1
|
Alzahrani AK, Imran M, Alshrari AS. Investigating the impact of SOD1 mutations on amyotrophic lateral sclerosis progression and potential drug repurposing through in silico analysis. J Biomol Struct Dyn 2024:1-16. [PMID: 39673548 DOI: 10.1080/07391102.2024.2439577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 05/29/2024] [Indexed: 12/16/2024]
Abstract
Superoxide dismutase 1 (SOD1) is a vital enzyme responsible for attenuating oxidative stress through its ability to facilitate the dismutation of the superoxide radical into oxygen and hydrogen peroxide. The progressive loss of motor neurons characterize amyotrophic lateral sclerosis (ALS), a crippling neurodegenerative disease that is caused by mutations in the SOD1 gene. In this study, in silico mutational analysis was performed to study the various mutations, the pathogenicity and stability ΔΔG (binding free energy) of the variant of SOD1. x in the protein variant analysis showed a considerable destabilizing effect with a ΔΔG value of -4.2 kcal/mol, signifying a notable impact on protein stability. Molecular dynamics simulations were conducted on both wild-type and C146R mutant SOD1. RMSD profiles indicated that both maintained consistent structural conformation over time. Additionally, virtual screening of 3067 FDA-approved drugs against the mutant SOD1 identified two potential binders, Tucatinib (51039094) and Regorafenib (11167602), which interacted with Leu106, similar to the control drug, Ebselen. Further simulations assessed the dynamic properties of SOD1 in monomeric and dimeric forms while bound to these compounds. 11167602 maintained stable interaction with the monomeric SOD1 mutant, whereas 51039094 and Ebselen dissociated from the monomeric protein's binding site. However, all three compounds were stably bound to the dimeric SOD1. MM/GBSA analysis revealed similar negative binding free energies for 11167602 and 51039094, identifying them as strong binders due to their interaction with Cys111. Experimental validation, including in vitro, cell-based, and in vivo assays are essential to confirm these candidates before advancing to clinical trials.
Collapse
Affiliation(s)
- A Khuzaim Alzahrani
- Department of Medical Laboratory Technology, Faculty of Medical Applied Science, Northern Border University, Arar, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Ahmed S Alshrari
- Department of Medical Laboratory Technology, Faculty of Medical Applied Science, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
2
|
Wang LQ, Ma Y, Zhang MY, Yuan HY, Li XN, Xia W, Zhao K, Huang X, Chen J, Li D, Zou L, Wang Z, Le W, Liu C, Liang Y. Amyloid fibril structures and ferroptosis activation induced by ALS-causing SOD1 mutations. SCIENCE ADVANCES 2024; 10:eado8499. [PMID: 39475611 PMCID: PMC11524188 DOI: 10.1126/sciadv.ado8499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024]
Abstract
Over 200 genetic mutations in copper-zinc superoxide dismutase (SOD1) have been linked to amyotrophic lateral sclerosis (ALS). Among these, two ALS-causing mutants, histidine-46→arginine (H46R) and glycine-85→arginine (G85R), exhibit a decreased capacity to bind metal ions. Here, we report two cryo-electron microscopy structures of amyloid fibrils formed by H46R and G85R. These mutations lead to the formation of amyloid fibrils with unique structures distinct from those of the native fibril. The core of these fibrils features a serpentine arrangement with seven or eight β strands, secured by a hydrophobic cavity and a salt bridge between arginine-85 and aspartic acid-101 in the G85R fibril. We demonstrate that these mutant fibrils are notably more toxic and capable of promoting the aggregation of wild-type SOD1 more effectively, causing mitochondrial impairment and activating ferroptosis in cell cultures, compared to wild-type SOD1 fibrils. Our study provides insights into the structural mechanisms by which SOD1 mutants aggregate and induce cytotoxicity in ALS.
Collapse
Affiliation(s)
- Li-Qiang Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Yeyang Ma
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mu-Ya Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Han-Ye Yuan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Xiang-Ning Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kun Zhao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xi Huang
- Department of Neurology, Shenzhen People’s Hospital (the First Affiliated Hospital of Southern University of Science and Technology), the Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Jie Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
- Wuhan University Shenzhen Research Institute, Shenzhen 518057, China
| | - Dan Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200030, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Liangyu Zou
- Department of Neurology, Shenzhen People’s Hospital (the First Affiliated Hospital of Southern University of Science and Technology), the Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Zhengzhi Wang
- School of Civil Engineering, Wuhan University, Wuhan 430072, China
| | - Weidong Le
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 200237, China
- Key Laboratory of Liaoning Province for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116021, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Shanghai 200032, China
| | - Yi Liang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
- Wuhan University Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
3
|
Tokuda E, Sakashita Y, Tokoro N, Date A, Kosuge Y, Miyasaka T. MS785-MS27 Reactive Misfolded/Non-Native Zn-Deficient SOD1 Species Exhibit Cytotoxicity and Adopt Heterozygous Conformations in Motor Neurons. Int J Mol Sci 2024; 25:5603. [PMID: 38891791 PMCID: PMC11171496 DOI: 10.3390/ijms25115603] [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: 04/04/2024] [Revised: 05/11/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
Misfolding of superoxide dismutase-1 (SOD1) is a pathological hallmark of amyotrophic lateral sclerosis (ALS) with SOD1 mutations. The development of antibodies specific for misfolded SOD1 deepens our understanding of how the protein participates in ALS pathogenesis. Since the term "misfolding" refers to various disordered conformers other than the natively folded one, which misfolded species are recognized by specific antibodies should be determined. Here, we molecularly characterized the recognition by MS785-MS27, an antibody cocktail experimentally confirmed to recognize over 100 ALS-linked SOD1 mutants. Indirect ELISA revealed that the antibody cocktail recognized Zn-deficient wild-type and mutated SOD1 species. It also recognized conformation-disordered wild-type and mutated SOD1 species, such as unfolded and oligomeric forms, but had less affinity for the aggregated form. Antibody-reactive SOD1 exhibited cytotoxicity to a motor neuron cell model, which was blocked by Zn treatment with Zn-deficient SOD1. Immunohistochemistry revealed antibody-reactive SOD1 mainly in spinal motor neurons of SOD1G93A mice throughout the disease course, and the distribution after symptomatic stages differed from that of other misfolded SOD1 species. This suggests that misfolded/non-native SOD1 species exist as heterogeneous populations. In conclusion, MS785-MS27 recognizes various conformation-disordered SOD1 species lacking the Zn ion.
Collapse
Affiliation(s)
- Eiichi Tokuda
- Laboratory of Clinical Medicine, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi 274-8555, Chiba, Japan
| | - Yume Sakashita
- Laboratory of Clinical Medicine, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi 274-8555, Chiba, Japan
| | - Naoya Tokoro
- Laboratory of Clinical Medicine, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi 274-8555, Chiba, Japan
| | - Ayano Date
- Laboratory of Clinical Medicine, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi 274-8555, Chiba, Japan
| | - Yasuhiro Kosuge
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi 274-8555, Chiba, Japan;
| | - Tomohiro Miyasaka
- Laboratory of Physiology and Anatomy, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi 274-8555, Chiba, Japan;
| |
Collapse
|
4
|
Han J. Copper trafficking systems in cells: insights into coordination chemistry and toxicity. Dalton Trans 2023; 52:15277-15296. [PMID: 37702384 DOI: 10.1039/d3dt02166a] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Transition metal ions, such as copper, are indispensable components in the biological system. Copper ions which primarily exist in two major oxidation states Cu(I) and Cu(II) play crucial roles in various cellular processes including antioxidant defense, biosynthesis of neurotransmitters, and energy metabolism, owing to their inherent redox activity. The disturbance in copper homeostasis can contribute to the development of copper metabolism disorders, cancer, and neurodegenerative diseases, highlighting the significance of understanding the copper trafficking system in cellular environments. This review aims to offer a comprehensive overview of copper homeostatic machinery, with an emphasis on the coordination chemistry of copper transporters and trafficking proteins. While copper chaperones and the corresponding metalloenzymes are thoroughly discussed, we also explore the potential existence of low-molecular-mass metal complexes within cellular systems. Furthermore, we summarize the toxicity mechanisms originating from copper deficiency or accumulation, which include the dysregulation of oxidative stress, signaling pathways, signal transduction, and amyloidosis. This perspective review delves into the current knowledge regarding the intricate aspects of the copper trafficking system, providing valuable insights into potential treatment strategies from the standpoint of bioinorganic chemistry.
Collapse
Affiliation(s)
- Jiyeon Han
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea.
| |
Collapse
|
5
|
Morichon L, Hirtz C, Tiers L, Mezghrani A, Raoul C, Esselin F, La Cruz ED, Julien JP, Camu W, Lehmann S. Ultrasensitive digital immunoassays for SOD1 conformation in amyotrophic lateral sclerosis. Bioanalysis 2023; 15:927-936. [PMID: 37650499 DOI: 10.4155/bio-2023-0103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
Aim: The aim of this study was to detect misfolded Cu/Zn SOD1 as a potential biomarker for amyotrophic lateral sclerosis (ALS). Materials & methods: Two ultrasensitive immunodetection assays were developed for the quantification of total and misfolded SOD1. Results: The detection of total and misfolded SOD1 was possible in human serum and cerebrospinal fluid. Total SOD1 was increased in cerebrospinal fluid from ALS patients. Misfolded SOD1 had low and variable expression in both control and ALS patient samples. Conclusion: These assays hold promise for improving our understanding of ALS and its detection, and could lead to more effective treatment options in the future. Further studies in larger cohorts are now required.
Collapse
Affiliation(s)
- Lisa Morichon
- LBPC-PPC, University of Montpellier, CHU Montpellier, INM INSERM, Montpellier, 34295, France
| | - Christophe Hirtz
- LBPC-PPC, University of Montpellier, CHU Montpellier, INM INSERM, Montpellier, 34295, France
| | - Laurent Tiers
- LBPC-PPC, University of Montpellier, CHU Montpellier, INM INSERM, Montpellier, 34295, France
| | | | - Cédric Raoul
- INM, University of Montpellier, INSERM, Montpellier, 34295, France
| | - Florence Esselin
- Department of Neurology, University of Montpellier, CHU Montpellier, INM, INSERM, Montpellier, 34295, France
| | - Elisa De La Cruz
- Department of Neurology, University of Montpellier, CHU Montpellier, INM, INSERM, Montpellier, 34295, France
| | - Jean-Pierre Julien
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, University Laval, Quebec City, 23027, Canada
| | - William Camu
- Department of Neurology, University of Montpellier, CHU Montpellier, INM, INSERM, Montpellier, 34295, France
| | - Sylvain Lehmann
- LBPC-PPC, University of Montpellier, CHU Montpellier, INM INSERM, Montpellier, 34295, France
- INM, University of Montpellier, INSERM, Montpellier, 34295, France
| |
Collapse
|
6
|
Park JH, Nordström U, Tsiakas K, Keskin I, Elpers C, Mannil M, Heller R, Nolan M, Alburaiky S, Zetterström P, Hempel M, Schara-Schmidt U, Biskup S, Steinacker P, Otto M, Weishaupt J, Hahn A, Santer R, Marquardt T, Marklund SL, Andersen PM. The motor system is exceptionally vulnerable to absence of the ubiquitously expressed superoxide dismutase-1. Brain Commun 2023; 5:fcad017. [PMID: 36793789 PMCID: PMC9924500 DOI: 10.1093/braincomms/fcad017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/21/2022] [Accepted: 01/24/2023] [Indexed: 01/30/2023] Open
Abstract
Superoxide dismutase-1 is a ubiquitously expressed antioxidant enzyme. Mutations in SOD1 can cause amyotrophic lateral sclerosis, probably via a toxic gain-of-function involving protein aggregation and prion-like mechanisms. Recently, homozygosity for loss-of-function mutations in SOD1 has been reported in patients presenting with infantile-onset motor neuron disease. We explored the bodily effects of superoxide dismutase-1 enzymatic deficiency in eight children homozygous for the p.C112Wfs*11 truncating mutation. In addition to physical and imaging examinations, we collected blood, urine and skin fibroblast samples. We used a comprehensive panel of clinically established analyses to assess organ function and analysed oxidative stress markers, antioxidant compounds, and the characteristics of the mutant Superoxide dismutase-1. From around 8 months of age, all patients exhibited progressive signs of both upper and lower motor neuron dysfunction, cerebellar, brain stem, and frontal lobe atrophy and elevated plasma neurofilament concentration indicating ongoing axonal damage. The disease progression seemed to slow down over the following years. The p.C112Wfs*11 gene product is unstable, rapidly degraded and no aggregates were found in fibroblast. Most laboratory tests indicated normal organ integrity and only a few modest deviations were found. The patients displayed anaemia with shortened survival of erythrocytes containing decreased levels of reduced glutathione. A variety of other antioxidants and oxidant damage markers were within normal range. In conclusion, non-neuronal organs in humans show a remarkable tolerance to absence of Superoxide dismutase-1 enzymatic activity. The study highlights the enigmatic specific vulnerability of the motor system to both gain-of-function mutations in SOD1 and loss of the enzyme as in the here depicted infantile superoxide dismutase-1 deficiency syndrome.
