearsplice or intronic regions of the gene. The pathogenicity of the c.358-10T>G nearsplice SOD1 variant was corroborated based on its high frequency (as the second most frequent SOD1 variant) in French fALS, the segregation analysis confirmed in eight affected members of a large pedigree, the typical SOD1-related phenotype observed (with lower limb onset and prominent lower motor neuron involvement), and findings on postmortem tissues showing SOD1 misaccumulation. CONCLUSIONS Our results highlighted nearsplice/intronic mutations in SOD1 are responsible for a significant portion of French fALS and suggested the systematic analysis of the SOD1 mRNA sequence could become the method of choice for SOD1 screening, not to miss these specific cases.
Collapse
Affiliation(s)
- François Muratet
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France
| | - Elisa Teyssou
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France
| | - Aude Chiot
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France
| | - Séverine Boillée
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France
| | - Christian S Lobsiger
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France
| | - Delphine Bohl
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France
| | - Beata Gyorgy
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France
| | - Justine Guegan
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France
| | - Yannick Marie
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France
| | - Maria Del Mar Amador
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France.,AP-HP, Département de Neurologie, Centre de référence SLA Ile de France, Hôpital de la Pitié-Salpêtrière, Paris, Île de France, France
| | - Francois Salachas
- AP-HP, Département de Neurologie, Centre de référence SLA Ile de France, Hôpital de la Pitié-Salpêtrière, Paris, Île de France, France
| | - Vincent Meininger
- Hôpital des Peupliers, Ramsay General Health Group, Paris, Île-de-France, France
| | - Emilien Bernard
- Centre de référence SLA, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Université de Lyon, Bron, Auvergne-Rhône-Alpes, France.,Institut NeuroMyoGène, CNRS UMR5310, INSERM U1217, Faculté de Médecine Rockefeller, Université Claude Bernard Lyon I, Lyon, Auvergne-Rhône-Alpes, France
| | - Jean-Christophe Antoine
- Service de Neurologie, Centre de Ressource et de Compétence SLA, Hôpital Nord, CHU de Saint-Etienne, Saint-Etienne, Rhône-Alpes, France
| | - Jean-Philippe Camdessanché
- Service de Neurologie, Centre de Ressource et de Compétence SLA, Hôpital Nord, CHU de Saint-Etienne, Saint-Etienne, Rhône-Alpes, France
| | - William Camu
- Centre de référence SLA, Hôpital Gui de Chauliac, CHU de Montpellier, Université de Montpellier, Montpellier, Languedoc-Roussillon, France
| | - Cécile Cazeneuve
- Département de Génétique et Cytogénétique, Unité Fonctionnelle de neurogénétique moléculaire et cellulaire, APHP, Hôpital Pitié-Salpêtrière, Paris, Île-de-France, France
| | - Anne-Laure Fauret-Amsellem
- Département de Génétique et Cytogénétique, Unité Fonctionnelle de neurogénétique moléculaire et cellulaire, APHP, Hôpital Pitié-Salpêtrière, Paris, Île-de-France, France
| | - Eric Leguern
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France.,Département de Génétique et Cytogénétique, Unité Fonctionnelle de neurogénétique moléculaire et cellulaire, APHP, Hôpital Pitié-Salpêtrière, Paris, Île-de-France, France
| | - Kevin Mouzat
- Laboratoire de Biochimie et Biologie Moleculaire, CHU Nimes, Nîmes, Languedoc-Roussillon, France.,Motoneuron Disease: Pathophysiology and Therapy, INM, INSERM, Université de Montpellier, Montpellier, Languedoc-Roussillon, France
| | - Claire Guissart
- Laboratoire de Biochimie et Biologie Moleculaire, CHU Nimes, Nîmes, Languedoc-Roussillon, France.,Motoneuron Disease: Pathophysiology and Therapy, INM, INSERM, Université de Montpellier, Montpellier, Languedoc-Roussillon, France
| | - Serge Lumbroso
- Laboratoire de Biochimie et Biologie Moleculaire, CHU Nimes, Nîmes, Languedoc-Roussillon, France.,Motoneuron Disease: Pathophysiology and Therapy, INM, INSERM, Université de Montpellier, Montpellier, Languedoc-Roussillon, France
| | - Philippe Corcia
- Centre de référence SLA, Département de Neurologie, CHRU Tours, Tours, Centre-Val de Loire, France.,UMR 1253, Université de Tours, Inserm, Tours, Centre-Val de Loire, France
| | - Patrick Vourc'h
- UMR 1253, Université de Tours, Inserm, Tours, Centre-Val de Loire, France.,Service de Biochimie et Biologie Moléculaire, CHU Tours, Tours, Centre-Val de Loire, France
| | - Aude-Marie Grapperon
- Centre de Référence pour les Maladies Neuromusculaire et la SLA, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, CHU de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Shahram Attarian
- Centre de Référence pour les Maladies Neuromusculaire et la SLA, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, CHU de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Annie Verschueren
- Centre de Référence pour les Maladies Neuromusculaire et la SLA, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, CHU de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Danielle Seilhean
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France.,Département de Neuropathologie, APHP, Hôpital Pitié-Salpêtrière, Paris, Île-de-France, France
| | - Stéphanie Millecamps
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM,Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, Île de France, France
| |
Collapse
|
19
|
Post J, Schaffrath A, Gering I, Hartwig S, Lehr S, Shah NJ, Langen KJ, Willbold D, Kutzsche J, Willuweit A. Oral Treatment with RD2RD2 Impedes Development of Motoric Phenotype and Delays Symptom Onset in SOD1 G93A Transgenic Mice. Int J Mol Sci 2021; 22:ijms22137066. [PMID: 34209129 PMCID: PMC8269060 DOI: 10.3390/ijms22137066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 11/16/2022] Open
Abstract
Neuroinflammation is a pathological hallmark of several neurodegenerative disorders and plays a key role in the pathogenesis of amyotrophic lateral sclerosis (ALS). It has been implicated as driver of disease progression and is observed in ALS patients, as well as in the transgenic SOD1G93A mouse model. Here, we explore and validate the therapeutic potential of the d-enantiomeric peptide RD2RD2 upon oral administration in SOD1G93A mice. Transgenic mice were treated daily with RD2RD2 or placebo for 10 weeks and phenotype progression was followed with several behavioural tests. At the end of the study, plasma cytokine levels and glia cell markers in brain and spinal cord were analysed. Treatment resulted in a significantly increased performance in behavioural and motor coordination tests and a decelerated neurodegenerative phenotype in RD2RD2-treated SOD1G93A mice. Additionally, we observed retardation of the average disease onset. Treatment of SOD1G93A mice led to significant reduction in glial cell activation and a rescue of neurons. Analysis of plasma revealed normalisation of several cytokines in samples of RD2RD2-treated SOD1G93A mice towards the levels of non-transgenic mice. In conclusion, these findings qualify RD2RD2 to be considered for further development and testing towards a disease modifying ALS treatment.
Collapse
Affiliation(s)
- Julia Post
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany; (J.P.); (A.S.); (I.G.)
| | - Anja Schaffrath
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany; (J.P.); (A.S.); (I.G.)
| | - Ian Gering
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany; (J.P.); (A.S.); (I.G.)
| | - Sonja Hartwig
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; (S.H.); (S.L.)
- German Center for Diabetes Research, Partner Düsseldorf, 85764 München-Neuherberg, Germany
| | - Stefan Lehr
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; (S.H.); (S.L.)
- German Center for Diabetes Research, Partner Düsseldorf, 85764 München-Neuherberg, Germany
| | - N. Jon Shah
- Institute of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Forschungszentrum Jülich, 52425 Jülich, Germany; (N.J.S.); (K.-J.L.)
- Institute of Neuroscience and Medicine 11, INM-11, JARA, Forschungszentrum Jülich, 52425 Jülich, Germany
- JARA-Brain-Translational Medicine, 52062 Aachen, Germany
- Department of Neurology, RWTH Aachen University, 52062 Aachen, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Forschungszentrum Jülich, 52425 Jülich, Germany; (N.J.S.); (K.-J.L.)
- Department of Nuclear Medicine, RWTH Aachen University, 52062 Aachen, Germany
| | - Dieter Willbold
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany; (J.P.); (A.S.); (I.G.)
- Institut für Physikalische Biologie, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
- Correspondence: (D.W.); (J.K.); (A.W.); Tel.: +49-2461-612100 (D.W.); +49-2461-619496 (J.K.); +49-2461-6196358 (A.W.); Fax: +49-2461-612023 (D.W.); +49-2461-619497 (J.K.); +49-2461-612302 (A.W.)
| | - Janine Kutzsche
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany; (J.P.); (A.S.); (I.G.)
- Correspondence: (D.W.); (J.K.); (A.W.); Tel.: +49-2461-612100 (D.W.); +49-2461-619496 (J.K.); +49-2461-6196358 (A.W.); Fax: +49-2461-612023 (D.W.); +49-2461-619497 (J.K.); +49-2461-612302 (A.W.)
| | - Antje Willuweit
- Institute of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Forschungszentrum Jülich, 52425 Jülich, Germany; (N.J.S.); (K.-J.L.)
