1
|
Gómez-Gálvez P, Navarro V, Castro AM, Paradas C, Escudero LM. Computational Analysis of SOD1-G93A Mouse Muscle Biomarkers for Comprehensive Assessment of ALS Progression. Neuropathol Appl Neurobiol 2025; 51:e70014. [PMID: 40164574 DOI: 10.1111/nan.70014] [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: 07/26/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
AIMS To identify potential image biomarkers of neuromuscular disease by analysing morphological and network-derived features in skeletal muscle biopsies from a murine model of amyotrophic lateral sclerosis (ALS), the SOD1G93A mouse and wild-type (WT) controls at distinct stages of disease progression. METHODS Using the NDICIA computational framework, we quantitatively evaluated histological differences between skeletal muscle biopsies from SOD1G93A and WT mice. The process involved the selection of a subset of features revealing these differences. A subset of discriminative features was selected to characterise these differences, and their temporal dynamics were assessed across disease stages. RESULTS Our findings demonstrate that muscle pathology in the mutant model evolves from early alterations in muscle fibre arrangement, detectable at the presymptomatic stage through graph theory features, to the subsequent development of the typical morphological pattern of neurogenic atrophy at more advanced disease stages. CONCLUSIONS Our assay identifies a neurogenic signature in mutant muscle biopsies, even when the disease is phenotypically imperceptible.
Collapse
Affiliation(s)
- Pedro Gómez-Gálvez
- MRC Laboratory of Molecular Biology, Cambridge, UK
- Department of Physiology, Development and Neurobiology, University of Cambridge, Cambridge, UK
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Biología Celular, Facultad de Biología, Universidad de Sevilla, Seville, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Victoria Navarro
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío CSIC/Universidad de Sevilla, Seville, Spain
| | - Ana M Castro
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Biología Celular, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | - Carmen Paradas
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío CSIC/Universidad de Sevilla, Seville, Spain
- Neuromuscular Disease Unit, Neurology Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Luis M Escudero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Biología Celular, Facultad de Biología, Universidad de Sevilla, Seville, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Bond S, Saxena S, Sierra-Delgado JA. Microglia in ALS: Insights into Mechanisms and Therapeutic Potential. Cells 2025; 14:421. [PMID: 40136670 PMCID: PMC11941390 DOI: 10.3390/cells14060421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by the loss of motor neurons, leading to escalating muscle weakness, atrophy, and eventually paralysis. While neurons are the most visibly affected, emerging data highlight microglia-the brain's resident immune cells-as key contributors to disease onset and progression. Rather than existing in a simple beneficial or harmful duality, microglia can adopt multiple functional states shaped by internal and external factors, including those in ALS. Collectively, these disease-specific forms are called disease-associated microglia (DAM). Research using rodent models, patient-derived cells, and human postmortem tissue shows that microglia can transition into DAM phenotypes, driving inflammation and neuronal injury. However, these cells can also fulfill protective roles under certain conditions, revealing their adaptable nature. This review explores recent discoveries regarding the multifaceted behavior of microglia in ALS, highlights important findings that link these immune cells to motor neuron deterioration, and discusses emerging therapies-some already used in clinical trials-that aim to recalibrate microglial functions and potentially slow disease progression.