Collapse
Affiliation(s)
- Julien H Park
- Department of Clinical Sciences, Neurosciences, Umeå University, 901 87 Umeå, Sweden,Department of General Paediatrics, University of Münster, 48149 Münster, Germany
| | - Ulrika Nordström
- Department of Clinical Sciences, Neurosciences, Umeå University, 901 87 Umeå, Sweden
| | - Konstantinos Tsiakas
- Department of Paediatrics, University Medical Centre Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Isil Keskin
- Department of Medical Biosciences, Pathology, Umeå University, 901 85 Umeå, Sweden
| | - Christiane Elpers
- Department of General Paediatrics, University of Münster, 48149 Münster, Germany
| | - Manoj Mannil
- Clinic for Radiology, University Hospital Münster, WWU University of Münster, 48149 Münster, Germany
| | - Raoul Heller
- Starship Children’s Health, Auckland City Hospital, Auckland 1142, New Zealand
| | - Melinda Nolan
- Starship Children’s Health, Auckland City Hospital, Auckland 1142, New Zealand
| | - Salam Alburaiky
- Starship Children’s Health, Auckland City Hospital, Auckland 1142, New Zealand
| | - Per Zetterström
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, 901 87 Umeå, Sweden
| | - Maja Hempel
- Department of Paediatrics, University Medical Centre Hamburg-Eppendorf, 20251 Hamburg, Germany,Current address: Institute of Human Genetics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | | | - Saskia Biskup
- CeGAT GmbH and Praxis für Humangenetik Tübingen, 72076 Tübingen, Germany
| | - Petra Steinacker
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Markus Otto
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Jochen Weishaupt
- Division for Neurodegenerative Diseases, Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Andreas Hahn
- Department of Child Neurology, Justus Liebig University, 35392 Giessen, Germany
| | - René Santer
- Department of Paediatrics, University Medical Centre Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Thorsten Marquardt
- Department of General Paediatrics, University of Münster, 48149 Münster, Germany
| | - Stefan L Marklund
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, 901 87 Umeå, Sweden
| | - Peter M Andersen
- Correspondence to: Peter Munch Andersen Department of Clinical Science, Neurosciences Umeå University, SE-901 85 Umeå, Sweden E-mail:
| |
Collapse
|
7
|
Nordström U, Lang L, Ekhtiari Bidhendi E, Zetterström P, Oliveberg M, Danielsson J, Andersen PM, Marklund SL. Mutant SOD1 aggregates formed in vitro and in cultured cells are polymorphic and differ from those arising in the CNS. J Neurochem 2023; 164:77-93. [PMID: 36326589 PMCID: PMC10099669 DOI: 10.1111/jnc.15718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Mutations in the human Superoxide dismutase 1 (hSOD1) gene are well-established cause of the motor neuron disease ALS. Patients and transgenic (Tg) ALS model mice carrying mutant variants develop hSOD1 aggregates in the CNS. We have identified two hSOD1 aggregate strains, which both transmit spreading template-directed aggregation and premature fatal paralysis when inoculated into adult transgenic mice. This prion-like spread of aggregation could be a primary disease mechanism in SOD1-induced ALS. Human SOD1 aggregation has been studied extensively both in cultured cells and under various conditions in vitro. To determine how the structure of aggregates formed in these model systems related to disease-associated aggregates in the CNS, we used a binary epitope-mapping assay to examine aggregates of hSOD1 variants G93A, G85R, A4V, D90A, and G127X formed in vitro, in four different cell lines and in the CNS of Tg mice. We found considerable variability between replicate sets of in vitro-generated aggregates. In contrast, there was a high similarity between replicates of a given hSOD1 mutant in a given cell line, but pronounced variations between different hSOD1 mutants and different cell lines in both structures and amounts of aggregates formed. The aggregates formed in vitro or in cultured cells did not replicate the aggregate strains that arise in the CNS. Our findings suggest that the distinct aggregate morphologies in the CNS could result from a micro-environment with stringent quality control combined with second-order selection by spreading ability. Explorations of pathogenesis and development of therapeutics should be conducted in models that replicate aggregate structures forming in the CNS.
Collapse
Affiliation(s)
- Ulrika Nordström
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Lisa Lang
- Department of Biochemistry and Biophysics, Arrhenius Laboratories of Natural Sciences, Stockholm University, Stockholm, Sweden
| | - Elaheh Ekhtiari Bidhendi
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden.,Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Per Zetterström
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Mikael Oliveberg
- Department of Biochemistry and Biophysics, Arrhenius Laboratories of Natural Sciences, Stockholm University, Stockholm, Sweden
| | - Jens Danielsson
- Department of Biochemistry and Biophysics, Arrhenius Laboratories of Natural Sciences, Stockholm University, Stockholm, Sweden
| | - Peter M Andersen
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Stefan L Marklund
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| |
Collapse
|
8
|
Garg P, Semmler S, Baudouin C, Velde CV, Plotkin SS. Misfolding-Associated Exposure of Natively Buried Residues in Mutant SOD1 Facilitates Binding to TRAF6. J Mol Biol 2022; 434:167697. [PMID: 35753527 DOI: 10.1016/j.jmb.2022.167697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/14/2022] [Accepted: 06/18/2022] [Indexed: 10/17/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease primarily impacting motor neurons. Mutations in superoxide dismutase 1 (SOD1) are the second most common cause of familial ALS. Several of these mutations lead to misfolding or toxic gain of function in the SOD1 protein. Recently, we reported that misfolded SOD1 interacts with TNF receptor-associated factor 6 (TRAF6) in the SOD1G93A rat model of ALS. Further, we showed in cultured cells that several mutant SOD1 proteins, but not wildtype SOD1 protein, interact with TRAF6 via the MATH domain. Here, we sought to uncover the structural details of this interaction through molecular dynamics (MD) simulations of a dimeric model system, coarse grained using the AWSEM force field. We used direct MD simulations to identify buried residues, and predict binding poses by clustering frames from the trajectories. Metadynamics simulations were also used to deduce preferred binding regions on the protein surfaces from the potential of the mean force in orientation space. Well-folded SOD1 was found to bind TRAF6 via co-option of its native homodimer interface. However, if loops IV and VII of SOD1 were disordered, as typically occurs in the absence of stabilizing Zn2+ ion binding, these disordered loops now participated in novel interactions with TRAF6. On TRAF6, multiple interaction hot-spots were distributed around the equatorial region of the MATH domain beta barrel. Expression of TRAF6 variants with mutations in this region in cultured cells demonstrated that TRAF6T475 facilitates interaction with different SOD1 mutants. These findings contribute to our understanding of the disease mechanism and uncover potential targets for the development of therapeutics.
Collapse
Affiliation(s)
- Pranav Garg
- Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Sabrina Semmler
- Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec H3A 2B4, Canada; Centre de Recherche du Centre Hospitalier de Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Charlotte Baudouin
- Centre de Recherche du Centre Hospitalier de Université de Montréal, Montréal, Quebec H2X 0A9, Canada; Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Christine Vande Velde
- Centre de Recherche du Centre Hospitalier de Université de Montréal, Montréal, Quebec H2X 0A9, Canada; Department of Neurosciences, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| | - Steven S Plotkin
- Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada; Genome Sciences and Technology Program, University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada.
| |
Collapse
|
9
|
Wang LQ, Ma Y, Yuan HY, Zhao K, Zhang MY, Wang Q, Huang X, Xu WC, Dai B, Chen J, Li D, Zhang D, Wang Z, Zou L, Yin P, Liu C, Liang Y. Cryo-EM structure of an amyloid fibril formed by full-length human SOD1 reveals its conformational conversion. Nat Commun 2022; 13:3491. [PMID: 35715417 PMCID: PMC9205981 DOI: 10.1038/s41467-022-31240-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 06/09/2022] [Indexed: 11/23/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease. Misfolded Cu, Zn-superoxide dismutase (SOD1) has been linked to both familial and sporadic ALS. SOD1 fibrils formed in vitro share toxic properties with ALS inclusions. Here we produced cytotoxic amyloid fibrils from full-length apo human SOD1 under reducing conditions and determined the atomic structure using cryo-EM. The SOD1 fibril consists of a single protofilament with a left-handed helix. The fibril core exhibits a serpentine fold comprising N-terminal segment (residues 3–55) and C-terminal segment (residues 86–153) with an intrinsic disordered segment. The two segments are zipped up by three salt bridge pairs. By comparison with the structure of apo SOD1 dimer, we propose that eight β-strands (to form a β-barrel) and one α-helix in the subunit of apo SOD1 convert into thirteen β-strands stabilized by five hydrophobic cavities in the SOD1 fibril. Our data provide insights into how SOD1 converts between structurally and functionally distinct states. Misfolded SOD1 has been linked to both familial and sporadic ALS. Here the authors have determined the cryo-EM structure of SOD1 fibrils, providing insights into the conversion of SOD1 from its immature form into an aggregated form during pathogenesis of ALS.
Collapse
Affiliation(s)
- Li-Qiang Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, China.,Wuhan University Shenzhen Research Institute, 518057, Shenzhen, China
| | - Yeyang Ma
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Han-Ye Yuan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, China.,Wuhan University Shenzhen Research Institute, 518057, Shenzhen, China
| | - Kun Zhao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Mu-Ya Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, China.,Wuhan University Shenzhen Research Institute, 518057, Shenzhen, China
| | - Qiang Wang
- National Key Laboratory of Crop Genetic Improvement and National Centre of Plant Gene Research, Huazhong Agricultural University, 430070, Wuhan, China
| | - Xi Huang
- Department of Neurology, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), 518020, Shenzhen, China
| | - Wen-Chang Xu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Bin Dai
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, China.,Wuhan University Shenzhen Research Institute, 518057, Shenzhen, China
| | - Dan Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Bio-X Institutes, Shanghai Jiao Tong University, 200030, Shanghai, China.,Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Delin Zhang
- National Key Laboratory of Crop Genetic Improvement and National Centre of Plant Gene Research, Huazhong Agricultural University, 430070, Wuhan, China
| | - Zhengzhi Wang
- School of Civil Engineering, Wuhan University, 430072, Wuhan, China
| | - Liangyu Zou
- Department of Neurology, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), 518020, Shenzhen, China
| | - Ping Yin
- National Key Laboratory of Crop Genetic Improvement and National Centre of Plant Gene Research, Huazhong Agricultural University, 430070, Wuhan, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201210, Shanghai, China.
| | - Yi Liang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, 430072, Wuhan, China. .,Wuhan University Shenzhen Research Institute, 518057, Shenzhen, China.
| |
Collapse
|
10
|
Furukawa Y. A pathological link between dysregulated copper binding in Cu/Zn-superoxide dismutase and amyotrophic lateral sclerosis. J Clin Biochem Nutr 2022; 71:73-77. [PMID: 36213785 PMCID: PMC9519421 DOI: 10.3164/jcbn.22-42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022] Open
Abstract
Mutations in the gene coding Cu/Zn-superoxide dismutase (SOD1) are linked to a familial form of amyotrophic lateral sclerosis (ALS), and its pathological hallmark includes abnormal accumulation of mutant SOD1 proteins in spinal motorneurons. Mutant SOD1 proteins are considered to be susceptible to misfolding, resulting in the accumulation as oligomers/aggregates. While it remains obscure how and why SOD1 becomes misfolded under pathological conditions in vivo, the failure to bind a copper and zinc ion in SOD1 in vitro leads to the significant destabilization of its natively folded structure. Therefore, genetic and pharmacological attempts to promote the metal binding in mutant SOD1 could serve as an effective treatment of ALS. Here, I briefly review the copper and zinc binding process of SOD1 in vivo and discuss a copper chaperone for SOD1 as a potential target for developing ALS therapeutics.