- Correspondence: (D.W.); (J.K.); (A.W.); Tel.: +49-2461-612100 (D.W.); +49-2461-619496 (J.K.); +49-2461-6196358 (A.W.); Fax: +49-2461-612023 (D.W.); +49-2461-619497 (J.K.); +49-2461-612302 (A.W.)
| |
Collapse
|
20
|
Jagaraj CJ, Parakh S, Atkin JD. Emerging Evidence Highlighting the Importance of Redox Dysregulation in the Pathogenesis of Amyotrophic Lateral Sclerosis (ALS). Front Cell Neurosci 2021; 14:581950. [PMID: 33679322 PMCID: PMC7929997 DOI: 10.3389/fncel.2020.581950] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022] Open
Abstract
The cellular redox state, or balance between cellular oxidation and reduction reactions, serves as a vital antioxidant defence system that is linked to all important cellular activities. Redox regulation is therefore a fundamental cellular process for aerobic organisms. Whilst oxidative stress is well described in neurodegenerative disorders including amyotrophic lateral sclerosis (ALS), other aspects of redox dysfunction and their contributions to pathophysiology are only just emerging. ALS is a fatal neurodegenerative disease affecting motor neurons, with few useful treatments. Hence there is an urgent need to develop more effective therapeutics in the future. Here, we discuss the increasing evidence for redox dysregulation as an important and primary contributor to ALS pathogenesis, which is associated with multiple disease mechanisms. Understanding the connection between redox homeostasis, proteins that mediate redox regulation, and disease pathophysiology in ALS, may facilitate a better understanding of disease mechanisms, and lead to the design of better therapeutic strategies.
Collapse
Affiliation(s)
- Cyril Jones Jagaraj
- Department of Biomedical Sciences, Macquarie University Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Sonam Parakh
- Department of Biomedical Sciences, Macquarie University Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Julie D Atkin
- Department of Biomedical Sciences, Macquarie University Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
21
|
Neuroprotective Effect of Vascular Endothelial Growth Factor on Motoneurons of the Oculomotor System. Int J Mol Sci 2021; 22:ijms22020814. [PMID: 33467517 PMCID: PMC7830098 DOI: 10.3390/ijms22020814] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 01/04/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) was initially characterized as a potent angiogenic factor based on its activity on the vascular system. However, it is now well established that VEGF also plays a crucial role as a neuroprotective factor in the nervous system. A deficit of VEGF has been related to motoneuronal degeneration, such as that occurring in amyotrophic lateral sclerosis (ALS). Strikingly, motoneurons of the oculomotor system show lesser vulnerability to neurodegeneration in ALS compared to other motoneurons. These motoneurons presented higher amounts of VEGF and its receptor Flk-1 than other brainstem pools. That higher VEGF level could be due to an enhanced retrograde input from their target muscles, but it can also be produced by the motoneurons themselves and act in an autocrine way. By contrast, VEGF’s paracrine supply from the vicinity cells, such as glial cells, seems to represent a minor source of VEGF for brainstem motoneurons. In addition, ocular motoneurons experiment an increase in VEGF and Flk-1 level in response to axotomy, not observed in facial or hypoglossal motoneurons. Therefore, in this review, we summarize the differences in VEGF availability that could contribute to the higher resistance of extraocular motoneurons to injury and neurodegenerative diseases.
Collapse
|
22
|
Guissart C, Mouzat K, Kantar J, Louveau B, Vilquin P, Polge A, Raoul C, Lumbroso S. Premature termination codons in SOD1 causing Amyotrophic Lateral Sclerosis are predicted to escape the nonsense-mediated mRNA decay. Sci Rep 2020; 10:20738. [PMID: 33244158 PMCID: PMC7691510 DOI: 10.1038/s41598-020-77716-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/12/2020] [Indexed: 11/09/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common and severe adult-onset motoneuron disease and has currently no effective therapy. Approximately 20% of familial ALS cases are caused by dominantly-inherited mutations in the gene encoding Cu/Zn superoxide dismutase (SOD1), which represents one of the most frequent genetic cause of ALS. Despite the overwhelming majority of ALS-causing missense mutations in SOD1, a minority of premature termination codons (PTCs) have been identified. mRNA harboring PTCs are known to be rapidly degraded by nonsense-mediated mRNA decay (NMD), which limits the production of truncated proteins. The rules of NMD surveillance varying with PTC location in mRNA, we analyzed the localization of PTCs in SOD1 mRNA to evaluate whether or not those PTCs can be triggered to degradation by the NMD pathway. Our study shows that all pathogenic PTCs described in SOD1 so far can theoretically escape the NMD, resulting in the production of truncated protein. This finding supports the hypothesis that haploinsufficiency is not an underlying mechanism of SOD1 mutant-associated ALS and suggests that PTCs found in the regions that trigger NMD are not pathogenic. Such a consideration is particularly important since the availability of SOD1 antisense strategies, in view of variant treatment assignment.
Collapse
Affiliation(s)
- Claire Guissart
- Laboratoire de Biochimie et Biologie Moléculaire, CHU Nimes, University of Montpellier, Nimes, France.
- The Neuroscience Institute of Montpellier, INM, INSERM, Univ Montpellier, Montpellier, France.
| | - Kevin Mouzat
- Laboratoire de Biochimie et Biologie Moléculaire, CHU Nimes, University of Montpellier, Nimes, France
- The Neuroscience Institute of Montpellier, INM, INSERM, Univ Montpellier, Montpellier, France
| | - Jovana Kantar
- Laboratoire de Biochimie et Biologie Moléculaire, CHU Nimes, University of Montpellier, Nimes, France
- The Neuroscience Institute of Montpellier, INM, INSERM, Univ Montpellier, Montpellier, France
| | - Baptiste Louveau
- Département de Pharmacologie et de Génomique des Tumeurs Solides, Hôpital Saint Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Paul Vilquin
- Département de Pharmacologie et de Génomique des Tumeurs Solides, Hôpital Saint Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Anne Polge
- Laboratoire de Biochimie et Biologie Moléculaire, CHU Nimes, University of Montpellier, Nimes, France
| | - Cédric Raoul
- The Neuroscience Institute of Montpellier, INM, INSERM, Univ Montpellier, Montpellier, France
| | - Serge Lumbroso
- Laboratoire de Biochimie et Biologie Moléculaire, CHU Nimes, University of Montpellier, Nimes, France
- The Neuroscience Institute of Montpellier, INM, INSERM, Univ Montpellier, Montpellier, France
| |
Collapse
|
23
|
Ungaro C, Sprovieri T, Morello G, Perrone B, Spampinato AG, Simone IL, Trojsi F, Monsurrò MR, Spataro R, La Bella V, Andò S, Cavallaro S, Conforti FL. Genetic investigation of amyotrophic lateral sclerosis patients in south Italy: a two-decade analysis. Neurobiol Aging 2020; 99:99.e7-99.e14. [PMID: 32951934 DOI: 10.1016/j.neurobiolaging.2020.08.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/21/2020] [Accepted: 08/22/2020] [Indexed: 10/23/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a multifactorial disease characterized by the interplay of genetic and environmental factors. In the majority of cases, ALS is sporadic, whereas familial forms occur in less than 10% of patients. Herein, we present the results of molecular analyses performed in a large cohort of Italian ALS patients, focusing on novel and already described variations in ALS-linked genes. Our analysis revealed that more than 10% of tested patients carried a mutation in one of the major ALS genes, with C9orf72 hexanucleotide expansion being the most common mutation. In addition, our study confirmed a significant association between ALS patients carrying the ATNX-1 intermediate repeat and the pathological C9orf72 expansion, supporting the involvement of this risk factor in neuronal degeneration. Overall, our study broadens the known mutational spectrum in ALS and provides new insights for a more accurate view of the genetic pattern of the disease.
Collapse
Affiliation(s)
- Carmine Ungaro
- Department of Earth and Environment, Institute of Atmospheric Pollution (IIA), National Research Council (CNR), Rende (CS), Italy
| | - Teresa Sprovieri
- Department of Earth and Environment, Institute of Atmospheric Pollution (IIA), National Research Council (CNR), Rende (CS), Italy
| | - Giovanna Morello
- Department of Biomedical Science, Institute for Research and Biomedical Innovation (IRIB), National Research Council (CNR), Catania, Italy
| | - Benedetta Perrone
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Antonio Gianmaria Spampinato
- Department of Biomedical Science, Institute for Research and Biomedical Innovation (IRIB), National Research Council (CNR), Catania, Italy
| | - Isabella Laura Simone
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Italy
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, MRI Research Center SUN-FISM, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Rosaria Monsurrò
- Department of Advanced Medical and Surgical Sciences, MRI Research Center SUN-FISM, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Vincenzo La Bella
- Department of Experimental Biomedicine and Clinical Neurosciences, ALS Clinical Research Center and Laboratory of Neurochemistry, University of Palermo, Palermo, Italy
| | - Sebastiano Andò
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy; Centro Sanitario, Università della Calabria, Rende (CS), Italy
| | - Sebastiano Cavallaro
- Department of Biomedical Science, Institute for Research and Biomedical Innovation (IRIB), National Research Council (CNR), Catania, Italy
| | - Francesca Luisa Conforti
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy.