Collapse
Affiliation(s)
- Silvano Bond
- Department of Physical Medicine and Rehabilitation, University of Missouri School of Medicine, Columbia, MO 65211, USA;
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA
| | - Smita Saxena
- Department of Physical Medicine and Rehabilitation, University of Missouri School of Medicine, Columbia, MO 65211, USA;
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA
| | - Julieth A. Sierra-Delgado
- Department of Physical Medicine and Rehabilitation, University of Missouri School of Medicine, Columbia, MO 65211, USA;
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
3
|
Petro TM, Esmael A, Pattee GL, Al-Sarmi F, Chiodo F, Agarkova IV, Dunigan DD, Van Etten JL. Expression of human superoxide dismutase (SOD) 1 G93A and chlorovirus ATCV-1 SOD increases the response of macrophages to inflammatory stimulants, including ATCV-1 major capsid protein glycans. Immunobiology 2025; 230:152881. [PMID: 39965330 DOI: 10.1016/j.imbio.2025.152881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/27/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
One cause of familial Amyotrophic Lateral Sclerosis (ALS) is a mutation in Super Oxide Dismutase 1 (SOD1) whereby amino acid 93 is alanine instead of glycine (SOD1-G93A). Transgenic mice expressing human SOD1-G93A pathogenic variant develop motor neuron disease (MND), similar to ALS. Humans with ALS and SOD1-G93A mice have elevated production of inflammatory cytokines, such as IL-6, which may promote MND. We previously showed that infection with the Chlorovirus Acanthocystis turfacea chlorella virus 1 (ATCV-1), which encodes a SOD1, accelerates onset of MND in these mice and induces macrophages to produce high levels of IL-6. We confirm here that ALS patients compared with healthy controls have significantly elevated levels of plasma IL-6 and Interferon-gamma (IFN-γ), but not IL-17. To determine if expression of ATCV-1 SOD1 or SOD1-G93A in mouse macrophages elevates expression of inflammatory cytokines, we transfected the RAW264.7 mouse macrophage cell line with plasmids encoding ATCV-1 SOD1, wild-type human SOD1, SOD1-G93A, or an empty vector. RAW264.7 cells stably expressing wtSOD1 or G93A-SOD1 were stimulated with poly I:C and Interferon-gamma, alone, or in combination to induce inflammatory factors, such as IL-6 and Nitric Oxide (NO), anti-inflammatory factors, such as IL-10, or activation of Interferon Stimulated Response Elements (ISRE) promoters. After stimulation, production of IL-6 and NO, but not IL-10 or ISRE promoter activity was significantly higher in RAW264.7 cells expressing SOD1-G93A compared with wt SOD1. Moreover, RAW264.7 cells expressing SOD1-G93A compared with wt SOD1 produced higher levels of IL-6 and NO in response to ATCV-1 glycoproteins. Finally, transfection of plasmid encoding ATCV-1 SOD1 into RAW264.7 cells significantly increased expression of inflammatory factors in responses to poly I:C and IFN-γ, primarily in an Interferon regulatory factor 3 (IRF3) dependent fashion. These data clearly show that expression of G93A-SOD1 or ATCV-1 SOD1 in macrophages significantly elevates expression of inflammatory factors following stimulations that mimic virus infection, viral components, or T cell cytokines, thereby suggesting one mechanism by which atypical SOD1 in macrophages can contribute to ALS-MND.
Collapse
Affiliation(s)
- Thomas M Petro
- Department of Oral Biology, University of Nebraska Medical Center, USA; Nebraska Center for Virology University of Nebraska Lincoln, Lincoln, Nebraska 68583, USA.
| | - Ahmed Esmael
- Nebraska Center for Virology University of Nebraska Lincoln, Lincoln, Nebraska 68583, USA; Benha University, College of Science, Egypt
| | - Gary L Pattee
- Bryan Health Physician Network, Lincoln, Nebraska, USA
| | - Fiyad Al-Sarmi
- Nebraska Center for Virology University of Nebraska Lincoln, Lincoln, Nebraska 68583, USA
| | - Fabrizio Chiodo
- Institute of Biomolecular Chemistry and Dept. of Agricultural Sciences, University of Napoli, Italy
| | - Irina V Agarkova
- Nebraska Center for Virology University of Nebraska Lincoln, Lincoln, Nebraska 68583, USA
| | - David D Dunigan
- Nebraska Center for Virology University of Nebraska Lincoln, Lincoln, Nebraska 68583, USA
| | - James L Van Etten
- Nebraska Center for Virology University of Nebraska Lincoln, Lincoln, Nebraska 68583, USA
| |
Collapse
|
4
|