Collapse
|
11
|
Hydrogen Peroxide and Amyotrophic Lateral Sclerosis: From Biochemistry to Pathophysiology. Antioxidants (Basel) 2021; 11:antiox11010052. [PMID: 35052556 PMCID: PMC8773294 DOI: 10.3390/antiox11010052] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/18/2021] [Accepted: 12/24/2021] [Indexed: 11/19/2022] Open
Abstract
Free radicals are unstable chemical reactive species produced during Redox dyshomeostasis (RDH) inside living cells and are implicated in the pathogenesis of various neurodegenerative diseases. One of the most complicated and life-threatening motor neurodegenerative diseases (MND) is amyotrophic lateral sclerosis (ALS) because of the poor understanding of its pathophysiology and absence of an effective treatment for its cure. During the last 25 years, researchers around the globe have focused their interest on copper/zinc superoxide dismutase (Cu/Zn SOD, SOD1) protein after the landmark discovery of mutant SOD1 (mSOD1) gene as a risk factor for ALS. Substantial evidence suggests that toxic gain of function due to redox disturbance caused by reactive oxygen species (ROS) changes the biophysical properties of native SOD1 protein thus, instigating its fibrillization and misfolding. These abnormal misfolding aggregates or inclusions of SOD1 play a role in the pathogenesis of both forms of ALS, i.e., Sporadic ALS (sALS) and familial ALS (fALS). However, what leads to a decrease in the stability and misfolding of SOD1 is still in question and our scientific knowledge is scarce. A large number of studies have been conducted in this area to explore the biochemical mechanistic pathway of SOD1 aggregation. Several studies, over the past two decades, have shown that the SOD1-catalyzed biochemical reaction product hydrogen peroxide (H2O2) at a pathological concentration act as a substrate to trigger the misfolding trajectories and toxicity of SOD1 in the pathogenesis of ALS. These toxic aggregates of SOD1 also cause aberrant localization of TAR-DNA binding protein 43 (TDP-43), which is characteristic of neuronal cytoplasmic inclusions (NCI) found in ALS. Here in this review, we present the evidence implicating the pivotal role of H2O2 in modulating the toxicity of SOD1 in the pathophysiology of the incurable and highly complex disease ALS. Also, highlighting the role of H2O2 in ALS, we believe will encourage scientists to target pathological concentrations of H2O2 thereby halting the misfolding of SOD1.
Collapse
|
12
|
Ayers JI, Xu G, Dillon K, Lu Q, Chen Z, Beckman J, Moreno-Romero AK, Zamora DL, Galaleldeen A, Borchelt DR. Variation in the vulnerability of mice expressing human superoxide dismutase 1 to prion-like seeding: a study of the influence of primary amino acid sequence. Acta Neuropathol Commun 2021; 9:92. [PMID: 34016165 PMCID: PMC8139116 DOI: 10.1186/s40478-021-01191-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/03/2021] [Indexed: 11/24/2022] Open
Abstract
Misfolded forms of superoxide dismutase 1 (SOD1) with mutations associated with familial amyotrophic lateral sclerosis (fALS) exhibit prion characteristics, including the ability to act as seeds to accelerate motor neuron disease in mouse models. A key feature of infectious prion seeding is that the efficiency of transmission is governed by the primary sequence of prion protein (PrP). Isologous seeding, where the sequence of the PrP in the seed matches that of the host, is generally much more efficient than when there is a sequence mis-match. Here, we used paradigms in which mutant SOD1 seeding homogenates were injected intraspinally in newborn mice or into the sciatic nerve of adult mice, to assess the influence of SOD1 primary sequence on seeding efficiency. We observed a spectrum of seeding efficiencies depending upon both the SOD1 expressed by mice injected with seeds and the origin of the seed preparations. Mice expressing WT human SOD1 or the disease variant G37R were resistant to isologous seeding. Mice expressing G93A SOD1 were also largely resistant to isologous seeding, with limited success in one line of mice that express at low levels. By contrast, mice expressing human G85R-SOD1 were highly susceptible to isologous seeding but resistant to heterologous seeding by homogenates from paralyzed mice over-expressing mouse SOD1-G86R. In other seeding experiments with G85R SOD1:YFP mice, we observed that homogenates from paralyzed animals expressing the H46R or G37R variants of human SOD1 were less effective than seeds prepared from mice expressing the human G93A variant. These sequence mis-match effects were less pronounced when we used purified recombinant SOD1 that had been fibrilized in vitro as the seeding preparation. Collectively, our findings demonstrate diversity in the abilities of ALS variants of SOD1 to initiate or sustain prion-like propagation of misfolded conformations that produce motor neuron disease.
Collapse
|
13
|
A Metal-Free, Disulfide Oxidized Form of Superoxide Dismutase 1 as a Primary Misfolded Species with Prion-Like Properties in the Extracellular Environments Surrounding Motor Neuron-Like Cells. Int J Mol Sci 2021; 22:ijms22084155. [PMID: 33923808 PMCID: PMC8074096 DOI: 10.3390/ijms22084155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 11/16/2022] Open
Abstract
Superoxide dismutase 1 (SOD1) is a metalloenzyme with high structural stability, but a lack of Cu and Zn ions decreases its stability and enhances the likelihood of misfolding, which is a pathological hallmark of amyotrophic lateral sclerosis (ALS). A growing body of evidence has demonstrated that misfolded SOD1 has prion-like properties such as transmissibility between cells and intracellular propagation of misfolding of natively folded SOD1. Recently, we found that SOD1 is misfolded in the cerebrospinal fluid of sporadic ALS patients, providing a route by which misfolded SOD1 spreads via the extracellular environment of the central nervous system. Unlike intracellular misfolded SOD1, it is unknown which extracellular misfolded species is most relevant to prion-like properties. Here, we determined a conformational feature of extracellular misfolded SOD1 that is linked to prion-like properties. Using culture media from motor neuron-like cells, NSC-34, extracellular misfolded wild-type, and four ALS-causing SOD1 mutants were characterized as a metal-free, disulfide oxidized form of SOD1 (apo-SOD1S-S). Extracellular misfolded apo-SOD1S-S exhibited cell-to-cell transmission from the culture medium to recipient cells as well as intracellular propagation of SOD1 misfolding in recipient cells. Furthermore, culture medium containing misfolded apo-SOD1S-S exerted cytotoxicity to motor neuron-like cells, which was blocked by removal of misfolded apo-SOD1S-S from the medium. We conclude that misfolded apo-SOD1S-S is a primary extracellular species that is linked to prion-like properties.
Collapse
|
14
|
Beitchman JA, Lifshitz J, Harris NG, Thomas TC, Lafrenaye AD, Hånell A, Dixon CE, Povlishock JT, Rowe RK. Spatial Distribution of Neuropathology and Neuroinflammation Elucidate the Biomechanics of Fluid Percussion Injury. Neurotrauma Rep 2021; 2:59-75. [PMID: 34223546 PMCID: PMC8240834 DOI: 10.1089/neur.2020.0046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Diffuse brain injury is better described as multi-focal, where pathology can be found adjacent to seemingly uninjured neural tissue. In experimental diffuse brain injury, pathology and pathophysiology have been reported far more lateral than predicted by the impact site. We hypothesized that local thickening of the rodent skull at the temporal ridges serves to focus the intracranial mechanical forces experienced during brain injury and generate predictable pathology. We demonstrated local thickening of the skull at the temporal ridges using contour analysis on magnetic resonance imaging. After diffuse brain injury induced by midline fluid percussion injury (mFPI), pathological foci along the anterior-posterior length of cortex under the temporal ridges were evident acutely (1, 2, and 7 days) and chronically (28 days) post-injury by deposition of argyophilic reaction product. Area CA3 of the hippocampus and lateral nuclei of the thalamus showed pathological change, suggesting that mechanical forces to or from the temporal ridges shear subcortical regions. A proposed model of mFPI biomechanics suggests that injury force vectors reflect off the skull base and radiate toward the temporal ridge, thereby injuring ventral thalamus, dorsolateral hippocampus, and sensorimotor cortex. Surgically thinning the temporal ridge before injury reduced injury-induced inflammation in the sensorimotor cortex. These data build evidence for temporal ridges of the rodent skull to contribute to the observed pathology, whether by focusing extracranial forces to enter the cranium or intracranial forces to escape the cranium. Pre-clinical investigations can take advantage of the predicted pathology to explore injury mechanisms and treatment efficacy.
Collapse
Affiliation(s)
- Joshua A. Beitchman
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA
- Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona, USA
- Midwestern University, Glendale, Arizona, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA
- Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona, USA
- Arizona State University, Tempe, Arizona, USA
- Phoenix VA Health Care System, Phoenix, Arizona, USA
| | - Neil G. Harris
- UCLA Brain Injury Research Center, Department of Neurosurgery, and Intellectual Development and Disabilities Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Theresa Currier Thomas
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA
- Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona, USA
- Arizona State University, Tempe, Arizona, USA
- Phoenix VA Health Care System, Phoenix, Arizona, USA
| | | | - Anders Hånell
- Virginia Commonwealth University, Richmond, Virginia, USA
- Uppsala University Hospital, Uppsala, Sweden
| | | | | | - Rachel K. Rowe
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, USA
- Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona, USA
- Phoenix VA Health Care System, Phoenix, Arizona, USA
| |
Collapse
|
15
|
Rozas P, Pinto C, Martínez Traub F, Díaz R, Pérez V, Becerra D, Ojeda P, Ojeda J, Wright MT, Mella J, Plate L, Henríquez JP, Hetz C, Medinas DB. Protein disulfide isomerase ERp57 protects early muscle denervation in experimental ALS. Acta Neuropathol Commun 2021; 9:21. [PMID: 33541434 PMCID: PMC7863244 DOI: 10.1186/s40478-020-01116-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive fatal neurodegenerative disease that affects motoneurons. Mutations in superoxide dismutase 1 (SOD1) have been described as a causative genetic factor for ALS. Mice overexpressing ALS-linked mutant SOD1 develop ALS symptoms accompanied by histopathological alterations and protein aggregation. The protein disulfide isomerase family member ERp57 is one of the main up-regulated proteins in tissue of ALS patients and mutant SOD1 mice, whereas point mutations in ERp57 were described as possible risk factors to develop the disease. ERp57 catalyzes disulfide bond formation and isomerization in the endoplasmic reticulum (ER), constituting a central component of protein quality control mechanisms. However, the actual contribution of ERp57 to ALS pathogenesis remained to be defined. Here, we studied the consequences of overexpressing ERp57 in experimental ALS using mutant SOD1 mice. Double transgenic SOD1G93A/ERp57WT animals presented delayed deterioration of electrophysiological activity and maintained muscle innervation compared to single transgenic SOD1G93A littermates at early-symptomatic stage, along with improved motor performance without affecting survival. The overexpression of ERp57 reduced mutant SOD1 aggregation, but only at disease end-stage, dissociating its role as an anti-aggregation factor from the protection of neuromuscular junctions. Instead, proteomic analysis revealed that the neuroprotective effects of ERp57 overexpression correlated with increased levels of synaptic and actin cytoskeleton proteins in the spinal cord. Taken together, our results suggest that ERp57 operates as a disease modifier at early stages by maintaining motoneuron connectivity.
Collapse
Affiliation(s)
- Pablo Rozas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Cristina Pinto
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Francisca Martínez Traub
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Rodrigo Díaz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Viviana Pérez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Daniela Becerra
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Patricia Ojeda
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Jorge Ojeda
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Madison T Wright
- Department of Chemistry and Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Jessica Mella
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Lars Plate
- Department of Chemistry and Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Juan Pablo Henríquez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, Santiago, Chile.
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.
- Buck Institute for Research on Aging, Novato, CA, USA.
| | - Danilo B Medinas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, Santiago, Chile.
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.
| |
Collapse
|
16
|
Zhang X, Wang F, Hu Y, Chen R, Meng D, Guo L, Lv H, Guan J, Jia Y. In vivo stress granule misprocessing evidenced in a FUS knock-in ALS mouse model. Brain 2020; 143:1350-1367. [PMID: 32358598 DOI: 10.1093/brain/awaa076] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 01/10/2020] [Accepted: 02/02/2020] [Indexed: 12/13/2022] Open
Abstract
Many RNA-binding proteins, including TDP-43, FUS, and TIA1, are stress granule components, dysfunction of which causes amyotrophic lateral sclerosis (ALS). However, whether a mutant RNA-binding protein disrupts stress granule processing in vivo in pathogenesis is unknown. Here we establish a FUS ALS mutation, p.R521C, knock-in mouse model that carries impaired motor ability and late-onset motor neuron loss. In disease-susceptible neurons, stress induces mislocalization of mutant FUS into stress granules and upregulation of ubiquitin, two hallmarks of disease pathology. Additionally, stress aggravates motor performance decline in the mutant mouse. By using two-photon imaging in TIA1-EGFP transduced animals, we document more intensely TIA1-EGFP-positive granules formed hours but cleared weeks after stress challenge in neurons in the mutant cortex. Moreover, neurons with severe granule misprocessing die days after stress challenge. Therefore, we argue that stress granule misprocessing is pathogenic in ALS, and the model we provide here is sound for further disease mechanistic study.