| |
Collapse
|
24
|
Borkowski LF, Craig TA, Stricklin OE, Johnson KA, Nichols NL. 5-HT2A/B receptor expression in the phrenic motor nucleus in a rat model of ALS (SOD1 G93A). Respir Physiol Neurobiol 2020; 279:103471. [PMID: 32504811 PMCID: PMC7384973 DOI: 10.1016/j.resp.2020.103471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/23/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
Despite respiratory motor neuron death, ventilation is preserved in SOD1G93A rats. Compensatory respiratory plasticity may counterbalance the loss of these neurons. Phrenic long-term facilitation (pLTF; a form of respiratory plasticity) in naïve rats is 5-HT2 and NADPH oxidase-dependent. Furthermore, 5-HT2A, not 5-HT2B, receptor-induced phrenic motor facilitation is NADPH oxidase-independent in naïve rats. pLTF is NADPH oxidase-dependent in pre-symptomatic, but not end-stage, SOD1G93A rats. Here, we hypothesized that in the putative phrenic motor nucleus (PMN) of SOD1G93A rats vs. wild-type littermates: 1) pre-symptomatic rats would have greater 5-HT2B receptor expression that decreases at end-stage; and 2) 5-HT2A receptor expression would increase from pre-symptomatic to end-stage. Putative PMN 5-HT2A receptor expression was reduced when comparing across (but not within) pre-symptomatic vs. end-stage groups (p < 0.05). In contrast, putative PMN 5-HT2B receptor expression was increased when comparing across pre-symptomatic vs. end-stage groups, and within end-stage groups (p < 0.05). These data suggest a potential role for 5-HT2 receptors in pLTF and breathing in SOD1G93A rats.
Collapse
Affiliation(s)
- Lauren F Borkowski
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States
| | - Taylor A Craig
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States
| | - Olivia E Stricklin
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States
| | - Katherine A Johnson
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States
| | - Nicole L Nichols
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States.
| |
Collapse
|
25
|
Martinelli I, Zucchi E, Gessani A, Fini N, Chiò A, Pecoraro V, Trenti T, Mandrioli J. A novel p.N66T mutation in exon 3 of the SOD1 gene: report of two families of ALS patients with early cognitive impairment. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:296-300. [PMID: 32248719 DOI: 10.1080/21678421.2020.1746344] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: To date more than 180 different mutations in the SOD1 gene have been described in ALS; some of these mutations are associated to peculiar clinical features and have contributed to the understanding of disease heterogeneity. Only 5% of SOD1 mutations involve exon 3. Here we report a novel mutation c.197A > C in the exon 3 of the SOD1 gene in two apparently unrelated ALS families with early respiratory and cognitive impairment.Case report: In the first family two brothers developed ALS in their seventies, with arm weakness followed by bulbar involvement and behavioral breakdown. An unrelated 57-year-old man presented with progressive leg weakness and mild compromised executive functions without known family history for ALS/FTD and underwent invasive ventilation in a few months. A novel missense mutation A to C at codon 197 in exon 3 causing aminoacid substitution of arginine by threonine (N66T) was found for all of them. Harmful consequences of c.197A > C mutation on SOD1 function were suggested by in silico prediction and homology with other known mutations at the same position.Discussion and conclusion: Here, we report two apparently unrelated ALS families carrying a novel SOD1 mutation (N66T), supporting its pathogenic role by primary analysis, and characterized by early bulbar, respiratory, and cognitive involvement. Early cognitive impairment has been rarely described in ALS caused by SOD1 mutations, and mainly in the later phases of the disease. This report provides additional data on the SOD1 mutation spectrum and clinical presentation of ALS, widening phenotypical characterization of SOD1 ALS.
Collapse
Affiliation(s)
- Ilaria Martinelli
- Department of Neuroscience, Ospedale Civile S. Agostino Estense, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Elisabetta Zucchi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Annalisa Gessani
- Department of Neuroscience, Ospedale Civile S. Agostino Estense, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Nicola Fini
- Department of Neuroscience, Ospedale Civile S. Agostino Estense, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Adriano Chiò
- ALS Center "Rita Levi Montalcini" Department of Neuroscience, University of Torino, Torino, Italy; Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Torino, Italy; The Neuroscience Institute of Torino, Torino, Italy
| | - Valentina Pecoraro
- Laboratory of Toxycology and Advanced Diagnostics, Department of Laboratory Medicine and Pathology, Ospedale Civile S. Agostino Estense, Modena, Italy
| | - Tommaso Trenti
- Laboratory of Toxycology and Advanced Diagnostics, Department of Laboratory Medicine and Pathology, Ospedale Civile S. Agostino Estense, Modena, Italy
| | - Jessica Mandrioli
- Department of Neuroscience, Ospedale Civile S. Agostino Estense, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| |
Collapse
|
26
|
Ghosh DK, Kumar A, Ranjan A. T54R mutation destabilizes the dimer of superoxide dismutase 1T54R by inducing steric clashes at the dimer interface. RSC Adv 2020; 10:10776-10788. [PMID: 35492906 PMCID: PMC9050410 DOI: 10.1039/c9ra09870d] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/28/2020] [Indexed: 11/29/2022] Open
Abstract
Mutations cause abnormalities in protein structure, function and oligomerization. Different mutations in the superoxide dismutase 1 (SOD1) protein cause its misfolding, loss of dimerization and aggravate its aggregation in the amyotrophic lateral sclerosis disease. In this study, we report the mechanistic details of how a threonine-to-arginine mutation at the 54th position (T54R) of SOD1 results in destabilization of the dimer interface of SOD1T54R. Using computational and experimental methods, we show that the T54R mutation increases fluctuation of the mutation-harboring loop (R54-loop) of SOD1T54R. Fluctuation of this loop causes steric clashes that involve arginine-54 (R54) and other residues of SOD1T54R, resulting in loss of inter-subunit contacts at the dimer interface. Since the T54 residue-containing loop is necessary for the dimerization of wild-type SOD1, fluctuation of the R54-loop, steric clashes involving R54 and loss of inter-subunit contacts give rise to the loss of SOD1T54R dimer stability. This correlates to energetically unfavorable tethering of the monomers of SOD1T54R. The outcome is gradual splitting of SOD1T54R dimers into monomers, thereby exposing the previously buried hydrophobic interface residues to the aqueous environment. This event finally leads to aggregation of SOD1T54R. T54R mutation has no effect in altering the relative positions of copper and zinc ion binding residues of SOD1T54R. The native SOD1 structure is stable, and there is no destabilizing effect at its dimer interface. Overall, our study reveals the intricate mechanism of T54R mutation-associated destabilization of the dimer of the SOD1T54R protein. T54R mutation destabilizes the dimer of SOD1T54R.![]()
Collapse
Affiliation(s)
- Debasish Kumar Ghosh
- Computational and Functional Genomics Group
- Centre for DNA Fingerprinting and Diagnostics
- Hyderabad 500039
- India
| | - Abhishek Kumar
- Computational and Functional Genomics Group
- Centre for DNA Fingerprinting and Diagnostics
- Hyderabad 500039
- India
- Graduate Studies
| | - Akash Ranjan
- Computational and Functional Genomics Group
- Centre for DNA Fingerprinting and Diagnostics
- Hyderabad 500039
- India
| |
Collapse
|
27
|
Chowdhury S, Sanyal D, Sen S, Uversky VN, Maulik U, Chattopadhyay K. Evolutionary Analyses of Sequence and Structure Space Unravel the Structural Facets of SOD1. Biomolecules 2019; 9:E826. [PMID: 31817166 PMCID: PMC6995586 DOI: 10.3390/biom9120826] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/09/2019] [Accepted: 11/16/2019] [Indexed: 01/08/2023] Open
Abstract
Superoxide dismutase (SOD) is the primary enzyme of the cellular antioxidant defense cascade. Misfolding, concomitant oligomerization, and higher order aggregation of human cytosolic SOD are linked to amyotrophic lateral sclerosis (ALS). Although, with two metal ion cofactors SOD1 is extremely robust, the de-metallated apo form is intrinsically disordered. Since the rise of oxygen-based metabolism and antioxidant defense systems are evolutionary coupled, SOD is an interesting protein with a deep evolutionary history. We deployed statistical analysis of sequence space to decode evolutionarily co-varying residues in this protein. These were validated by applying graph theoretical modelling to understand the impact of the presence of metal ion co-factors in dictating the disordered (apo) to hidden disordered (wild-type SOD1) transition. Contact maps were generated for different variants, and the selected significant residues were mapped on separate structure networks. Sequence space analysis coupled with structure networks helped us to map the evolutionarily coupled co-varying patches in the SOD1 and its metal-depleted variants. In addition, using structure network analysis, the residues with a major impact on the internal dynamics of the protein structure were investigated. Our results reveal that the bulk of these evolutionarily co-varying residues are localized in the loop regions and positioned differentially depending upon the metal residence and concomitant steric restrictions of the loops.
Collapse
Affiliation(s)
- Sourav Chowdhury
- Protein Folding and Dynamics Group, Structural Biology and Bio-informatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C.Mullick Road, Kolkata 700032, India; (S.C.); (D.S.)
- Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA
| | - Dwipanjan Sanyal
- Protein Folding and Dynamics Group, Structural Biology and Bio-informatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C.Mullick Road, Kolkata 700032, India; (S.C.); (D.S.)
| | - Sagnik Sen
- Department of Computer Science, Jadavpur University, Kolkata 700032, India; (S.S.); (U.M.)
| | - Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd. MDC07, Tampa, FL 33612, USA;
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino 142290, Moscow Region, Russia
| | - Ujjwal Maulik
- Department of Computer Science, Jadavpur University, Kolkata 700032, India; (S.S.); (U.M.)
| | - Krishnananda Chattopadhyay
- Protein Folding and Dynamics Group, Structural Biology and Bio-informatics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C.Mullick Road, Kolkata 700032, India; (S.C.); (D.S.)
| |
Collapse
|
28
|
Garcia C, Vidal-Taboada JM, Syriani E, Salvado M, Morales M, Gamez J. Haplotype Analysis of the First A4V- SOD1 Spanish Family: Two Separate Founders or a Single Common Founder? Front Genet 2019; 10:1109. [PMID: 31781168 PMCID: PMC6857184 DOI: 10.3389/fgene.2019.01109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 10/16/2019] [Indexed: 12/02/2022] Open
Abstract
Despite the genetic heterogeneity reported in familial amyotrophic lateral sclerosis (ALS) (fALS), Cu/Zn superoxide-dismutase (SOD1) gene mutations are the second most common cause of the disease, accounting for around 20% of all families (ALS1) and isolated sporadic cases (sALS). At least 186 different mutations in the SOD1 gene have been reported to date. The possibility of a single founder and separate founders have been investigated for D90A (p.D91A) and A4V (p.A5V), the most common mutations worldwide. High-throughput single nucleotide polymorphism genotyping studies have suggested two founders for A4V (one for the Amerindian population and another for the European population) although the possibility that the two populations are descended from a single ancient founder cannot be ruled out. We used 15 genetic variants spanning the human chromosome 21 from the SOD1 gene to the SCAF4 gene, comparing them with the population reference panels, to demonstrate that the first A4V Spanish pedigree shared the genetic background reported in the European population.
Collapse
Affiliation(s)
- Cecilia Garcia
- ALS Unit, Neurology Department, Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain
| | - Jose Manuel Vidal-Taboada
- ALS Unit, Neurology Department, Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain
| | - Enrique Syriani
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain
| | - Maria Salvado
- ALS Unit, Neurology Department, Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Miguel Morales
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain
| | - Josep Gamez
- ALS Unit, Neurology Department, Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
29
|
Abstract
Few proteins have come under such intense scrutiny as superoxide dismutase-1 (SOD1). For almost a century, scientists have dissected its form, function and then later its malfunction in the neurodegenerative disease amyotrophic lateral sclerosis (ALS). We now know SOD1 is a zinc and copper metalloenzyme that clears superoxide as part of our antioxidant defence and respiratory regulation systems. The possibility of reduced structural integrity was suggested by the first crystal structures of human SOD1 even before deleterious mutations in the sod1 gene were linked to the ALS. This concept evolved in the intervening years as an impressive array of biophysical studies examined the characteristics of mutant SOD1 in great detail. We now recognise how ALS-related mutations perturb the SOD1 maturation processes, reduce its ability to fold and reduce its thermal stability and half-life. Mutant SOD1 is therefore predisposed to monomerisation, non-canonical self-interactions, the formation of small misfolded oligomers and ultimately accumulation in the tell-tale insoluble inclusions found within the neurons of ALS patients. We have also seen that several post-translational modifications could push wild-type SOD1 down this toxic pathway. Recently we have come to view ALS as a prion-like disease where both the symptoms, and indeed SOD1 misfolding itself, are transmitted to neighbouring cells. This raises the possibility of intervention after the initial disease presentation. Several small-molecule and biologic-based strategies have been devised which directly target the SOD1 molecule to change the behaviour thought to be responsible for ALS. Here we provide a comprehensive review of the many biophysical advances that sculpted our view of SOD1 biology and the recent work that aims to apply this knowledge for therapeutic outcomes in ALS.
Collapse
|
30
|
Ragagnin AMG, Shadfar S, Vidal M, Jamali MS, Atkin JD. Motor Neuron Susceptibility in ALS/FTD. Front Neurosci 2019; 13:532. [PMID: 31316328 PMCID: PMC6610326 DOI: 10.3389/fnins.2019.00532] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/08/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the death of both upper and lower motor neurons (MNs) in the brain, brainstem and spinal cord. The neurodegenerative mechanisms leading to MN loss in ALS are not fully understood. Importantly, the reasons why MNs are specifically targeted in this disorder are unclear, when the proteins associated genetically or pathologically with ALS are expressed ubiquitously. Furthermore, MNs themselves are not affected equally; specific MNs subpopulations are more susceptible than others in both animal models and human patients. Corticospinal MNs and lower somatic MNs, which innervate voluntary muscles, degenerate more readily than specific subgroups of lower MNs, which remain resistant to degeneration, reflecting the clinical manifestations of ALS. In this review, we discuss the possible factors intrinsic to MNs that render them uniquely susceptible to neurodegeneration in ALS. We also speculate why some MN subpopulations are more vulnerable than others, focusing on both their molecular and physiological properties. Finally, we review the anatomical network and neuronal microenvironment as determinants of MN subtype vulnerability and hence the progression of ALS.
Collapse
Affiliation(s)
- Audrey M G Ragagnin
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Sina Shadfar
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Marta Vidal
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Md Shafi Jamali
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Julie D Atkin
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
31
|
Kuuluvainen L, Kaivola K, Mönkäre S, Laaksovirta H, Jokela M, Udd B, Valori M, Pasanen P, Paetau A, Traynor BJ, Stone DJ, Schleutker J, Pöyhönen M, Tienari PJ, Myllykangas L. Oligogenic basis of sporadic ALS: The example of SOD1 p.Ala90Val mutation. Neurol Genet 2019; 5:e335. [PMID: 31086828 PMCID: PMC6481226 DOI: 10.1212/nxg.0000000000000335] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/01/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To characterize the clinical and neuropathologic features of patients with amyotrophic lateral sclerosis (ALS) with the superoxide dismutase 1 (SOD1) p.Ala90Val mutation, as well as the mutation frequency and the role of oligogenic mechanisms in disease penetrance. METHODS An index patient with autopsy-proven ALS was discovered to have the SOD1 p.Ala90Val mutation, which was screened in 2 Finnish ALS cohorts (n = 453). Additional contributing variants were analyzed from whole-genome or whole-exome sequencing data. RESULTS Seven screened patients (1.5%) were found to carry the SOD1 heterozygous mutation. Allele-sharing analysis suggested a common founder haplotype. Common clinical features included limb-onset, long disease course, and sensory symptoms. No TDP43 pathology was observed. All cases were apparently sporadic, and pedigree analysis demonstrated that the mutation has reduced penetrance. Analysis of other contributing genes revealed a unique set of additional variants in each patient. These included previously described rare ANG and SPG11 mutations. One patient was compound heterozygous for SOD1 p.Ala90Val and p.Asp91Ala. CONCLUSIONS Our data suggest that the penetrance of SOD1 p.Ala90Val is modulated by other genes and indicates highly individual oligogenic basis of apparently sporadic ALS. Additional genetic variants likely contributing to disease penetrance were very heterogeneous, even among Finnish patients carrying the SOD1 founder mutation.
Collapse
Affiliation(s)
- Liina Kuuluvainen
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Karri Kaivola
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Saana Mönkäre
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hannu Laaksovirta
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Manu Jokela
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Bjarne Udd
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Miko Valori
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Petra Pasanen
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anders Paetau
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Bryan J Traynor
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - David J Stone
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Johanna Schleutker
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Minna Pöyhönen
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pentti J Tienari
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Liisa Myllykangas
- Department of Clinical Genetics (L.K.), Helsinki University Hospital; Department of Medical Genetics (L.K.), University of Helsinki, Helsinki, Finland; Molecular Neurology (K.K., M.V., P.J.T.), Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Department of Medical Genetics (S.M.), University of Helsinki, Helsinki, Finland and Turku; University Hospital (S.M.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Neurology (H.L.), Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center (M.J., B.U.), Tampere University Hospital and University of Tampere, Tampere, Finland; Division of Clinical Neurosciences (M.J.), Turku University Hospital and University of Turku, Turku, Finland; Folkhälsan Research Center (B.U.), Biomedicum, University of Helsinki, Helsinki, Finland; Institute of Biomedicine (P.P., J.S.), University of Turku; Turku University Hospital (P.P., J.S.), Laboratory Division, Genetics and Saske, Department of Medical Genetics, Turku, Finland; Department of Pathology (A.P.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Laboratory of Neurogenetics (B.J.T.), National Institute on Aging, National Institutes of Health, Bethesda, MD; Merck & Co. (D.J.S.), Inc., West Point, PA; Department of Clinical Genetics (M.P.), Helsinki University Hospital; Department of Medical Genetics (M.P.), University of Helsinki, Helsinki, Finland; Department of Neurology (P.J.T.), Helsinki University Hospital; and Department of Pathology (L.M.), University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
32
|
Parakh S, Perri ER, Jagaraj CJ, Ragagnin AMG, Atkin JD. Rab-dependent cellular trafficking and amyotrophic lateral sclerosis. Crit Rev Biochem Mol Biol 2019; 53:623-651. [PMID: 30741580 DOI: 10.1080/10409238.2018.1553926] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Rab GTPases are becoming increasingly implicated in neurodegenerative disorders, although their role in amyotrophic lateral sclerosis (ALS) has been somewhat overlooked. However, dysfunction of intracellular transport is gaining increasing attention as a pathogenic mechanism in ALS. Many previous studies have focused axonal trafficking, and the extreme length of axons in motor neurons may contribute to their unique susceptibility in this disorder. In contrast, the role of transport defects within the cell body has been relatively neglected. Similarly, whilst Rab GTPases control all intracellular membrane trafficking events, their role in ALS is poorly understood. Emerging evidence now highlights this family of proteins in ALS, particularly the discovery that C9orf72 functions in intra transport in conjunction with several Rab GTPases. Here, we summarize recent updates on cellular transport defects in ALS, with a focus on Rab GTPases and how their dysfunction may specifically target neurons and contribute to pathophysiology. We discuss the molecular mechanisms associated with dysfunction of Rab proteins in ALS. Finally, we also discuss dysfunction in other modes of transport recently implicated in ALS, including nucleocytoplasmic transport and the ER-mitochondrial contact regions (MAM compartment), and speculate whether these may also involve Rab GTPases.