Asai Y, Yano K, Higashino T, Yoshihara D, Sakiyama H, Eguchi H, Fukushima K, Suzuki K, Fujiwara N. The Ile35 Residue of the ALS-Associated Mutant SOD1 Plays a Crucial Role in the Intracellular Aggregation of the Molecule. Mol Neurobiol 2025; 62:2023-2038. [PMID: 39060907 DOI: 10.1007/s12035-024-04369-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with an unknown pathogenesis. It has been reported that mutations in the gene for Cu/Zn superoxide dismutase (SOD1) cause familial ALS. Mutant SOD1 undergoes aggregation and forms amyloid more easily, and SOD1-immunopositive inclusions have been observed in the spinal cords of ALS patients. Because of this, SOD1 aggregation is thought to be related to the pathogenesis of ALS. Some core regions of amyloid have been identified, but the issue of whether these regions form aggregates in living cells remains unclear, and the mechanism responsible for intracellular SOD1 aggregation also remains unclear. The findings reported in this study indicate that the aggregation of the ALS-linked mutant SOD1-EGFP was significantly enhanced when the BioID2 gene was fused to the N-terminus of the mutant SOD1-EGFP plasmid for cellular expression. Expression of a series of BioID2-(C-terminal deletion peptides of SOD1)-EGFP permitted us to identify 1-35 as a minimal N-terminal sequence and Ile35 as an essential amino acid residue that contributes to the intracellular aggregation of SOD1. The findings also showed that an additional substitution of Ile35 with Ser into the ALS mutant SOD1 resulted in the significant suppression of aggregate formation. The fact that no Ile35 mutations have been reported to date in ALS patients indicates that all ALS mutant SOD1s contain Ile35. Taken together, we propose that Ile35 plays a pivotal role in the aggregation of the ALS-linked SOD1 and that this study will contribute to our understanding of the mechanism responsible for SOD1 aggregation.
Collapse
Affiliation(s)
- Yoshiyuki Asai
- Department of Biochemistry, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Kyoka Yano
- Department of Biochemistry, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Tomoyuki Higashino
- Department of Biochemistry, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Daisaku Yoshihara
- Department of Biochemistry, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
- Labolatory of Biochemistry, School of Pharmacy, Hyogo Medical University, Kobe, Hyogo, 650-8530, Japan
| | - Haruhiko Sakiyama
- Department of Biochemistry, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
- Faculty of Nutrition, Department of Food and Nutrition, Senri Kinran University, Suita, Osaka, 565-0873, Japan
| | - Hironobu Eguchi
- Department of Biochemistry, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Kazuaki Fukushima
- Department of Chemistry, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, 663-8501, Japan
| | - Keiichiro Suzuki
- Department of Biochemistry, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Noriko Fujiwara
- Department of Biochemistry, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan.
| |
Collapse
|
5
|
Cabeza-Fernández S, Hernández-Rojas R, Casillas-Bajo A, Patel N, de la Fuente AG, Cabedo H, Gomez-Sanchez JA. Schwann cell JUN expression worsens motor performance in an amyotrophic lateral sclerosis mouse model. Glia 2024; 72:2178-2189. [PMID: 39149866 DOI: 10.1002/glia.24604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease characterized by motor neuron death and distal axonopathy. Despite its clinical severity and profound impact in the patients and their families, many questions about its pathogenesis remain still unclear, including the role of Schwann cells and axon-glial signaling in disease progression. Upon axonal injury, upregulation of JUN transcription factor promotes Schwann cell reprogramming into a repair phenotype that favors axon regrowth and neuronal survival. To study the potential role of repair Schwann cells on motoneuron survival in amyotrophic lateral sclerosis, we generated a mouse line that over-expresses JUN in the Schwann cells of the SOD1G93A mutant, a mouse model of this disease. Then, we explored disease progression by evaluating survival, motor performance and histology of peripheral nerves and spinal cord of these mice. We found that Schwann cell JUN overexpression does not prevent axon degeneration neither motor neuron death in the SOD1G93A mice. Instead, it induces a partial demyelination of medium and large size axons, worsening motor performance and resulting in more aggressive disease phenotype.