Collapse
Affiliation(s)
- Xue Zhang
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Fengchao Wang
- Animal core facility, National Institute of Biological Sciences, Beijing, China
| | - Yi Hu
- School of Life Sciences, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Runze Chen
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Dawei Meng
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Liang Guo
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Hailong Lv
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Jisong Guan
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Yichang Jia
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| |
Collapse
|
17
|
Nikseresht S, Hilton JB, Kysenius K, Liddell JR, Crouch PJ. Copper-ATSM as a Treatment for ALS: Support from Mutant SOD1 Models and Beyond. Life (Basel) 2020; 10:E271. [PMID: 33158182 PMCID: PMC7694234 DOI: 10.3390/life10110271] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/22/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier permeant, copper-containing compound, CuII(atsm), has successfully progressed from fundamental research outcomes in the laboratory through to phase 2/3 clinical assessment in patients with the highly aggressive and fatal neurodegenerative condition of amyotrophic lateral sclerosis (ALS). The most compelling outcomes to date to indicate potential for disease-modification have come from pre-clinical studies utilising mouse models that involve transgenic expression of mutated superoxide dismutase 1 (SOD1). Mutant SOD1 mice provide a very robust mammalian model of ALS with high validity, but mutations in SOD1 account for only a small percentage of ALS cases in the clinic, with the preponderant amount of cases being sporadic and of unknown aetiology. As per other putative drugs for ALS developed and tested primarily in mutant SOD1 mice, this raises important questions about the pertinence of CuII(atsm) to broader clinical translation. This review highlights some of the challenges associated with the clinical translation of new treatment options for ALS. It then provides a brief account of pre-clinical outcomes for CuII(atsm) in SOD1 mouse models of ALS, followed by an outline of additional studies which report positive outcomes for CuII(atsm) when assessed in cell and mouse models of neurodegeneration which do not involve mutant SOD1. Clinical evidence for CuII(atsm) selectively targeting affected regions of the CNS in patients is also presented. Overall, this review summarises the existing evidence which indicates why clinical relevance of CuII(atsm) likely extends beyond the context of cases of ALS caused by mutant SOD1.
Collapse
Affiliation(s)
- Sara Nikseresht
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC 3010, Australia; (S.N.); (J.B.H.); (J.R.L.)
| | - James B.W. Hilton
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC 3010, Australia; (S.N.); (J.B.H.); (J.R.L.)
| | - Kai Kysenius
- Department of Pharmacology and Therapeutics and Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Jeffrey R. Liddell
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC 3010, Australia; (S.N.); (J.B.H.); (J.R.L.)
| | - Peter J. Crouch
- Department of Pharmacology and Therapeutics and Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3010, Australia;
| |
Collapse
|
18
|
Niu B, Mackness BC, Zitzewitz JA, Matthews CR, Gross ML. Trifluoroethanol Partially Unfolds G93A SOD1 Leading to Protein Aggregation: A Study by Native Mass Spectrometry and FPOP Protein Footprinting. Biochemistry 2020; 59:3650-3659. [PMID: 32924445 DOI: 10.1021/acs.biochem.0c00425] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Misfolding of Cu, Zn superoxide dismutase (SOD1) variants may lead to protein aggregation and ultimately amyotrophic lateral sclerosis (ALS). The mechanism and protein conformational changes during this process are complex and remain unclear. To study SOD1 variant aggregation at the molecular level and in solution, we chemically induced aggregation of a mutant variant (G93A SOD1) with trifluoroethanol (TFE) and used both native mass spectrometry (MS) to analyze the intact protein and fast photochemical oxidation of proteins (FPOP) to characterize the structural changes induced by TFE. We found partially unfolded G93A SOD1 monomers prior to oligomerization and identified regions of the N-terminus, C-terminus, and strands β5, β6 accountable for the partial unfolding. We propose that exposure of hydrophobic interfaces of these unstructured regions serves as a precursor to aggregation. Our results provide a possible mechanism and molecular basis for ALS-linked SOD1 misfolding and aggregation.
Collapse
Affiliation(s)
- Ben Niu
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Brian C Mackness
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, United States
| | - Jill A Zitzewitz
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, United States
| | - C Robert Matthews
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, United States
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| |
Collapse
|
19
|
Does wild-type Cu/Zn-superoxide dismutase have pathogenic roles in amyotrophic lateral sclerosis? Transl Neurodegener 2020; 9:33. [PMID: 32811540 PMCID: PMC7437001 DOI: 10.1186/s40035-020-00209-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by adult-onset progressive degeneration of upper and lower motor neurons. Increasing numbers of genes are found to be associated with ALS; among those, the first identified gene, SOD1 coding a Cu/Zn-superoxide dismutase protein (SOD1), has been regarded as the gold standard in the research on a pathomechanism of ALS. Abnormal accumulation of misfolded SOD1 in affected spinal motor neurons has been established as a pathological hallmark of ALS caused by mutations in SOD1 (SOD1-ALS). Nonetheless, involvement of wild-type SOD1 remains quite controversial in the pathology of ALS with no SOD1 mutations (non-SOD1 ALS), which occupies more than 90% of total ALS cases. In vitro studies have revealed post-translationally controlled misfolding and aggregation of wild-type as well as of mutant SOD1 proteins; therefore, SOD1 proteins could be a therapeutic target not only in SOD1-ALS but also in more prevailing cases, non-SOD1 ALS. In order to search for evidence on misfolding and aggregation of wild-type SOD1 in vivo, we reviewed pathological studies using mouse models and patients and then summarized arguments for and against possible involvement of wild-type SOD1 in non-SOD1 ALS as well as in SOD1-ALS.
Collapse
|
20
|
Perri ER, Parakh S, Vidal M, Mehta P, Ma Y, Walker AK, Atkin JD. The Cysteine (Cys) Residues Cys-6 and Cys-111 in Mutant Superoxide Dismutase 1 (SOD1) A4V Are Required for Induction of Endoplasmic Reticulum Stress in Amyotrophic Lateral Sclerosis. J Mol Neurosci 2020; 70:1357-1368. [PMID: 32445072 DOI: 10.1007/s12031-020-01551-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/13/2020] [Indexed: 12/21/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the degeneration of motor neurons. Between 12 and 20% of inherited cases and approximately 1-2% of all cases are caused by mutations in the gene encoding dismutase 1 (SOD1). Mutant SOD1 A4V (alanine to valine) induces endoplasmic reticulum (ER) stress, which is increasingly implicated as a pathway to motor neuron degeneration and death in ALS. However, it remains unclear how ER stress is induced by mutant SOD1 A4V. Previous studies have established that it is induced early in pathophysiology and it precedes the formation of mutant SOD1 inclusions. SOD1 contains four cysteine residues, two of which form an intra-subunit disulphide bond involving Cys-57 and Cys-146. The remaining two cysteines, Cys-6 and Cys-111, remain unpaired and have been implicated in mutant SOD1 aggregation. In this study, we examined the relationship between the SOD1 A4V cysteine residues and aggregation, ER stress induction and toxicity. We report here that mutation of Cys-6 and Cys-111 in mutant SOD1 A4V, but not Cys-57 or Cys-146, ameliorates ER stress, inclusion formation and apoptosis in neuronal cell lines. These results imply that protein misfolding, induced by Cys-6 and Cys-111, is required for these pathological events in neuronal cells.
Collapse
Affiliation(s)
- Emma R Perri
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Sonam Parakh
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Marta Vidal
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Prachi Mehta
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Yi Ma
- Department of General Surgery, Monash Health, Melbourne, Victoria, Australia
| | - Adam K Walker
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia.,Neurodegeneration Pathobiology Laboratory, Queensland Brain Institute, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Julie D Atkin
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia. .,Department of General Surgery, Monash Health, Melbourne, Victoria, Australia.
| |
Collapse
|
21
|
Filipi T, Hermanova Z, Tureckova J, Vanatko O, Anderova M. Glial Cells-The Strategic Targets in Amyotrophic Lateral Sclerosis Treatment. J Clin Med 2020; 9:E261. [PMID: 31963681 PMCID: PMC7020059 DOI: 10.3390/jcm9010261] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease, which is characterized by the degeneration of motor neurons in the motor cortex and the spinal cord and subsequently by muscle atrophy. To date, numerous gene mutations have been linked to both sporadic and familial ALS, but the effort of many experimental groups to develop a suitable therapy has not, as of yet, proven successful. The original focus was on the degenerating motor neurons, when researchers tried to understand the pathological mechanisms that cause their slow death. However, it was soon discovered that ALS is a complicated and diverse pathology, where not only neurons, but also other cell types, play a crucial role via the so-called non-cell autonomous effect, which strongly deteriorates neuronal conditions. Subsequently, variable glia-based in vitro and in vivo models of ALS were established and used for brand-new experimental and clinical approaches. Such a shift towards glia soon bore its fruit in the form of several clinical studies, which more or less successfully tried to ward the unfavourable prognosis of ALS progression off. In this review, we aimed to summarize current knowledge regarding the involvement of each glial cell type in the progression of ALS, currently available treatments, and to provide an overview of diverse clinical trials covering pharmacological approaches, gene, and cell therapies.
Collapse
Affiliation(s)
- Tereza Filipi
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
- 2nd Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
- 2nd Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| | - Ondrej Vanatko
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| |
Collapse
|
22
|
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
|
23
|
Keskin I, Forsgren E, Lehmann M, Andersen PM, Brännström T, Lange DJ, Synofzik M, Nordström U, Zetterström P, Marklund SL, Gilthorpe JD. The molecular pathogenesis of superoxide dismutase 1-linked ALS is promoted by low oxygen tension. Acta Neuropathol 2019; 138:85-101. [PMID: 30863976 PMCID: PMC6570705 DOI: 10.1007/s00401-019-01986-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 12/13/2022]
Abstract
Mutations in superoxide dismutase 1 (SOD1) cause amyotrophic lateral sclerosis (ALS). Disease pathogenesis is linked to destabilization, disorder and aggregation of the SOD1 protein. However, the non-genetic factors that promote disorder and the subsequent aggregation of SOD1 have not been studied. Mainly located to the reducing cytosol, mature SOD1 contains an oxidized disulfide bond that is important for its stability. Since O2 is required for formation of the bond, we reasoned that low O2 tension might be a risk factor for the pathological changes associated with ALS development. By combining biochemical approaches in an extensive range of genetically distinct patient-derived cell lines, we show that the disulfide bond is an Achilles heel of the SOD1 protein. Culture of patient-derived fibroblasts, astrocytes, and induced pluripotent stem cell-derived mixed motor neuron and astrocyte cultures (MNACs) under low O2 tensions caused reductive bond cleavage and increases in disordered SOD1. The effects were greatest in cells derived from patients carrying ALS-linked mutations in SOD1. However, significant increases also occurred in wild-type SOD1 in cultures derived from non-disease controls, and patients carrying mutations in other common ALS-linked genes. Compared to fibroblasts, MNACs showed far greater increases in SOD1 disorder and even aggregation of mutant SOD1s, in line with the vulnerability of the motor system to SOD1-mediated neurotoxicity. Our results show for the first time that O2 tension is a principal determinant of SOD1 stability in human patient-derived cells. Furthermore, we provide a mechanism by which non-genetic risk factors for ALS, such as aging and other conditions causing reduced vascular perfusion, could promote disease initiation and progression.
Collapse
Affiliation(s)
- Isil Keskin
- Department of Medical Biosciences, Pathology, Umeå University, 90185, Umeå, Sweden
| | - Elin Forsgren
- Department of Pharmacology and Clinical Neuroscience, Umeå University, 90187, Umeå, Sweden
| | - Manuela Lehmann
- Department of Pharmacology and Clinical Neuroscience, Umeå University, 90187, Umeå, Sweden
| | - Peter M Andersen
- Department of Pharmacology and Clinical Neuroscience, Umeå University, 90187, Umeå, Sweden
| | - Thomas Brännström
- Department of Medical Biosciences, Pathology, Umeå University, 90185, Umeå, Sweden
| | - Dale J Lange
- Department of Neurology, Hospital for Special Surgery and Weill Cornell Medical Center, New York, NY, 10021, USA
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Research Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany
| | - Ulrika Nordström
- Department of Pharmacology and Clinical Neuroscience, Umeå University, 90187, Umeå, Sweden
| | - Per Zetterström
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, 90185, Umeå, Sweden
| | - Stefan L Marklund
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, 90185, Umeå, Sweden.
| | - Jonathan D Gilthorpe
- Department of Pharmacology and Clinical Neuroscience, Umeå University, 90187, Umeå, Sweden.
| |
Collapse
|
24
|
Kuo MTH, Beckman JS, Shaw CA. Neuroprotective effect of CuATSM on neurotoxin-induced motor neuron loss in an ALS mouse model. Neurobiol Dis 2019; 130:104495. [PMID: 31181282 DOI: 10.1016/j.nbd.2019.104495] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/27/2019] [Accepted: 06/05/2019] [Indexed: 02/02/2023] Open
Abstract
CuATSM is a PET-imaging agent that has recently received attention for its success in extending the lifespan in animals in several neurodegenerative disease models. In the SOD1G93A model of ALS, CuATSM prolonged mouse longevity far longer than any previously tested therapeutic agents. The mechanism underlying this outcome has not been fully understood, but studies suggest that this copper complex contributes to maintaining copper homeostasis in mitochondria. More specifically for the SOD1 model, the molecule supplies copper back to the SOD1 protein. Additionally, CuATSM demonstrated similar protective effects in various in vivo Parkinson's disease mouse models. In the current pilot study, we utilized a neurodegenerative mouse model of motor neuron degeneration induced by the neurotoxin β-sitosterol β-D-glucoside. In this model, slow but distinct and progressive features of sporadic ALS occur. Treatment with CuATSM kept animal behavioural performance on par with the controls and prevented the extensive motor neuron degeneration and microglia activation seen in the untreated animals. These outcomes support a broader neuroprotective role for CuATSM beyond mutant SOD models of ALS.