Collapse
Affiliation(s)
- S Parakh
- a Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Centre for MND Research , Macquarie University , Sydney , Australia.,b Department of Biochemistry and Genetics , La Trobe Institute for Molecular Science, La Trobe University , Melbourne , Australia
| | - E R Perri
- a Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Centre for MND Research , Macquarie University , Sydney , Australia.,b Department of Biochemistry and Genetics , La Trobe Institute for Molecular Science, La Trobe University , Melbourne , Australia
| | - C J Jagaraj
- a Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Centre for MND Research , Macquarie University , Sydney , Australia
| | - A M G Ragagnin
- a Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Centre for MND Research , Macquarie University , Sydney , Australia
| | - J D Atkin
- a Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Centre for MND Research , Macquarie University , Sydney , Australia.,b Department of Biochemistry and Genetics , La Trobe Institute for Molecular Science, La Trobe University , Melbourne , Australia
| |
Collapse
|
33
|
Czell D. [Genetics of Amyotrophic Lateral Sclerosis]. PRAXIS 2019; 108:37-44. [PMID: 30621546 DOI: 10.1024/1661-8157/a003153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Genetics of Amyotrophic Lateral Sclerosis Abstract. In recent years, the knowledge of gene mutation that can lead to amyotrophic lateral sclerosis (ALS), frontotemporal dementia, or a FTD-ALS complex has been drastically improvedwith the help of next-generation sequencing (NGS), so that many new genes and their molecular genetic mechanisms and symptoms of the patients could be described. For example it was found that in the same family with the same gene mutation patients with ALS, FTD or another neurodegenerative disease can be found. The knowledge about the genes does not only contribute to the understanding of these diseases, but should help develop new therapeutic possibilities.
Collapse
Affiliation(s)
- David Czell
- 1 Klinik für Innere Medizin/Neurologie, Spital Linth, Uznach
| |
Collapse
|
34
|
Canosa A, De Marco G, Lomartire A, Rinaudo MT, Di Cunto F, Turco E, Barberis M, Brunetti M, Casale F, Moglia C, Calvo A, Marklund SL, Andersen PM, Mora G, Chiò A. A novel p.Ser108LeufsTer15 SOD1 mutation leading to the formation of a premature stop codon in an apparently sporadic ALS patient: insights into the underlying pathomechanisms. Neurobiol Aging 2018; 72:189.e11-189.e17. [PMID: 30236613 DOI: 10.1016/j.neurobiolaging.2018.08.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 07/12/2018] [Accepted: 08/15/2018] [Indexed: 12/14/2022]
Abstract
We report an apparently sporadic amyotrophic lateral sclerosis patient carrying a heterozygous novel frameshift SOD1 mutation (p.Ser108LeufsTer15), predicted to cause a premature protein truncation. RT-PCR analysis of SOD1 mRNA and SDS-PAGE/Western blot analysis of PBMC demonstrated that mRNA from the mutant allele is expressed at levels similar to those of the wild-type allele, but the truncated protein is undetectable also in the insoluble fraction and after proteasome inhibition. Accordingly, the dismutation activity in erythrocytes is halved. Thus, the pathogenic mechanism associated with this mutation might be based on an insufficient activity of SOD1 that would make motor neurons more vulnerable to oxidative injury. However, it cannot be excluded that p.Ser108LeufsTer15 SOD1 is present in the nervous tissue and, being less charged and hence having less repulsive forces than the wild-type protein, may trigger toxic mechanisms as a consequence of its propensity to aggregate.
Collapse
Affiliation(s)
- Antonio Canosa
- ALS Centre, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy.
| | - Giovanni De Marco
- ALS Centre, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy
| | - Annarosa Lomartire
- ALS Centre, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy
| | | | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Orbassano, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Marco Barberis
- ALS Centre, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy
| | - Maura Brunetti
- ALS Centre, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy
| | - Federico Casale
- ALS Centre, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy
| | - Cristina Moglia
- ALS Centre, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy; Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, S.C. Neurologia 2 U, Turin, Italy
| | - Andrea Calvo
- ALS Centre, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy; Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, S.C. Neurologia 2 U, Turin, Italy; Neuroscience Institute of Turin (NIT), Turin, Italy
| | - Stefan L Marklund
- Department of Pharmacology and Clinical Neurosciences, Umeå University, Umeå, Sweden
| | - Peter M Andersen
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Gabriele Mora
- ALS Centre, "Salvatore Maugeri" Clinical-Scientific Institutes, IRCCS, Neurological Rehabilitation Unit, Milan, Italy
| | - Adriano Chiò
- ALS Centre, "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy; Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, S.C. Neurologia 2 U, Turin, Italy; Neuroscience Institute of Turin (NIT), Turin, Italy; Institute of Cognitive Sciences and Technologies, C.N.R, Rome, Italy
| |
Collapse
|
35
|
Müller K, Brenner D, Weydt P, Meyer T, Grehl T, Petri S, Grosskreutz J, Schuster J, Volk AE, Borck G, Kubisch C, Klopstock T, Zeller D, Jablonka S, Sendtner M, Klebe S, Knehr A, Günther K, Weis J, Claeys KG, Schrank B, Sperfeld AD, Hübers A, Otto M, Dorst J, Meitinger T, Strom TM, Andersen PM, Ludolph AC, Weishaupt JH. Comprehensive analysis of the mutation spectrum in 301 German ALS families. J Neurol Neurosurg Psychiatry 2018; 89:817-827. [PMID: 29650794 DOI: 10.1136/jnnp-2017-317611] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/25/2018] [Accepted: 03/07/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Recent advances in amyotrophic lateral sclerosis (ALS) genetics have revealed that mutations in any of more than 25 genes can cause ALS, mostly as an autosomal-dominant Mendelian trait. Detailed knowledge about the genetic architecture of ALS in a specific population will be important for genetic counselling but also for genotype-specific therapeutic interventions. METHODS Here we combined fragment length analysis, repeat-primed PCR, Southern blotting, Sanger sequencing and whole exome sequencing to obtain a comprehensive profile of genetic variants in ALS disease genes in 301 German pedigrees with familial ALS. We report C9orf72 mutations as well as variants in consensus splice sites and non-synonymous variants in protein-coding regions of ALS genes. We furthermore estimate their pathogenicity by taking into account type and frequency of the respective variant as well as segregation within the families. RESULTS 49% of our German ALS families carried a likely pathogenic variant in at least one of the earlier identified ALS genes. In 45% of the ALS families, likely pathogenic variants were detected in C9orf72, SOD1, FUS, TARDBP or TBK1, whereas the relative contribution of the other ALS genes in this familial ALS cohort was 4%. We identified several previously unreported rare variants and demonstrated the absence of likely pathogenic variants in some of the recently described ALS disease genes. CONCLUSIONS We here present a comprehensive genetic characterisation of German familial ALS. The present findings are of importance for genetic counselling in clinical practice, for molecular research and for the design of diagnostic gene panels or genotype-specific therapeutic interventions in Europe.