Collapse
Affiliation(s)
- Sonia Cabeza-Fernández
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Alicante, Spain
- Molecular control of neuronal axon myelination laboratory, Instituto de Neurociencias UMH-CSIC, Sant Joan d'Alacant, Spain
| | - Rubí Hernández-Rojas
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Alicante, Spain
- Molecular control of neuronal axon myelination laboratory, Instituto de Neurociencias UMH-CSIC, Sant Joan d'Alacant, Spain
| | - Angeles Casillas-Bajo
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Alicante, Spain
- Molecular control of neuronal axon myelination laboratory, Instituto de Neurociencias UMH-CSIC, Sant Joan d'Alacant, Spain
| | - Nikiben Patel
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Alicante, Spain
- Molecular control of neuronal axon myelination laboratory, Instituto de Neurociencias UMH-CSIC, Sant Joan d'Alacant, Spain
| | - Alerie G de la Fuente
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Alicante, Spain
- Molecular control of neuronal axon myelination laboratory, Instituto de Neurociencias UMH-CSIC, Sant Joan d'Alacant, Spain
| | - Hugo Cabedo
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Alicante, Spain
- Molecular control of neuronal axon myelination laboratory, Instituto de Neurociencias UMH-CSIC, Sant Joan d'Alacant, Spain
| | - Jose A Gomez-Sanchez
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Alicante, Spain
- Molecular control of neuronal axon myelination laboratory, Instituto de Neurociencias UMH-CSIC, Sant Joan d'Alacant, Spain
| |
Collapse
|
6
|
Du M, Akerman SC, Fare CM, Ruan L, Vidensky S, Mamedova L, Lee J, Rothstein JD. Divergent and Convergent TMEM106B Pathology in Murine Models of Neurodegeneration and Human Disease. RESEARCH SQUARE 2024:rs.3.rs-5306005. [PMID: 39606446 PMCID: PMC11601866 DOI: 10.21203/rs.3.rs-5306005/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
TMEM106B is a lysosomal/late endosome protein that is a potent genetic modifier of multiple neurodegenerative diseases as well as general aging. Recently, TMEM106B was shown to form insoluble aggregates in postmortem human brain tissue, drawing attention to TMEM106B pathology and the potential role of TMEM106B aggregation in disease. In the context of neurodegenerative diseases, TMEM106B has been studied in vivo using animal models of neurodegeneration, but these studies rely on overexpression or knockdown approaches. To date, endogenous TMEM106B pathology and its relationship to known canonical pathology in animal models has not been reported. Here, we analyze histological patterns of TMEM106B in murine models of C9ORF72-related amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD), SOD1-related ALS, and tauopathy and compare these to postmortem human tissue from patients with C9-ALS/FTD, Alzheimer's disease (AD), and AD with limbic-predominant age-related TDP-43 encephalopathy (AD/LATE). We show that there are significant differences between TMEM106B pathology in mouse models and human patient tissue. Importantly, we also identified convergent evidence from both murine models and human patients that links TMEM106B pathology to TDP-43 nuclear clearance specifically in C9-ALS. Similarly, we find a relationship at the cellular level between TMEM106B pathology and phosphorylated Tau burden in Alzheimer's disease. By characterizing endogenous TMEM106B pathology in both mice and human postmortem tissue, our work reveals considerations that must be taken into account when analyzing data from in vivo mouse studies and elucidates new insights supporting the involvement of TMEM106B in the pathogenesis and progression of multiple neurodegenerative diseases.
Collapse
Affiliation(s)
- Muzi Du
- Johns Hopkins University School of Medicine
| | | | | | | | | | | | - Joshua Lee
- Johns Hopkins University School of Medicine
| | | |
Collapse
|
7
|
Luan T, Li Q, Huang Z, Feng Y, Xu D, Zhou Y, Hu Y, Wang T. Axonopathy Underlying Amyotrophic Lateral Sclerosis: Unraveling Complex Pathways and Therapeutic Insights. Neurosci Bull 2024; 40:1789-1810. [PMID: 39097850 PMCID: PMC11607281 DOI: 10.1007/s12264-024-01267-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/08/2024] [Indexed: 08/05/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex neurodegenerative disorder characterized by progressive axonopathy, jointly leading to the dying back of the motor neuron, disrupting both nerve signaling and motor control. In this review, we highlight the roles of axonopathy in ALS progression, driven by the interplay of multiple factors including defective trafficking machinery, protein aggregation, and mitochondrial dysfunction. Dysfunctional intracellular transport, caused by disruptions in microtubules, molecular motors, and adaptors, has been identified as a key contributor to disease progression. Aberrant protein aggregation involving TDP-43, FUS, SOD1, and dipeptide repeat proteins further amplifies neuronal toxicity. Mitochondrial defects lead to ATP depletion, oxidative stress, and Ca2+ imbalance, which are regarded as key factors underlying the loss of neuromuscular junctions and axonopathy. Mitigating these defects through interventions including neurotrophic treatments offers therapeutic potential. Collaborative research efforts aim to unravel ALS complexities, opening avenues for holistic interventions that target diverse pathological mechanisms.