Collapse
Affiliation(s)
- Michael T H Kuo
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Joseph S Beckman
- Linus Pauling Institute, Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, United States
| | - Christopher A Shaw
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada; Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada; Program in Experimental Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
25
|
Experimental Mutations in Superoxide Dismutase 1 Provide Insight into Potential Mechanisms Involved in Aberrant Aggregation in Familial Amyotrophic Lateral Sclerosis. G3-GENES GENOMES GENETICS 2019; 9:719-728. [PMID: 30622123 PMCID: PMC6404617 DOI: 10.1534/g3.118.200787] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mutations in more than 80 different positions in superoxide dismutase 1 (SOD1) have been associated with amyotrophic lateral sclerosis (fALS). There is substantial evidence that a common consequence of these mutations is to induce the protein to misfold and aggregate. How these mutations perturb native structure to heighten the propensity to misfold and aggregate is unclear. In the present study, we have mutagenized Glu residues at positions 40 and 133 that are involved in stabilizing the β-barrel structure of the native protein and a critical Zn binding domain, respectively, to examine how specific mutations may cause SOD1 misfolding and aggregation. Mutations associated with ALS as well as experimental mutations were introduced into these positions. We used an assay in which mutant SOD1 was fused to yellow fluorescent protein (SOD1:YFP) to visualize the formation of cytosolic inclusions by mutant SOD1. We then used existing structural data on SOD1, to predict how different mutations might alter local 3D conformation. Our findings reveal an association between mutant SOD1 aggregation and amino acid substitutions that are predicted to introduce steric strain, sometimes subtly, in the 3D conformation of the peptide backbone.
Collapse
|
26
|
De Giorgio F, Maduro C, Fisher EMC, Acevedo-Arozena A. Transgenic and physiological mouse models give insights into different aspects of amyotrophic lateral sclerosis. Dis Model Mech 2019; 12:dmm037424. [PMID: 30626575 PMCID: PMC6361152 DOI: 10.1242/dmm.037424] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A wide range of genetic mouse models is available to help researchers dissect human disease mechanisms. Each type of model has its own distinctive characteristics arising from the nature of the introduced mutation, as well as from the specific changes to the gene of interest. Here, we review the current range of mouse models with mutations in genes causative for the human neurodegenerative disease amyotrophic lateral sclerosis. We focus on the two main types of available mutants: transgenic mice and those that express mutant genes at physiological levels from gene targeting or from chemical mutagenesis. We compare the phenotypes for genes in which the two classes of model exist, to illustrate what they can teach us about different aspects of the disease, noting that informative models may not necessarily mimic the full trajectory of the human condition. Transgenic models can greatly overexpress mutant or wild-type proteins, giving us insight into protein deposition mechanisms, whereas models expressing mutant genes at physiological levels may develop slowly progressing phenotypes but illustrate early-stage disease processes. Although no mouse models fully recapitulate the human condition, almost all help researchers to understand normal and abnormal biological processes, providing that the individual characteristics of each model type, and how these may affect the interpretation of the data generated from each model, are considered and appreciated.
Collapse
Affiliation(s)
- Francesca De Giorgio
- Department of Neuromuscular Diseases, UCL Institute of Neurology, and MRC Centre for Neuromuscular Disease, University College London, Queen Square, London WC1N 3BG, UK
| | - Cheryl Maduro
- Department of Neuromuscular Diseases, UCL Institute of Neurology, and MRC Centre for Neuromuscular Disease, University College London, Queen Square, London WC1N 3BG, UK
| | - Elizabeth M C Fisher
- Department of Neuromuscular Diseases, UCL Institute of Neurology, and MRC Centre for Neuromuscular Disease, University College London, Queen Square, London WC1N 3BG, UK
| | - Abraham Acevedo-Arozena
- Unidad de Investigación Hospital Universitario de Canarias, Fundación Canaria de Investigación Sanitaria and Instituto de Tecnologías Biomédicas (ITB), La Laguna, 38320 Tenerife, Spain
| |
Collapse
|
27
|
Boyd SD, Calvo JS, Liu L, Ullrich MS, Skopp A, Meloni G, Winkler DD. The yeast copper chaperone for copper-zinc superoxide dismutase (CCS1) is a multifunctional chaperone promoting all levels of SOD1 maturation. J Biol Chem 2018; 294:1956-1966. [PMID: 30530491 DOI: 10.1074/jbc.ra118.005283] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/30/2018] [Indexed: 11/06/2022] Open
Abstract
Copper (Cu) is essential for the survival of aerobic organisms through its interaction with molecular oxygen (O2). However, Cu's chemical properties also make it toxic, requiring specific cellular mechanisms for Cu uptake and handling, mediated by Cu chaperones. CCS1, the budding yeast (S. cerevisiae) Cu chaperone for Cu-zinc (Zn) superoxide dismutase (SOD1) activates by directly promoting both Cu delivery and disulfide formation in SOD1. The complete mechanistic details of this transaction along with recently proposed molecular chaperone-like functions for CCS1 remain undefined. Here, we present combined structural, spectroscopic, kinetic, and thermodynamic data that suggest a multifunctional chaperoning role(s) for CCS1 during SOD1 activation. We observed that CCS1 preferentially binds a completely immature form of SOD1 and that the SOD1·CCS1 interaction promotes high-affinity Zn(II) binding in SOD1. Conserved aromatic residues within the CCS1 C-terminal domain are integral in these processes. Previously, we have shown that CCS1 delivers Cu(I) to an entry site at the SOD1·CCS1 interface upon binding. We show here that Cu(I) is transferred from CCS1 to the entry site and then to the SOD1 active site by a thermodynamically driven affinity gradient. We also noted that efficient transfer from the entry site to the active site is entirely dependent upon the oxidation of the conserved intrasubunit disulfide bond in SOD1. Our results herein provide a solid foundation for proposing a complete molecular mechanism for CCS1 activity and reclassification as a first-of-its-kind "dual chaperone."
Collapse
Affiliation(s)
| | - Jenifer S Calvo
- Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080
| | - Li Liu
- From the Departments of Biological Sciences and
| | | | | | - Gabriele Meloni
- Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080
| | | |
Collapse
|
28
|
Maier M, Welt T, Wirth F, Montrasio F, Preisig D, McAfoose J, Vieira FG, Kulic L, Späni C, Stehle T, Perrin S, Weber M, Hock C, Nitsch RM, Grimm J. A human-derived antibody targets misfolded SOD1 and ameliorates motor symptoms in mouse models of amyotrophic lateral sclerosis. Sci Transl Med 2018; 10:10/470/eaah3924. [DOI: 10.1126/scitranslmed.aah3924] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/15/2017] [Accepted: 05/16/2018] [Indexed: 12/13/2022]
Abstract
Mutations in the gene encoding superoxide dismutase 1 (SOD1) lead to misfolding and aggregation of SOD1 and cause familial amyotrophic lateral sclerosis (FALS). However, the implications of wild-type SOD1 misfolding in sporadic forms of ALS (SALS) remain unclear. By screening human memory B cells from a large cohort of healthy elderly subjects, we generated a recombinant human monoclonal antibody (α-miSOD1) that selectively bound to misfolded SOD1, but not to physiological SOD1 dimers. On postmortem spinal cord sections from 121 patients with ALS, α-miSOD1 antibody identified misfolded SOD1 in a majority of cases, regardless of their SOD1 genotype. In contrast, the α-miSOD1 antibody did not bind to its epitope in most of the 41 postmortem spinal cord sections from non-neurological control (NNC) patients. In transgenic mice overexpressing disease-causing human SOD1G37R or SOD1G93A mutations, treatment with the α-miSOD1 antibody delayed the onset of motor symptoms, extended survival by up to 2 months, and reduced aggregation of misfolded SOD1 and motor neuron degeneration. These effects were obtained whether α-miSOD1 antibody treatment was administered by direct brain infusion or peripheral administration. These results support the further development of α-miSOD1 antibody as a candidate treatment for ALS involving misfolding of SOD1.
Collapse
|
29
|
Tryptophan 32 mediates SOD1 toxicity in a in vivo motor neuron model of ALS and is a promising target for small molecule therapeutics. Neurobiol Dis 2018; 124:297-310. [PMID: 30528257 DOI: 10.1016/j.nbd.2018.11.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 11/11/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022] Open
Abstract
SOD1 misfolding, toxic gain of function, and spread are proposed as a pathological basis of amyotrophic lateral sclerosis (ALS), but the nature of SOD1 toxicity has been difficult to elucidate. Uniquely in SOD1 proteins from humans and other primates, and rarely in other species, a tryptophan residue at position 32 (W32) is predicted to be solvent exposed and to participate in SOD1 misfolding. We hypothesized that W32 is influential in SOD1 acquiring toxicity, as it is known to be important in template-directed misfolding. We tested if W32 contributes to SOD1 cytotoxicity and if it is an appropriate drug target to ameliorate ALS-like neuromuscular deficits in a zebrafish model of motor neuron axon morphology and function (swimming). Embryos injected with human SOD1 variant with W32 substituted for a serine (SOD1W32S) had reduced motor neuron axonopathy and motor deficits compared to those injected with wildtype or disease-associated SOD1. A library of FDA-approved small molecules was ranked with virtual screening based on predicted binding to W32, and subsequently filtered for analogues using a pharmacophore model based on molecular features of the uracil moiety of a small molecule previously predicted to interact with W32 (5'-fluorouridine or 5'-FUrd). Along with testing 5'-FUrd and uridine, a lead candidate from this list was selected based on its lower toxicity and improved blood brain barrier penetrance; telbivudine significantly rescued SOD1 toxicity in a dose-dependent manner. The mechanisms whereby the small molecules ameliorated motor neuron phenotypes were specifically mediated through human SOD1 and its residue W32, because these therapeutics had no measurable impact on the effects of UBQLN4D90A, EtOH, or tryptophan-deficient human SOD1W32S. By substituting W32 for a more evolutionarily conserved residue (serine), we confirmed the significant influence of W32 on human SOD1 toxicity to motor neuron morphology and function; further, we performed pharmaceutical targeting of the W32 residue for rescuing SOD1 toxicity. This unique residue offers future novel insights into SOD1 stability and toxic gain of function, and therefore poses an potential target for drug therapy.
Collapse
|
30
|
Mutant superoxide dismutase aggregates from human spinal cord transmit amyotrophic lateral sclerosis. Acta Neuropathol 2018; 136:939-953. [PMID: 30284034 PMCID: PMC6280858 DOI: 10.1007/s00401-018-1915-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/21/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022]
Abstract
Motor neurons containing aggregates of superoxide dismutase 1 (SOD1) are hallmarks of amyotrophic lateral sclerosis (ALS) caused by mutations in the gene encoding SOD1. We have previously reported that two strains of mutant human (h) SOD1 aggregates (denoted A and B) can arise in hSOD1-transgenic models for ALS and that inoculation of such aggregates into the lumbar spinal cord of mice results in rostrally spreading, templated hSOD1 aggregation and premature fatal ALS-like disease. Here, we explored whether mutant hSOD1 aggregates with prion-like properties also exist in human ALS. Aggregate seeds were prepared from spinal cords from an ALS patient carrying the hSOD1G127Gfs*7 truncation mutation and from mice transgenic for the same mutation. To separate from mono-, di- or any oligomeric hSOD1 species, the seed preparation protocol included ultracentrifugation through a density cushion. The core structure of hSOD1G127Gfs*7 aggregates present in mice was strain A-like. Inoculation of the patient- or mouse-derived seeds into lumbar spinal cord of adult hSOD1-expressing mice induced strain A aggregation propagating along the neuraxis and premature fatal ALS-like disease (p < 0.0001). Inoculation of human or murine control seeds had no effect. The potencies of the ALS patient-derived seed preparations were high and disease was initiated in the transgenic mice by levels of hSOD1G127Gfs*7 aggregates much lower than those found in the motor system of patients carrying the mutation. The results suggest that prion-like growth and spread of hSOD1 aggregation could be the primary pathogenic mechanism, not only in hSOD1 transgenic rodent models, but also in human ALS.