Collapse
Affiliation(s)
| | - David Brenner
- Department of Neurology, Ulm University, Ulm, Germany
| | - Patrick Weydt
- Department of Neurology, Ulm University, Ulm, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, Bonn University, Bonn, Germany
| | - Thomas Meyer
- Department of Neurology, Charité Hospital, Humboldt University, Berlin, Germany
| | - Torsten Grehl
- Department of Neurology, Alfried Krupp Hospital, Essen, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | | | - Alexander E Volk
- Institute of Human Genetics, Ulm University, Ulm, Germany.,Institute of Human Genetics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Guntram Borck
- Institute of Human Genetics, Ulm University, Ulm, Germany
| | - Christian Kubisch
- Institute of Human Genetics, Ulm University, Ulm, Germany.,Institute of Human Genetics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institut, University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Daniel Zeller
- Department of Neurology, University of Würzburg, Würzburg, Germany
| | - Sibylle Jablonka
- Institute of Clinical Neurobiology, University Hospital of Würzburg, Würzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital of Würzburg, Würzburg, Germany
| | - Stephan Klebe
- Department of Neurology, University of Würzburg, Würzburg, Germany.,Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Antje Knehr
- Department of Neurology, Ulm University, Ulm, Germany
| | | | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Kristl G Claeys
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany.,Department of Neurology, RWTH Aachen University Hospital, Aachen, Germany.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium.,Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, Experimental Neurology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Berthold Schrank
- Department of Neurology, DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany
| | - Anne-Dorte Sperfeld
- Department of Neurology, Martin Luther University of Halle-Wittenberg, Halle/Saale, Germany
| | | | - Markus Otto
- Department of Neurology, Ulm University, Ulm, Germany
| | | | - Thomas Meitinger
- Institute of Human Genetics, Technische Universität München, Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Munich Cluster for Systems Neurology (SNergy), Munich, Germany
| | - Tim M Strom
- Institute of Human Genetics, Technische Universität München, Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Munich Cluster for Systems Neurology (SNergy), Munich, Germany
| | - Peter M Andersen
- Department of Neurology, Ulm University, Ulm, Germany.,Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | | | | | | |
Collapse
|
36
|
Vejux A, Namsi A, Nury T, Moreau T, Lizard G. Biomarkers of Amyotrophic Lateral Sclerosis: Current Status and Interest of Oxysterols and Phytosterols. Front Mol Neurosci 2018; 11:12. [PMID: 29445325 PMCID: PMC5797798 DOI: 10.3389/fnmol.2018.00012] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 01/09/2018] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a non-demyelinating neurodegenerative disease in adults with motor disorders. Two forms exist: a sporadic form (90% of cases) and a family form due to mutations in more than 20 genes including the Superoxide dismutase 1, TAR DNA Binding Protein, Fused in Sarcoma, chromosome 9 open reading frame 72 and VAPB genes. The mechanisms associated with this pathology are beginning to be known: oxidative stress, glutamate excitotoxicity, protein aggregation, reticulum endoplasmic stress, neuroinflammation, alteration of RNA metabolism. In various neurodegenerative diseases, such as Alzheimer's disease or multiple sclerosis, the involvement of lipids is increasingly suggested based on lipid metabolism modifications. With regard to ALS, research has also focused on the possible involvement of lipids. Lipid involvement was suggested for clinical arguments where changes in cholesterol and LDL/HDL levels were reported with, however, differences in positivity between studies. Since lipids are involved in the membrane structure and certain signaling pathways, it may be considered to look for oxysterols, mainly 25-hydroxycholesterol and its metabolites involved in immune response, or phytosterols to find suitable biomarkers for this pathology.
Collapse
Affiliation(s)
- Anne Vejux
- Team Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism EA 7270, INSERM, University of Bourgogne Franche-Comté, Dijon, France
| | - Amira Namsi
- Team Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism EA 7270, INSERM, University of Bourgogne Franche-Comté, Dijon, France.,Laboratoire de Neurophysiologie Fonctionnelle et Pathologies, UR11ES/09, Faculté des Sciences Mathématiques, Physiques et Naturelles de Tunis, Université de Tunis El Manar - Bienvenue, Tunis, Tunisia
| | - Thomas Nury
- Team Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism EA 7270, INSERM, University of Bourgogne Franche-Comté, Dijon, France
| | - Thibault Moreau
- Team Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism EA 7270, INSERM, University of Bourgogne Franche-Comté, Dijon, France.,Department of Neurology, University Hospital/University Bourgogne Franche-Comté, Dijon, France
| | - Gérard Lizard
- Team Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism EA 7270, INSERM, University of Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
37
|
Ticozzi N, Silani V. Genotypic and Phenotypic Heterogeneity in Amyotrophic Lateral Sclerosis. NEURODEGENER DIS 2018. [DOI: 10.1007/978-3-319-72938-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
38
|
Goutman SA, Chen KS, Paez-Colasante X, Feldman EL. Emerging understanding of the genotype-phenotype relationship in amyotrophic lateral sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2018; 148:603-623. [PMID: 29478603 DOI: 10.1016/b978-0-444-64076-5.00039-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, noncurable neurodegenerative disorder of the upper and lower motor neurons causing weakness and death within a few years of symptom onset. About 10% of patients with ALS have a family history of the disease; however, ALS-associated genetic mutations are also found in sporadic cases. There are over 100 ALS-associated mutations, and importantly, several genetic mutations, including C9ORF72, SOD1, and TARDBP, have led to mechanistic insight into this complex disease. In the clinical realm, knowledge of ALS genetics can also help explain phenotypic heterogeneity, aid in genetic counseling, and in the future may help direct treatment efforts.
Collapse
Affiliation(s)
- Stephen A Goutman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States.
| | - Kevin S Chen
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | | | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
39
|
Tompa DR, Kadhirvel S. Molecular dynamics of a far positioned SOD1 mutant V14M reveals pathogenic misfolding behavior. J Biomol Struct Dyn 2017; 36:4085-4098. [PMID: 29157189 DOI: 10.1080/07391102.2017.1407675] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Human superoxide dismutase (Cu/Zn SOD1) is a homodimeric enzyme. Mutations in Cu/Zn SOD1 causes a familial form of amyotrophic lateral sclerosis (fALS), and aggregation of mutant SOD1 has been proposed to play a role in neurodegeneration. Though a majority of the mutations are point substitutions, there are a few changes that result in amino acid deletions or truncations of the polypeptide. These pathogenic mutations are scattered throughout the three-dimensional structure of the dimeric enzyme, which creates a puzzling pattern to investigate the molecular determinants of fALS. The most common hypothesis proposed that the misfolding of SOD1 mutants are primarily triggered by decreased affinity for metal ions. However, this hypothesis is challenging, as a significant number of disease-causing mutations are located far away from the metal-binding site and dimer interface. So in the present study, we have investigated the influence of such a far positioned pathogenic mutation, V14M, in altering the stability and folding of the Cu/Zn SOD1. Though the location of Val14 is far positioned, it has a vital role in the stability of SOD1 by preserving its hydrophobic cluster at one end of the β barrel domain. We have performed MD simulations of the V14M mutant for 80 ns timescale. The results reveal the fact that irrespective of its location, V14M mutation triggers a conformational change that is more similar to that of the metal-deficient holo form and could resemble an intermediate state in the folding reaction which results in protein misfolding and aggregation.
Collapse
Affiliation(s)
- Dharma Rao Tompa
- a Biomolecular Crystallography Laboratory, Department of Bioinformatics, School of Chemical and Biotechnology , SASTRA University , Thanjavur 613 401 , India
| | - Saraboji Kadhirvel
- a Biomolecular Crystallography Laboratory, Department of Bioinformatics, School of Chemical and Biotechnology , SASTRA University , Thanjavur 613 401 , India
| |
Collapse
|
40
|
Abu-Hamad S, Kahn J, Leyton-Jaimes MF, Rosenblatt J, Israelson A. Misfolded SOD1 Accumulation and Mitochondrial Association Contribute to the Selective Vulnerability of Motor Neurons in Familial ALS: Correlation to Human Disease. ACS Chem Neurosci 2017; 8:2225-2234. [PMID: 28715630 DOI: 10.1021/acschemneuro.7b00140] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder, with a 10% genetic linkage, of which 20% of these cases may be attributed to mutations in superoxide dismutase (SOD1). Specific mutations in SOD1 have been associated with disease duration, which can be highly variable ranging from a life expectancy of 3 to beyond 10 years. SOD1 neurotoxicity has been attributed to aberrant accumulation of misfolded SOD1, which in its soluble form binds to intracellular organelles disrupting their function or forms insoluble toxic aggregates. To understand whether these biophysical properties of the mutant protein may influence disease onset and duration, we generated 19 point mutations in the SOD1 gene, based on available clinical data of disease onset and progression from patients. By overexpressing these mutants in motor-neuron-like NSC-34 cells, we demonstrate a variability in misfolding capacity between the different mutants with a correlation between the degree of protein misfolding and mutation severity. We also show a clear variation of the different SOD1 mutants to associate with mitochondrial-enriched fractions with a correlation between mutation severity and this association. In summary, these findings reveal a correlation between the accumulation of misfolded SOD1 species and their mitochondrial association with disease duration but not with disease onset, and they have implications for the potential therapeutic role of suppressing the accumulation of misfolded SOD1.