Collapse
|
31
|
Yang F, Wang H, Logan DT, Mu X, Danielsson J, Oliveberg M. The Cost of Long Catalytic Loops in Folding and Stability of the ALS-Associated Protein SOD1. J Am Chem Soc 2018; 140:16570-16579. [PMID: 30359015 DOI: 10.1021/jacs.8b08141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A conspicuous feature of the amyotrophic lateral sclerosis (ALS)-associated protein SOD1 is that its maturation into a functional enzyme relies on local folding of two disordered loops into a catalytic subdomain. To drive the disorder-to-order transition, the protein employs a single Zn2+ ion. The question is then if the entropic penalty of maintaining such disordered loops in the immature apoSOD1 monomer is large enough to explain its unusually low stability, slow folding, and pathological aggregation in ALS. To find out, we determined the effects of systematically altering the SOD1-loop lengths by protein redesign. The results show that the loops destabilize the apoSOD1 monomer by ∼3 kcal/mol, rendering the protein marginally stable and accounting for its aggregation behavior. Yet the effect on the global folding kinetics remains much smaller with a transition-state destabilization of <1 kcal/mol. Notably, this 1/3 transition-state to folded-state stability ratio provides a clear-cut example of the enigmatic disagreement between the Leffler α value from loop-length alterations (typically 1/3) and the "standard" reaction coordinates based on solvent perturbations (typically >2/3). Reconciling the issue, we demonstrate that the disagreement disappears when accounting for the progressive loop shortening that occurs along the folding pathway. The approach assumes a consistent Flory loop entropy scaling factor of c = 1.48 for both equilibrium and kinetic data and has the added benefit of verifying the tertiary interactions of the folding nucleus as determined by phi-value analysis. Thus, SOD1 not only represents a case where evolution of key catalytic function has come with the drawback of a destabilized apo state but also stands out as a well-suited model system for exploring the physicochemical details of protein self-organization.
Collapse
Affiliation(s)
- Fan Yang
- Department of Biochemistry and Biophysics, Arrhenius Laboratories of Natural Sciences , Stockholm University , S-106 91 Stockholm , Sweden
| | - Huabing Wang
- Department of Biochemistry and Biophysics, Arrhenius Laboratories of Natural Sciences , Stockholm University , S-106 91 Stockholm , Sweden
| | - Derek T Logan
- Division of Biochemistry & Structural Biology, Department of Chemistry , Lund University , Box 124, 22100 Lund , Sweden
| | - Xin Mu
- Department of Biochemistry and Biophysics, Arrhenius Laboratories of Natural Sciences , Stockholm University , S-106 91 Stockholm , Sweden
| | - Jens Danielsson
- Department of Biochemistry and Biophysics, Arrhenius Laboratories of Natural Sciences , Stockholm University , S-106 91 Stockholm , Sweden
| | - Mikael Oliveberg
- Department of Biochemistry and Biophysics, Arrhenius Laboratories of Natural Sciences , Stockholm University , S-106 91 Stockholm , Sweden
| |
Collapse
|
32
|
Peng X, Cashman NR, Plotkin SS. Prediction of Misfolding-Specific Epitopes in SOD1 Using Collective Coordinates. J Phys Chem B 2018; 122:11662-11676. [PMID: 30351123 DOI: 10.1021/acs.jpcb.8b07680] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We introduce a global, collective coordinate bias into molecular dynamics simulations that partially unfolds a protein, in order to predict misfolding-specific epitopes based on the regions that locally unfold. Several metrics are used to measure local disorder, including solvent exposed surface area (SASA), native contacts ( Q), and root mean squared fluctuations (RMSF). The method is applied to Cu, Zn superoxide dismutase (SOD1). For this protein, the processes of monomerization, metal loss, and conformational unfolding due to microenvironmental stresses are all separately taken into account. Several misfolding-specific epitopes are predicted, and consensus epitopes are calculated. These predicted epitopes are consistent with the "lower-resolution" peptide sequences used to raise disease-specific antibodies, but the epitopes derived from collective coordinates contain shorter, more refined sequences for the key residues constituting the epitope.
Collapse
Affiliation(s)
- Xubiao Peng
- Department of Physics and Astronomy , University of British Columbia , Vancouver , British Columbia V6T 1Z1 , Canada.,Center for Quantum Technology Research, School of Physics , Beijing Institute of Technology , Haidian, Beijing 100081 , China
| | - Neil R Cashman
- Brain Research Centre , University of British Columbia , Vancouver , British Columbia V6T 2B5 , Canada
| | - Steven S Plotkin
- Department of Physics and Astronomy, and Genome Sciences and Technology Program , University of British Columbia , Vancouver , British Columbia V6T 1Z1 , Canada
| |
Collapse
|
33
|
Paré B, Lehmann M, Beaudin M, Nordström U, Saikali S, Julien JP, Gilthorpe JD, Marklund SL, Cashman NR, Andersen PM, Forsberg K, Dupré N, Gould P, Brännström T, Gros-Louis F. Misfolded SOD1 pathology in sporadic Amyotrophic Lateral Sclerosis. Sci Rep 2018; 8:14223. [PMID: 30242181 PMCID: PMC6155098 DOI: 10.1038/s41598-018-31773-z] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/15/2018] [Indexed: 11/30/2022] Open
Abstract
Aggregation of mutant superoxide dismutase 1 (SOD1) is a pathological hallmark of a subset of familial ALS patients. However, the possible role of misfolded wild type SOD1 in human ALS is highly debated. To ascertain whether or not misfolded SOD1 is a common pathological feature in non-SOD1 ALS, we performed a blinded histological and biochemical analysis of post mortem brain and spinal cord tissues from 19 sporadic ALS, compared with a SOD1 A4V patient as well as Alzheimer’s disease (AD) and non-neurological controls. Multiple conformation- or misfolded-specific antibodies for human SOD1 were compared. These were generated independently by different research groups and were compared using standardized conditions. Five different misSOD1 staining patterns were found consistently in tissue sections from SALS cases and the SOD1 A4V patient, but were essentially absent in AD and non-neurological controls. We have established clear experimental protocols and provide specific guidelines for working, with conformational/misfolded SOD1-specific antibodies. Adherence to these guidelines will aid in the comparison of the results of future studies and better interpretation of staining patterns. This blinded, standardized and unbiased approach provides further support for a possible pathological role of misSOD1 in SALS.
Collapse
Affiliation(s)
- Bastien Paré
- Laval University Experimental Organogenesis Research Center/LOEX, Division of Regenerative Medicine, CHU de Québec Research Center - Enfant-Jésus Hospital, Québec, Canada.,Department of Surgery, Faculty of Medicine, Laval University, Québec, Canada
| | - Manuela Lehmann
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Marie Beaudin
- Neuroscience Division of the CHU de Québec and Department of Medicine of the Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Ulrika Nordström
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Stephan Saikali
- Department of Medical Biology, Division of Anatomic Pathology and Neuropathology, CHU de Québec, Hôpital de l'Enfant-Jésus, Québec, Canada
| | - Jean-Pierre Julien
- Department of Psychiatry and Neuroscience, Laval University, Québec City, Québec, Canada.,Centre de Recherche CERVO, Québec City, Québec, Canada
| | - Jonathan D Gilthorpe
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Stefan L Marklund
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Neil R Cashman
- Department of Medicine (Neurology), Brain Research Center, University of British Columbia, Vancouver, BC, Canada
| | - Peter M Andersen
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Karin Forsberg
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Nicolas Dupré
- Neuroscience Division of the CHU de Québec and Department of Medicine of the Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Peter Gould
- Department of Medical Biology, Division of Anatomic Pathology and Neuropathology, CHU de Québec, Hôpital de l'Enfant-Jésus, Québec, Canada
| | - Thomas Brännström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - François Gros-Louis
- Laval University Experimental Organogenesis Research Center/LOEX, Division of Regenerative Medicine, CHU de Québec Research Center - Enfant-Jésus Hospital, Québec, Canada. .,Department of Surgery, Faculty of Medicine, Laval University, Québec, Canada.
| |
Collapse
|
34
|
Medinas DB, Rozas P, Martínez Traub F, Woehlbier U, Brown RH, Bosco DA, Hetz C. Endoplasmic reticulum stress leads to accumulation of wild-type SOD1 aggregates associated with sporadic amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A 2018; 115:8209-8214. [PMID: 30038021 PMCID: PMC6094144 DOI: 10.1073/pnas.1801109115] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abnormal modifications to mutant superoxide dismutase 1 (SOD1) are linked to familial amyotrophic lateral sclerosis (fALS). Misfolding of wild-type SOD1 (SOD1WT) is also observed in postmortem tissue of a subset of sporadic ALS (sALS) cases, but cellular and molecular mechanisms generating abnormal SOD1WT species are unknown. We analyzed aberrant human SOD1WT species over the lifetime of transgenic mice and found the accumulation of disulfide-cross-linked high-molecular-weight SOD1WT aggregates during aging. Subcellular fractionation of spinal cord tissue and protein overexpression in NSC-34 motoneuron-like cells revealed that endoplasmic reticulum (ER) localization favors oxidation and disulfide-dependent aggregation of SOD1WT We established a pharmacological paradigm of chronic ER stress in vivo, which recapitulated SOD1WTaggregation in young transgenic mice. These species were soluble in nondenaturing detergents and did not react with a SOD1 conformation-specific antibody. Interestingly, SOD1WT aggregation under ER stress correlated with astrocyte activation in the spinal cord of transgenic mice. Finally, the disulfide-cross-linked SOD1WT species were also found augmented in spinal cord tissue of sALS patients, correlating with the presence of ER stress markers. Overall, this study suggests that ER stress increases the susceptibility of SOD1WT to aggregate during aging, operating as a possible risk factor for developing ALS.
Collapse
Affiliation(s)
- Danilo B Medinas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile 8380453;
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile 7800003
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile 8380453
| | - Pablo Rozas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile 8380453
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile 7800003
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile 8380453
| | - Francisca Martínez Traub
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile 8380453
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile 7800003
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile 8380453
| | - Ute Woehlbier
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile 8380453
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile 8380453
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Santiago, Chile 8580745
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Daryl A Bosco
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile 8380453;
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile 7800003
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile 8380453
- Buck Institute for Research on Aging, Novato, CA 94945
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115
| |
Collapse
|
35
|
Boyd SD, Liu L, Bulla L, Winkler DD. Quantifying the Interaction between Copper-Zinc Superoxide Dismutase (Sod1) and its Copper Chaperone (Ccs1). ACTA ACUST UNITED AC 2018; 11. [PMID: 29950795 PMCID: PMC6018003 DOI: 10.4172/jpb.1000473] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Immature copper-zinc superoxide dismutase (Sod1) is activated by its copper chaperone (Ccs1). Ccs1 delivers a single copper ion and catalyzes oxidation of an intra-subunit disulfide bond within each Sod1 monomer through a mechanistically ambiguous process. Here, we use residue specific fluorescent labeling of immature Sod1 to quantitate the thermodynamics of the Sod1•Ccs1 interaction while determining a more complete view of Ccs1 function. Ccs1 preferentially binds a completely immature form of Sod1 that is metal deficient and disulfide reduced (E, E-Sod1SH). However, binding induces structural changes that promote high-affinity zinc binding by the Ccs1-bound Sod1 molecule. This adds further support to the notion that Ccs1 likely plays dual chaperoning roles during the Sod1 maturation process. Further analysis reveals that in addition to the copper-dependent roles during Sod1 activation, the N- and C-terminal domains of Ccs1 also have synergistic roles in securing both Sod1 recognition and its own active conformation. These results provide new and measurable analyses of the molecular determinants guiding Ccs1-mediated Sod1 activation.
Collapse
Affiliation(s)
- Stefanie D Boyd
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Li Liu
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Lee Bulla
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Duane D Winkler
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| |
Collapse
|
36
|
Manjula R, Wright GSA, Strange RW, Padmanabhan B. Assessment of ligand binding at a site relevant to
SOD
1 oxidation and aggregation. FEBS Lett 2018; 592:1725-1737. [DOI: 10.1002/1873-3468.13055] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Ramu Manjula
- Department of Biophysics National Institute of Mental Health and Neurosciences (NIMHANS) Bangalore India
| | - Gareth S. A. Wright
- Molecular Biophysics Group Institute of Integrative Biology Faculty of Health and Life Sciences University of Liverpool UK
| | | | - Balasundaram Padmanabhan
- Department of Biophysics National Institute of Mental Health and Neurosciences (NIMHANS) Bangalore India
| |
Collapse
|
37
|
The cysteine-reactive small molecule ebselen facilitates effective SOD1 maturation. Nat Commun 2018; 9:1693. [PMID: 29703933 PMCID: PMC5923229 DOI: 10.1038/s41467-018-04114-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 04/04/2018] [Indexed: 11/20/2022] Open
Abstract
Superoxide dismutase-1 (SOD1) mutants, including those with unaltered enzymatic activity, are known to cause amyotrophic lateral sclerosis (ALS). Several destabilizing factors contribute to pathogenicity including a reduced ability to complete the normal maturation process which comprises folding, metal cofactor acquisition, intra-subunit disulphide bond formation and dimerization. Immature SOD1 forms toxic oligomers and characteristic large insoluble aggregates within motor system cells. Here we report that the cysteine-reactive molecule ebselen efficiently confers the SOD1 intra-subunit disulphide and directs correct SOD1 folding, depopulating the globally unfolded precursor associated with aggregation and toxicity. Assisted formation of the unusual SOD1 cytosolic disulphide bond could have potential therapeutic applications. In less reducing environments, ebselen forms a selenylsulphide with Cys111 and restores the monomer–dimer equilibrium of A4V SOD1 to wild-type. Ebselen is therefore a potent bifunctional pharmacological chaperone for SOD1 that combines properties of the SOD1 chaperone hCCS and the recently licenced antioxidant drug, edaravone. Mutations in superoxide dismutase-1 (SOD1) cause amyotrophic lateral sclerosis (ALS). Here the authors present the SOD1 crystal structure bound to the small cysteine-reactive molecule ebselen and show that ebselen is a chaperone for SOD1.
Collapse
|
38
|
Tokuda E, Nomura T, Ohara S, Watanabe S, Yamanaka K, Morisaki Y, Misawa H, Furukawa Y. A copper-deficient form of mutant Cu/Zn-superoxide dismutase as an early pathological species in amyotrophic lateral sclerosis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2119-2130. [PMID: 29551730 DOI: 10.1016/j.bbadis.2018.03.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 02/27/2018] [Accepted: 03/14/2018] [Indexed: 12/13/2022]
Abstract
Dominant mutations in the gene encoding copper and zinc-binding superoxide dismutase (SOD1) cause amyotrophic lateral sclerosis (ALS). Abnormal accumulation of misfolded SOD1 proteins in spinal motoneurons is a major pathological hallmark in SOD1-related ALS. Dissociation of copper and/or zinc ions from SOD1 has been shown to trigger the protein aggregation/oligomerization in vitro, but the pathological contribution of such metal dissociation to the SOD1 misfolding still remains obscure. Here, we tested the relevance of the metal-deficient SOD1 in the misfolding in vivo by developing a novel antibody (anti-apoSOD), which exclusively recognized mutant SOD1 deficient in metal ions at its copper-binding site. Notably, anti-apoSOD-reactive species were detected specifically in the spinal cords of the ALS model mice only at their early pre-symptomatic stages but not at the end stage of the disease. The cerebrospinal fluid as well as the spinal cord homogenate of one SOD1-ALS patient also contained the anti-apoSOD-reactive species. Our results thus suggest that metal-deficiency in mutant SOD1 at its copper-binding site is one of the earliest pathological features in SOD1-ALS.
Collapse
Affiliation(s)
- Eiichi Tokuda
- Laboratory for Mechanistic Chemistry of Biomolecules, Department of Chemistry, Keio University, Yokohama 223-8522, Japan
| | - Takao Nomura
- Laboratory for Mechanistic Chemistry of Biomolecules, Department of Chemistry, Keio University, Yokohama 223-8522, Japan.
| | - Shinji Ohara
- Department of Neurology, Matsumoto Medical Center, Matsumoto 399-0021, Japan.
| | - Seiji Watanabe
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan.
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan.
| | - Yuta Morisaki
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Hidemi Misawa
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan.
| | - Yoshiaki Furukawa
- Laboratory for Mechanistic Chemistry of Biomolecules, Department of Chemistry, Keio University, Yokohama 223-8522, Japan.
| |
Collapse
|
39
|
Enge TG, Ecroyd H, Jolley DF, Yerbury JJ, Kalmar B, Dosseto A. Assessment of metal concentrations in the SOD1 G93A mouse model of amyotrophic lateral sclerosis and its potential role in muscular denervation, with particular focus on muscle tissue. Mol Cell Neurosci 2018. [PMID: 29524628 DOI: 10.1016/j.mcn.2018.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is among the most common of the motor neuron diseases, and arguably the most devastating. During the course of this fatal neurodegenerative disorder, motor neurons undergo progressive degeneration. The currently best-understood animal models of ALS are based on the over-expression of mutant isoforms of Cu/Zn superoxide dismutase 1 (SOD1); these indicate that there is a perturbation in metal homeostasis with disease progression. Copper metabolism in particular is affected in the central nervous system (CNS) and muscle tissue. METHODS This present study assessed previously published and newly gathered concentrations of transition metals (Cu, Zn, Fe and Se) in CNS (brain and spinal cord) and non-CNS (liver, intestine, heart and muscle) tissues from transgenic mice over-expressing the G93A mutant SOD1 isoform (SOD1G93A), transgenic mice over-expressing wildtype SOD1 (SOD1WT) and non-transgenic controls. RESULTS Cu accumulates in non-CNS tissues at pre-symptomatic stages in SOD1G93A tissues. This accumulation represents a potentially pathological feature that cannot solely be explained by the over-expression of mSOD1. As a result of the lack of Cu uptake into the CNS there may be a deficiency of Cu for the over-expressed mutant SOD1 in these tissues. Elevated Cu concentrations in muscle tissue also preceded the onset of symptoms and were found to be pathological and not be the result of SOD1 over-expression. CONCLUSIONS It is hypothesized that the observed Cu accumulations may represent a pathologic feature of ALS, which may actively contribute to axonal retraction leading to muscular denervation, and possibly significantly contributing to disease pathology. Therefore, it is proposed that the toxic-gain-of-function and dying-back hypotheses to explain the molecular drivers of ALS may not be separate, individual processes; rather our data suggests that they are parallel processes.
Collapse
Affiliation(s)
- T Gabriel Enge
- Wollongong Isotope Geochronology Laboratory and School of Earth and Environmental Sciences, University of Wollongong, Australia.
| | - Heath Ecroyd
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong, Australia
| | - Dianne F Jolley
- Center for Medical and Molecular Bioscience and School of Chemistry, University of Wollongong, Australia
| | - Justin J Yerbury
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong, Australia
| | - Bernadett Kalmar
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, UK
| | - Anthony Dosseto
- Wollongong Isotope Geochronology Laboratory and School of Earth and Environmental Sciences, University of Wollongong, Australia
| |
Collapse
|
40
|
Ayers JI, Cashman NR. Prion-like mechanisms in amyotrophic lateral sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2018; 153:337-354. [PMID: 29887144 DOI: 10.1016/b978-0-444-63945-5.00018-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The prion hypothesis - a protein conformation capable of replicating without a nucleic acid genome - was heretical at the time of its discovery. However, the characteristics of the disease-misfolded prion protein and its ability to transmit disease, replicate, and spread are now widely accepted throughout the scientific community. In fact, in the last decade a wealth of evidence has emerged supporting similar properties observed for many of the misfolded proteins implicated in other neurodegenerative diseases, such as Alzheimer disease, Parkinson disease, tauopathies, and as described in this chapter, amyotrophic lateral sclerosis (ALS). Multiple studies have now demonstrated the ability for superoxide dismutase-1, 43-kDa transactive response (TAR) DNA-binding protein, fused-in sarcoma, and most recently, C9orf72-encoded polypeptides to display properties similar to those of prions. The majority of these are cell-free and in vitro assays, while superoxide dismutase-1 remains the only ALS-linked protein to demonstrate several prion-like properties in vivo. In this chapter, we provide an introduction to ALS and review the recent literature linking several proteins implicated in the familial forms of the disease to properties of the prion protein.
Collapse
Affiliation(s)
- Jacob I Ayers
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease (CTRND), University of Florida, Gainesville, FL, United States
| | - Neil R Cashman
- Department of Medicine, Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
41
|
Uversky VN. The roles of intrinsic disorder-based liquid-liquid phase transitions in the "Dr. Jekyll-Mr. Hyde" behavior of proteins involved in amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Autophagy 2017; 13:2115-2162. [PMID: 28980860 DOI: 10.1080/15548627.2017.1384889] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pathological developments leading to amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are associated with misbehavior of several key proteins, such as SOD1 (superoxide dismutase 1), TARDBP/TDP-43, FUS, C9orf72, and dipeptide repeat proteins generated as a result of the translation of the intronic hexanucleotide expansions in the C9orf72 gene, PFN1 (profilin 1), GLE1 (GLE1, RNA export mediator), PURA (purine rich element binding protein A), FLCN (folliculin), RBM45 (RNA binding motif protein 45), SS18L1/CREST, HNRNPA1 (heterogeneous nuclear ribonucleoprotein A1), HNRNPA2B1 (heterogeneous nuclear ribonucleoprotein A2/B1), ATXN2 (ataxin 2), MAPT (microtubule associated protein tau), and TIA1 (TIA1 cytotoxic granule associated RNA binding protein). Although these proteins are structurally and functionally different and have rather different pathological functions, they all possess some levels of intrinsic disorder and are either directly engaged in or are at least related to the physiological liquid-liquid phase transitions (LLPTs) leading to the formation of various proteinaceous membrane-less organelles (PMLOs), both normal and pathological. This review describes the normal and pathological functions of these ALS- and FTLD-related proteins, describes their major structural properties, glances at their intrinsic disorder status, and analyzes the involvement of these proteins in the formation of normal and pathological PMLOs, with the ultimate goal of better understanding the roles of LLPTs and intrinsic disorder in the "Dr. Jekyll-Mr. Hyde" behavior of those proteins.
Collapse
Affiliation(s)
- Vladimir N Uversky
- a Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute , Morsani College of Medicine , University of South Florida , Tampa , FL , USA.,b Institute for Biological Instrumentation of the Russian Academy of Sciences , Pushchino, Moscow region , Russia
| |
Collapse
|
42
|
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: 2.8] [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
|
43
|
Abdolvahabi A, Shi Y, Rasouli S, Croom CM, Aliyan A, Martí AA, Shaw BF. Kaplan-Meier Meets Chemical Kinetics: Intrinsic Rate of SOD1 Amyloidogenesis Decreased by Subset of ALS Mutations and Cannot Fully Explain Age of Disease Onset. ACS Chem Neurosci 2017; 8:1378-1389. [PMID: 28290665 DOI: 10.1021/acschemneuro.7b00029] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Over 150 mutations in SOD1 (superoxide dismutase-1) cause amyotrophic lateral sclerosis (ALS), presumably by accelerating SOD1 amyloidogenesis. Like many nucleation processes, SOD1 fibrillization is stochastic (in vitro), which inhibits the determination of aggregation rates (and obscures whether rates correlate with patient phenotypes). Here, we diverged from classical chemical kinetics and used Kaplan-Meier estimators to quantify the probability of apo-SOD1 fibrillization (in vitro) from ∼103 replicate amyloid assays of wild-type (WT) SOD1 and nine ALS variants. The probability of apo-SOD1 fibrillization (expressed as a Hazard ratio) is increased by certain ALS-linked SOD1 mutations but is decreased or remains unchanged by other mutations. Despite this diversity, Hazard ratios of fibrillization correlated linearly with (and for three mutants, approximately equaled) Hazard ratios of patient survival (R2 = 0.67; Pearson's r = 0.82). No correlation exists between Hazard ratios of fibrillization and age of initial onset of ALS (R2 = 0.09). Thus, Hazard ratios of fibrillization might explain rates of disease progression but not onset. Classical kinetic metrics of fibrillization, i.e., mean lag time and propagation rate, did not correlate as strongly with phenotype (and ALS mutations did not uniformly accelerate mean rate of nucleation or propagation). A strong correlation was found, however, between mean ThT fluorescence at lag time and patient survival (R2 = 0.93); oligomers of SOD1 with weaker fluorescence correlated with shorter survival. This study suggests that SOD1 mutations trigger ALS by altering a property of SOD1 or its oligomers other than the intrinsic rate of amyloid nucleation (e.g., oligomer stability; rates of intercellular propagation; affinity for membrane surfaces; and maturation rate).
Collapse
Affiliation(s)
- Alireza Abdolvahabi
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Yunhua Shi
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Sanaz Rasouli
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76706, United States
| | - Corbin M. Croom
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Amir Aliyan
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Angel A. Martí
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Bryan F. Shaw
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| |
Collapse
|
44
|
Keskin I, Birve A, Berdynski M, Hjertkvist K, Rofougaran R, Nilsson TK, Glass JD, Marklund SL, Andersen PM. Comprehensive analysis to explain reduced or increased SOD1 enzymatic activity in ALS patients and their relatives. Amyotroph Lateral Scler Frontotemporal Degener 2017; 18:457-463. [DOI: 10.1080/21678421.2017.1301481] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Isil Keskin
- Department of Pharmacology and Clinical Neurosciences, Umeå University, Umeå, Sweden,
| | - Anna Birve
- Department of Pharmacology and Clinical Neurosciences, Umeå University, Umeå, Sweden,
| | - Mariusz Berdynski
- Department of Pharmacology and Clinical Neurosciences, Umeå University, Umeå, Sweden,
- Department of Neurodegenerative Disorders, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland,
| | - Karin Hjertkvist
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden, and
| | - Reza Rofougaran
- Department of Pharmacology and Clinical Neurosciences, Umeå University, Umeå, Sweden,
| | - Torbjörn K. Nilsson
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden, and
| | - Jonathan D. Glass
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Stefan L. Marklund
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden, and
| | - Peter M. Andersen
- Department of Pharmacology and Clinical Neurosciences, Umeå University, Umeå, Sweden,
| |
Collapse
|
45
|
Antinone SE, Ghadge GD, Ostrow LW, Roos RP, Green WN. S-acylation of SOD1, CCS, and a stable SOD1-CCS heterodimer in human spinal cords from ALS and non-ALS subjects. Sci Rep 2017; 7:41141. [PMID: 28120938 PMCID: PMC5264640 DOI: 10.1038/srep41141] [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: 09/22/2016] [Accepted: 12/15/2016] [Indexed: 11/09/2022] Open
Abstract
Previously, we found that human Cu, Zn-superoxide dismutase (SOD1) is S-acylated (palmitoylated) in vitro and in amyotrophic lateral sclerosis (ALS) mouse models, and that S-acylation increased for ALS-causing SOD1 mutants relative to wild type. Here, we use the acyl resin-assisted capture (acyl-RAC) assay to demonstrate S-acylation of SOD1 in human post-mortem spinal cord homogenates from ALS and non-ALS subjects. Acyl-RAC further revealed that endogenous copper chaperone for SOD1 (CCS) is S-acylated in both human and mouse spinal cords, and in vitro in HEK293 cells. SOD1 and CCS formed a highly stable heterodimer in human spinal cord homogenates that was resistant to dissociation by boiling, denaturants, or reducing agents and was not observed in vitro unless both SOD1 and CCS were overexpressed. Cysteine mutations that attenuate SOD1 maturation prevented the SOD1-CCS heterodimer formation. The degree of S-acylation was highest for SOD1-CCS heterodimers, intermediate for CCS monomers, and lowest for SOD1 monomers. Given that S-acylation facilitates anchoring of soluble proteins to cell membranes, our findings suggest that S-acylation and membrane localization may play an important role in CCS-mediated SOD1 maturation. Furthermore, the highly stable S-acylated SOD1-CCS heterodimer may serve as a long-lived maturation intermediate in human spinal cord.