Collapse
Affiliation(s)
- Salah Abu-Hamad
- Department
of Physiology and Cell Biology, Faculty of Health Sciences, ‡Department of Industrial
Engineering and Management, and §The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel
| | - Joy Kahn
- Department
of Physiology and Cell Biology, Faculty of Health Sciences, ‡Department of Industrial
Engineering and Management, and §The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel
| | - Marcel F. Leyton-Jaimes
- Department
of Physiology and Cell Biology, Faculty of Health Sciences, ‡Department of Industrial
Engineering and Management, and §The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel
| | - Jonathan Rosenblatt
- Department
of Physiology and Cell Biology, Faculty of Health Sciences, ‡Department of Industrial
Engineering and Management, and §The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel
| | - Adrian Israelson
- Department
of Physiology and Cell Biology, Faculty of Health Sciences, ‡Department of Industrial
Engineering and Management, and §The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, P.O.B. 653, Beer Sheva 84105, Israel
| |
Collapse
|
41
|
Naruse H, Ishiura H, Mitsui J, Date H, Takahashi Y, Matsukawa T, Tanaka M, Ishii A, Tamaoka A, Hokkoku K, Sonoo M, Segawa M, Ugawa Y, Doi K, Yoshimura J, Morishita S, Goto J, Tsuji S. Molecular epidemiological study of familial amyotrophic lateral sclerosis in Japanese population by whole-exome sequencing and identification of novel HNRNPA1 mutation. Neurobiol Aging 2017; 61:255.e9-255.e16. [PMID: 29033165 DOI: 10.1016/j.neurobiolaging.2017.08.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 08/27/2017] [Accepted: 08/30/2017] [Indexed: 12/13/2022]
Abstract
To elucidate the genetic epidemiology of familial amyotrophic lateral sclerosis (FALS) in the Japanese population, we conducted whole-exome sequencing analysis of 30 FALS families in whom causative mutations have not been identified in previous studies. Consequently, whole-exome sequencing analysis revealed novel mutations in HNRNPA1, TBK1, and VCP. Taken together with our previous results of mutational analyses by direct nucleotide sequencing analysis, a microarray-based resequencing method, or repeat-primed PCR analysis, causative mutations were identified in 41 of the 68 families (60.3%) with SOD1 being the most frequent cause of FALS (39.7%). Of the mutations identified in this study, a novel c.862/1018C>G (p.P288A/340A) mutation in HNRNPA1 located in the nuclear localization signal domain of hnRNPA1, enhances the recruitment of mutant hnRNPA1 into stress granules, indicating that an altered nuclear localization signal activity plays an essential role in amyotrophic lateral sclerosis pathogenesis.
Collapse
Affiliation(s)
- Hiroya Naruse
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Ishiura
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Mitsui
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hidetoshi Date
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuji Takahashi
- Department of Neurology, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takashi Matsukawa
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaki Tanaka
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akiko Ishii
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Akira Tamaoka
- Department of Neurology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Keiichi Hokkoku
- Department of Neurology, Teikyo University School of Medicine, Tokyo, Japan
| | - Masahiro Sonoo
- Department of Neurology, Teikyo University School of Medicine, Tokyo, Japan
| | - Mari Segawa
- Department of Neurology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yoshikazu Ugawa
- Department of Neurology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Koichiro Doi
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Jun Yoshimura
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Shinichi Morishita
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Jun Goto
- Department of Neurology, International University of Health and Welfare Mita Hospital, Tokyo, Japan
| | - Shoji Tsuji
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
42
|
Park JH, Jang HR, Lee IY, Oh HK, Choi EJ, Rhim H, Kang S. Amyotrophic lateral sclerosis-related mutant superoxide dismutase 1 aggregates inhibit 14-3-3-mediated cell survival by sequestration into the JUNQ compartment. Hum Mol Genet 2017; 26:3615-3629. [DOI: 10.1093/hmg/ddx250] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/26/2017] [Indexed: 12/11/2022] Open
|
43
|
Shahheydari H, Ragagnin A, Walker AK, Toth RP, Vidal M, Jagaraj CJ, Perri ER, Konopka A, Sultana JM, Atkin JD. Protein Quality Control and the Amyotrophic Lateral Sclerosis/Frontotemporal Dementia Continuum. Front Mol Neurosci 2017; 10:119. [PMID: 28539871 PMCID: PMC5423993 DOI: 10.3389/fnmol.2017.00119] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/10/2017] [Indexed: 12/11/2022] Open
Abstract
Protein homeostasis, or proteostasis, has an important regulatory role in cellular function. Protein quality control mechanisms, including protein folding and protein degradation processes, have a crucial function in post-mitotic neurons. Cellular protein quality control relies on multiple strategies, including molecular chaperones, autophagy, the ubiquitin proteasome system, endoplasmic reticulum (ER)-associated degradation (ERAD) and the formation of stress granules (SGs), to regulate proteostasis. Neurodegenerative diseases are characterized by the presence of misfolded protein aggregates, implying that protein quality control mechanisms are dysfunctional in these conditions. Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative diseases that are now recognized to overlap clinically and pathologically, forming a continuous disease spectrum. In this review article, we detail the evidence for dysregulation of protein quality control mechanisms across the whole ALS-FTD continuum, by discussing the major proteins implicated in ALS and/or FTD. We also discuss possible ways in which protein quality mechanisms could be targeted therapeutically in these disorders and highlight promising protein quality control-based therapeutics for clinical trials.
Collapse
Affiliation(s)
- Hamideh Shahheydari
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie UniversitySydney, NSW, Australia
| | - Audrey Ragagnin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie UniversitySydney, NSW, Australia
| | - Adam K Walker
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie UniversitySydney, NSW, Australia
| | - Reka P Toth
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie UniversitySydney, NSW, Australia
| | - Marta Vidal
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie UniversitySydney, NSW, Australia
| | - Cyril J Jagaraj
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie UniversitySydney, NSW, Australia
| | - Emma R Perri
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie UniversitySydney, NSW, Australia
| | - Anna Konopka
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie UniversitySydney, NSW, Australia
| | - Jessica M Sultana
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie UniversitySydney, NSW, Australia
| | - Julie D Atkin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie UniversitySydney, NSW, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe UniversityMelbourne, VIC, Australia
| |
Collapse
|
44
|
Wei Q, Zhou Q, Chen Y, Ou R, Cao B, Xu Y, Yang J, Shang HF. Analysis of SOD1 mutations in a Chinese population with amyotrophic lateral sclerosis: a case-control study and literature review. Sci Rep 2017; 7:44606. [PMID: 28291249 PMCID: PMC5349524 DOI: 10.1038/srep44606] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/09/2017] [Indexed: 02/05/2023] Open
Abstract
Although the copper/zinc superoxide dismutase-1 (SOD1) gene has been identified in both familial ALS (FALS) and sporadic ALS (SALS), it has rarely been studied in Chinese patients with ALS, and there are few studies with large samples. This study sought to assess the prevalence of SOD1 mutations in Chinese ALS patients. We screened a cohort of 499 ALS patients (487 SALS and 12 FALS) from the Department of Neurology at the West China Hospital of Sichuan University and analyzed all coding exons of SOD1 by Sanger sequencing. In addition, we reviewed the mutation frequencies of common ALS causative genes in Chinese populations. Eight missense mutations in SOD1 were found in 8 ALS individuals: two novel mutations (p.G73D and p.V120F) and six previously reported mutations. The frequencies of SOD1 mutations were 1.03% (5/487) in SALS and 25% (3/12) in FALS from Southwest China. A literature review indicated that the mutation rates of major ALS causative genes were 53.55% in FALS and 6.29% in SALS. In Chinese SALS and FALS, the highest mutation frequency was in the SOD1 gene. Our results suggest that SOD1 mutation is the most common cause of ALS in Chinese populations and that the mutation spectrum of ALS varies among different ethnic populations.
Collapse
Affiliation(s)
- QianQian Wei
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - QingQing Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - YongPing Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - RuWei Ou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bei Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - YaQian Xu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Yang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui-Fang Shang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
45
|
Bali T, Self W, Liu J, Siddique T, Wang LH, Bird TD, Ratti E, Atassi N, Boylan KB, Glass JD, Maragakis NJ, Caress JB, McCluskey LF, Appel SH, Wymer JP, Gibson S, Zinman L, Mozaffar T, Callaghan B, McVey AL, Jockel-Balsarotti J, Allred P, Fisher ER, Lopate G, Pestronk A, Cudkowicz ME, Miller TM. Defining SOD1 ALS natural history to guide therapeutic clinical trial design. J Neurol Neurosurg Psychiatry 2017; 88:99-105. [PMID: 27261500 PMCID: PMC5136332 DOI: 10.1136/jnnp-2016-313521] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/26/2016] [Accepted: 05/11/2016] [Indexed: 12/12/2022]
Abstract
IMPORTANCE Understanding the natural history of familial amyotrophic lateral sclerosis (ALS) caused by SOD1 mutations (ALSSOD1) will provide key information for optimising clinical trials in this patient population. OBJECTIVE To establish an updated natural history of ALSSOD1. DESIGN, SETTING AND PARTICIPANTS Retrospective cohort study from 15 medical centres in North America evaluated records from 175 patients with ALS with genetically confirmed SOD1 mutations, cared for after the year 2000. MAIN OUTCOMES AND MEASURES Age of onset, survival, ALS Functional Rating Scale (ALS-FRS) scores and respiratory function were analysed. Patients with the A4V (Ala-Val) SOD1 mutation (SOD1A4V), the largest mutation population in North America with an aggressive disease progression, were distinguished from other SOD1 mutation patients (SOD1non-A4V) for analysis. RESULTS Mean age of disease onset was 49.7±12.3 years (mean±SD) for all SOD1 patients, with no statistical significance between SOD1A4V and SOD1non-A4V (p=0.72, Kruskal-Wallis). Total SOD1 patient median survival was 2.7 years. Mean disease duration for all SOD1 was 4.6±6.0 and 1.4±0.7 years for SOD1A4V. SOD1A4V survival probability (median survival 1.2 years) was significantly decreased compared with SOD1non-A4V (median survival 6.8 years; p<0.0001, log-rank). A statistically significant increase in ALS-FRS decline in SOD1A4V compared with SOD1non-A4V participants (p=0.02) was observed, as well as a statistically significant increase in ALS-forced vital capacity decline in SOD1A4V compared with SOD1non-A4V (p=0.02). CONCLUSIONS AND RELEVANCE SOD1A4V is an aggressive, but relatively homogeneous form of ALS. These SOD1-specific ALS natural history data will be important for the design and implementation of clinical trials in the ALSSOD1 patient population.