Collapse
Affiliation(s)
- Sarah E Antinone
- University of Chicago, Department of Neurobiology, Chicago, 60637, USA
| | | | - Lyle W Ostrow
- Johns Hopkins University, Department of Neurology, Baltimore, 21205, USA
| | - Raymond P Roos
- University of Chicago, Department of Neurology, Chicago, 60637, USA
| | - William N Green
- University of Chicago, Department of Neurobiology, Chicago, 60637, USA
| |
Collapse
|
46
|
Tokuda E, Anzai I, Nomura T, Toichi K, Watanabe M, Ohara S, Watanabe S, Yamanaka K, Morisaki Y, Misawa H, Furukawa Y. Immunochemical characterization on pathological oligomers of mutant Cu/Zn-superoxide dismutase in amyotrophic lateral sclerosis. Mol Neurodegener 2017; 12:2. [PMID: 28057013 PMCID: PMC5216565 DOI: 10.1186/s13024-016-0145-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/20/2016] [Indexed: 12/11/2022] Open
Abstract
Background Dominant mutations in Cu/Zn-superoxide dismutase (SOD1) gene cause a familial form of amyotrophic lateral sclerosis (SOD1-ALS) with accumulation of misfolded SOD1 proteins as intracellular inclusions in spinal motor neurons. Oligomerization of SOD1 via abnormal disulfide crosslinks has been proposed as one of the misfolding pathways occurring in mutant SOD1; however, the pathological relevance of such oligomerization in the SOD1-ALS cases still remains obscure. Methods We prepared antibodies exclusively recognizing the SOD1 oligomers cross-linked via disulfide bonds in vitro. By using those antibodies, immunohistochemical examination and ELISA were mainly performed on the tissue samples of transgenic mice expressing mutant SOD1 proteins and also of human SOD1-ALS cases. Results We showed the recognition specificity of our antibodies exclusively toward the disulfide-crosslinked SOD1 oligomers by ELISA using various forms of purified SOD1 proteins in conformationally distinct states in vitro. Furthermore, the epitope of those antibodies was buried and inaccessible in the natively folded structure of SOD1. The antibodies were then found to specifically detect the pathological SOD1 species in the spinal motor neurons of the SOD1-ALS patients as well as the transgenic model mice. Conclusions Our findings here suggest that the SOD1 oligomerization through the disulfide-crosslinking associates with exposure of the SOD1 structural interior and is a pathological process occurring in the SOD1-ALS cases. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0145-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eiichi Tokuda
- Laboratory for Mechanistic Chemistry of Biomolecules, Department of Chemistry, Keio University, 3-14-1 Hiyoshi, Kohoku, Yokohama, Kanagawa, 223-8522, Japan
| | - Itsuki Anzai
- Laboratory for Mechanistic Chemistry of Biomolecules, Department of Chemistry, Keio University, 3-14-1 Hiyoshi, Kohoku, Yokohama, Kanagawa, 223-8522, Japan
| | - Takao Nomura
- Laboratory for Mechanistic Chemistry of Biomolecules, Department of Chemistry, Keio University, 3-14-1 Hiyoshi, Kohoku, Yokohama, Kanagawa, 223-8522, Japan
| | - Keisuke Toichi
- Laboratory for Mechanistic Chemistry of Biomolecules, Department of Chemistry, Keio University, 3-14-1 Hiyoshi, Kohoku, Yokohama, Kanagawa, 223-8522, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
| | - Shinji Ohara
- Department of Neurology, Matsumoto Medical Center, Matsumoto, 399-0021, Japan
| | - Seiji Watanabe
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan
| | - Yuta Morisaki
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Hidemi Misawa
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Yoshiaki Furukawa
- Laboratory for Mechanistic Chemistry of Biomolecules, Department of Chemistry, Keio University, 3-14-1 Hiyoshi, Kohoku, Yokohama, Kanagawa, 223-8522, Japan.
| |
Collapse
|
47
|
Wright GSA, Antonyuk SV, Hasnain SS. A faulty interaction between SOD1 and hCCS in neurodegenerative disease. Sci Rep 2016; 6:27691. [PMID: 27282955 PMCID: PMC4901319 DOI: 10.1038/srep27691] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/19/2016] [Indexed: 01/12/2023] Open
Abstract
A proportion of Amyotrophic lateral sclerosis (ALS) cases result from impaired mutant superoxide dismutase-1 (SOD1) maturation. The copper chaperone for SOD1 (hCCS) forms a transient complex with SOD1 and catalyses the final stages of its maturation. We find that a neurodegenerative disease-associated hCCS mutation abrogates the interaction with SOD1 by inhibiting hCCS zinc binding. Analogously, SOD1 zinc loss has a detrimental effect on the formation, structure and disassociation of the hCCS-SOD1 heterodimer. This suggests that hCCS functionality is impaired by ALS mutations that reduce SOD1 zinc affinity. Furthermore, stabilization of wild-type SOD1 by chemical modification including cisplatination, inhibits complex formation. We hypothesize that drug molecules designed to stabilize ALS SOD1 mutants that also target the wild-type form will lead to characteristics common in SOD1 knock-outs. Our work demonstrates the applicability of chromatographic SAXS when studying biomolecules predisposed to aggregation or dissociation; attributes frequently reported for complexes involved in neurodegenerative disease.
Collapse
Affiliation(s)
- Gareth S. A. Wright
- Molecular Biophysics Group, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, UK
| | - Svetlana V. Antonyuk
- Molecular Biophysics Group, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, UK
| | - S. Samar Hasnain
- Molecular Biophysics Group, Institute of Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, UK
| |
Collapse
|
48
|
Álvarez-Zaldiernas C, Lu J, Zheng Y, Yang H, Blasi J, Solsona C, Holmgren A. Cellular Redox Systems Impact the Aggregation of Cu,Zn Superoxide Dismutase Linked to Familial Amyotrophic Lateral Sclerosis. J Biol Chem 2016; 291:17197-208. [PMID: 27261461 DOI: 10.1074/jbc.m115.708230] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Indexed: 12/11/2022] Open
Abstract
Protein misfolding is implicated in neurodegenerative diseases such as ALS, where mutations of superoxide dismutase 1 (SOD1) account for about 20% of the inherited mutations. Human SOD1 (hSOD1) contains four cysteines, including Cys(57) and Cys(146), which have been linked to protein stability and folding via forming a disulfide bond, and Cys(6) and Cys(111) as free thiols. But the roles of the cellular oxidation-reduction (redox) environment in SOD1 folding and aggregation are not well understood. Here we explore the effects of cellular redox systems on the aggregation of hSOD1 proteins. We found that the known hSOD1 mutations G93A and A4V increased the capability of the thioredoxin and glutaredoxin systems to reduce hSOD1 compared with wild-type hSOD1. Treatment with inhibitors of these redox systems resulted in an increase of hSOD1 aggregates in the cytoplasm of cells transfected with mutants but not in cells transfected with wild-type hSOD1 or those containing a secondary C111G mutation. This aggregation may be coupled to changes in the redox state of the G93A and A4V mutants upon mild oxidative stress. These results strongly suggest that the thioredoxin and glutaredoxin systems are the key regulators for hSOD1 aggregation and may play critical roles in the pathogenesis of ALS.
Collapse
Affiliation(s)
- Cristina Álvarez-Zaldiernas
- From the Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden, the Department of Pathology and Experimental Therapeutics, Faculty of Medicine, Campus Bellvitge, University of Barcelona, Feixa Llarga s/n. Hospitalet de Llobregat, 08907 Barcelona, Spain, and the Bellvitge Biomedical Research Institute, Gran Via de l'Hospitalet, 199-203, L'Hospitalet de Llobregat, Barcelona, 08908 Barcelona, Spain
| | - Jun Lu
- From the Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden,
| | - Yujuan Zheng
- From the Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Hongqian Yang
- From the Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Juan Blasi
- the Department of Pathology and Experimental Therapeutics, Faculty of Medicine, Campus Bellvitge, University of Barcelona, Feixa Llarga s/n. Hospitalet de Llobregat, 08907 Barcelona, Spain, and the Bellvitge Biomedical Research Institute, Gran Via de l'Hospitalet, 199-203, L'Hospitalet de Llobregat, Barcelona, 08908 Barcelona, Spain
| | - Carles Solsona
- the Department of Pathology and Experimental Therapeutics, Faculty of Medicine, Campus Bellvitge, University of Barcelona, Feixa Llarga s/n. Hospitalet de Llobregat, 08907 Barcelona, Spain, and the Bellvitge Biomedical Research Institute, Gran Via de l'Hospitalet, 199-203, L'Hospitalet de Llobregat, Barcelona, 08908 Barcelona, Spain
| | - Arne Holmgren
- From the Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177, Stockholm, Sweden,
| |
Collapse
|
49
|
Bidhendi EE, Bergh J, Zetterström P, Andersen PM, Marklund SL, Brännström T. Two superoxide dismutase prion strains transmit amyotrophic lateral sclerosis-like disease. J Clin Invest 2016; 126:2249-53. [PMID: 27140399 DOI: 10.1172/jci84360] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 03/10/2016] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset degeneration of motor neurons that is commonly caused by mutations in the gene encoding superoxide dismutase 1 (SOD1). Both patients and Tg mice expressing mutant human SOD1 (hSOD1) develop aggregates of unknown importance. In Tg mice, 2 different strains of hSOD1 aggregates (denoted A and B) can arise; however, the role of these aggregates in disease pathogenesis has not been fully characterized. Here, minute amounts of strain A and B hSOD1 aggregate seeds that were prepared by centrifugation through a density cushion were inoculated into lumbar spinal cords of 100-day-old mice carrying a human SOD1 Tg. Mice seeded with A or B aggregates developed premature signs of ALS and became terminally ill after approximately 100 days, which is 200 days earlier than for mice that had not been inoculated or were given a control preparation. Concomitantly, exponentially growing strain A and B hSOD1 aggregations propagated rostrally throughout the spinal cord and brainstem. The phenotypes provoked by the A and B strains differed regarding progression rates, distribution, end-stage aggregate levels, and histopathology. Together, our data indicate that the aggregate strains are prions that transmit a templated, spreading aggregation of hSOD1, resulting in a fatal ALS-like disease.
Collapse
|
50
|
Copper Homeostasis as a Therapeutic Target in Amyotrophic Lateral Sclerosis with SOD1 Mutations. Int J Mol Sci 2016; 17:ijms17050636. [PMID: 27136532 PMCID: PMC4881462 DOI: 10.3390/ijms17050636] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/03/2016] [Accepted: 04/22/2016] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a lethal neurodegenerative disease affecting both upper and lower motor neurons, and currently, there is no cure or effective treatment. Mutations in a gene encoding a ubiquitous antioxidant enzyme, Cu,Zn-superoxide dismutase (SOD1), have been first identified as a cause of familial forms of ALS. It is widely accepted that mutant SOD1 proteins cause the disease through a gain in toxicity but not through a loss of its physiological function. SOD1 is a major copper-binding protein and regulates copper homeostasis in the cell; therefore, a toxicity of mutant SOD1 could arise from the disruption of copper homeostasis. In this review, we will briefly review recent studies implying roles of copper homeostasis in the pathogenesis of SOD1-ALS and highlight the therapeutic interventions focusing on pharmacological as well as genetic regulations of copper homeostasis to modify the pathological process in SOD1-ALS.
Collapse
|