Collapse
Affiliation(s)
- Taha Bali
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Wade Self
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jingxia Liu
- Division of Public Health Sciences, Washington University School of Medicine, St Louis, Missouri, USA
| | - Teepu Siddique
- Davee Department of Neurology and Clinical Neurosciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leo H Wang
- Department of Neurology at University of Washington Medical Center, University of Washington, Seattle, Washington, USA
| | - Thomas D Bird
- Department of Neurology at University of Washington Medical Center, University of Washington, Seattle, Washington, USA.,Geriatrics Research at Seattle Veterans Affairs Medical Center, Seattle, Washington, USA
| | - Elena Ratti
- Massachusetts General Hospital, Neurology Clinical Research Institute, Boston, Massachusetts, USA
| | - Nazem Atassi
- Massachusetts General Hospital, Neurology Clinical Research Institute, Boston, Massachusetts, USA
| | - Kevin B Boylan
- Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Jonathan D Glass
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - James B Caress
- Department of Neurology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Leo F McCluskey
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stanley H Appel
- Department of Neurology, Methodist Neurological Institute, The Methodist Hospital, Houston, Texas, USA
| | - James P Wymer
- The Neurosciences Institute, Albany Medical Center, Albany, New York, USA
| | - Summer Gibson
- Department of Neurology, University of Utah, Salt Lake City, Utah, USA
| | - Lorne Zinman
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Tahseen Mozaffar
- Department of Neurology, University of California, Irvine, California, USA
| | - Brian Callaghan
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - April L McVey
- Department of Neurology, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | - Peggy Allred
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Elena R Fisher
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Glenn Lopate
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Alan Pestronk
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Merit E Cudkowicz
- Massachusetts General Hospital, Neurology Clinical Research Institute, Boston, Massachusetts, USA
| | - Timothy M Miller
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
46
|
Abstract
Amyotrophic lateral sclerosis (ALS) is a highly heterogeneous disease with no effective treatment. Drug development has been hampered by the lack of biomarkers that aid in early diagnosis, demonstrate target engagement, monitor disease progression, and can serve as surrogate endpoints to assess the efficacy of treatments. Fluid-based biomarkers may potentially address these issues. An ideal biomarker should exhibit high specificity and sensitivity for distinguishing ALS from control (appropriate disease mimics and other neurologic diseases) populations and monitor disease progression within individual patients. Significant progress has been made using cerebrospinal fluid, serum, and plasma in the search for ALS biomarkers, with urine and saliva biomarkers still in earlier stages of development. A few of these biomarker candidates have demonstrated use in patient stratification, predicting disease course (fast vs slow progression) and severity, or have been used in preclinical and clinical applications. However, while ALS biomarker discovery has seen tremendous advancements in the last decade, validating biomarkers and moving them towards the clinic remains more elusive. In this review, we highlight biomarkers that are moving towards clinical utility and the challenges that remain in order to implement biomarkers at all stages of the ALS drug development process.
Collapse
Affiliation(s)
- Lucas T Vu
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 West Thomas Road, Phoenix, AZ, 85013, USA
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 West Thomas Road, Phoenix, AZ, 85013, USA
| | - Robert Bowser
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 West Thomas Road, Phoenix, AZ, 85013, USA.
- Department of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 West Thomas Road, Phoenix, AZ, 85013, USA.
| |
Collapse
|
47
|
Motor neurons derived from ALS-related mouse iPS cells recapitulate pathological features of ALS. Exp Mol Med 2016; 48:e276. [PMID: 27932790 PMCID: PMC5192071 DOI: 10.1038/emm.2016.113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/06/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a late-onset progressive neurodegenerative disease characterized by the loss of motor neurons in the spinal cord and brain. Mutations in Cu/Zn superoxide dismutase 1 (SOD1) are known to induce ALS. Although many research models have been developed, the exact pathological mechanism of ALS remains unknown. The recently developed induced pluripotent stem (iPS) cell technology is expected to illuminate the pathological mechanisms and new means of treatment for neurodegenerative diseases. To determine the pathological mechanism of ALS, we generated mouse iPS (miPS) cells from experimental ALS transgenic mice and control mice and characterized the cells using molecular biological methods. The generated miPS cells expressed many pluripotent genes and differentiated into three germ layers in vitro and in vivo. Motor neurons derived from ALS-related miPS cells recapitulated the pathological features of ALS. The ALS-model motor neurons showed SOD1 aggregates, as well as decreased cell survival rate and neurite length compared with wild-type motor neurons. Our study will be helpful in revealing the mechanism of motor neuronal cell death in ALS.
Collapse
|
48
|
Screening of SOD1, FUS and TARDBP genes in patients with amyotrophic lateral sclerosis in central-southern China. Sci Rep 2016; 6:32478. [PMID: 27604643 PMCID: PMC5015023 DOI: 10.1038/srep32478] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/08/2016] [Indexed: 01/26/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting motor neurons of the brain, brainstem and spinal cord. To date, mutations in more than 30 genes have been linked to the pathogenesis of ALS. Among them, SOD1, FUS and TARDBP are ranked as the three most common genes associated with ALS. However, no mutation analysis has been reported in central-southern China. In this study, we sequenced SOD1, FUS and TARDBP in a central-southern Chinese cohort of 173 patients with ALS (15 familial ALS and 158 sporadic ALS) to detect mutations. As a result, five missense mutations in SOD1, namely, p.D101N, p.D101G, p.C111Y, p.N86S and p.V87A, were identified in three unrelated familial probands and three sporadic cases; two mutations in FUS were found in two unrelated familial probands, including an insertion mutation (p.P525_Y526insY) and a missense mutation (p.R521H); no variants of TARDBP were observed in patients. Therefore, SOD1 mutations were present in 20.0% of familial ALS patients and 1.9% of sporadic ALS patients, while FUS mutations were responsible for 13.3% of familial ALS cases, and TARDBP mutations were rare in either familial or sporadic ALS cases. This study broadens the known mutational spectrum in patients with ALS and further demonstrates the necessity for genetic screening in ALS patients from central-southern China.
Collapse
|
49
|
Nithya K, Angeline T, Isabel W, Asirvatham AJ. SOD1 Gene +35A/C (exon3/intron3) Polymorphism in Type 2 Diabetes Mellitus among South Indian Population. GENETICS RESEARCH INTERNATIONAL 2016; 2016:3787268. [PMID: 27190652 PMCID: PMC4852105 DOI: 10.1155/2016/3787268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/17/2016] [Indexed: 02/02/2023]
Abstract
Superoxide dismutase is an antioxidant enzyme that is involved in defence mechanisms against oxidative stress. Cu/Zn SOD is a variant that is located in exon3/intron3 boundary. The aim of the present study was to investigate whether the Cu/Zn SOD (+35A/C) gene polymorphism is associated with the susceptibility to type 2 diabetes mellitus among south Indian population. The study included patients with type 2 diabetes mellitus (n = 100) and healthy controls (n = 75). DNA was isolated from the blood and genotyping of Cu/Zn SOD gene polymorphism was done by polymerase chain reaction based restriction fragment length polymorphism method. Occurrence of different genotypes and normal (A) and mutant (C) allele frequencies were determined. The frequency of the three genotypes of the total subjects was as follows: homozygous wild-type A/A (95%), heterozygous genotype A/C (3%), and homozygous mutant C/C (2%). The mutant (C) allele and the mutant genotypes (AC/CC) were found to be completely absent among the patients with type 2 diabetes mellitus. Absence of mutant genotype (CC) shows that the Cu/Zn SOD gene polymorphism may not be associated with the susceptibility to type 2 diabetes mellitus among south Indian population.
Collapse
Affiliation(s)
- K. Nithya
- PG & Research Department of Zoology & Biotechnology, Lady Doak College, Madurai, Tamil Nadu 625 002, India
| | - T. Angeline
- PG & Research Department of Zoology & Biotechnology, Lady Doak College, Madurai, Tamil Nadu 625 002, India
| | - W. Isabel
- PG & Research Department of Zoology & Biotechnology, Lady Doak College, Madurai, Tamil Nadu 625 002, India
| | | |
Collapse
|
50
|
Alemasov NA, Ivanisenko NV, Medvedev SP, Zakian SM, Kolchanov NA, Ivanisenko VA. Dynamic properties of SOD1 mutants can predict survival time of patients carrying familial amyotrophic lateral sclerosis. J Biomol Struct Dyn 2016; 35:645-656. [DOI: 10.1080/07391102.2016.1158666] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Nikolay A. Alemasov
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nikita V. Ivanisenko
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Laboratory of Molecular Epidemiology and Bioinformatics, Novosibirsk State University, Novosibirsk, Russia
| | - Sergey P. Medvedev
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Suren M. Zakian
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nikolay A. Kolchanov
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Vladimir A. Ivanisenko
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|