Collapse
Affiliation(s)
- Tongshu Luan
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Qing Li
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Zhi Huang
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yu Feng
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Duo Xu
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yujie Zhou
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yiqing Hu
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Tong Wang
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
8
|
Du M, Akerman SC, Fare CM, Ruan L, Vidensky S, Mamedova L, Lee J, Rothstein JD. Divergent and Convergent TMEM106B Pathology in Murine Models of Neurodegeneration and Human Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.16.618765. [PMID: 39464100 PMCID: PMC11507888 DOI: 10.1101/2024.10.16.618765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
TMEM106B is a lysosomal/late endosome protein that is a potent genetic modifier of multiple neurodegenerative diseases as well as general aging. Recently, TMEM106B was shown to form insoluble aggregates in postmortem human brain tissue, drawing attention to TMEM106B pathology and the potential role of TMEM106B aggregation in disease. In the context of neurodegenerative diseases, TMEM106B has been studied in vivo using animal models of neurodegeneration, but these studies rely on overexpression or knockdown approaches. To date, endogenous TMEM106B pathology and its relationship to known canonical pathology in animal models has not been reported. Here, we analyze histological patterns of TMEM106B in murine models of C9ORF72-related amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD), SOD1-related ALS, and tauopathy and compare these to postmortem human tissue from patients with C9-ALS/FTD, Alzheimer's disease (AD), and AD with limbic-predominant age-related TDP-43 encephalopathy (AD/LATE). We show that there are significant differences between TMEM106B pathology in mouse models and human patient tissue. Importantly, we also identified convergent evidence from both murine models and human patients that links TMEM106B pathology to TDP-43 nuclear clearance specifically in C9-ALS. Similarly, we find a relationship at the cellular level between TMEM106B pathology and phosphorylated Tau burden in Alzheimer's disease. By characterizing endogenous TMEM106B pathology in both mice and human postmortem tissue, our work reveals considerations that must be taken into account when analyzing data from in vivo mouse studies and elucidates new insights supporting the involvement of TMEM106B in the pathogenesis and progression of multiple neurodegenerative diseases.
Collapse
Affiliation(s)
- Muzi Du
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Suleyman C. Akerman
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Charlotte M. Fare
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Linhao Ruan
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Svetlana Vidensky
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Lyudmila Mamedova
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Joshua Lee
- Department of Psychological and Brain Sciences, Johns Hopkins University Krieger School of Arts and Sciences, Baltimore, MD, 21218, USA
| | - Jeffrey D. Rothstein
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
9
|
Duan C, Kang M, Pan X, Gan Z, Huang V, Li G, Place RF, Li LC. Intrathecal administration of a novel siRNA modality extends survival and improves motor function in the SOD1 G93A ALS mouse model. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102147. [PMID: 38435120 PMCID: PMC10907209 DOI: 10.1016/j.omtn.2024.102147] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 02/12/2024] [Indexed: 03/05/2024]
Abstract
Antisense oligonucleotides (ASOs) were the first modality to pioneer targeted gene knockdown in the treatment of amyotrophic lateral sclerosis (ALS) caused by mutant superoxide dismutase 1 (SOD1). RNA interference (RNAi) is another mechanism of gene silencing in which short interfering RNAs (siRNAs) effectively degrade complementary transcripts. However, delivery to extrahepatic tissues like the CNS has been a bottleneck in the clinical development of RNAi. Herein, we identify potent siRNA duplexes for the knockdown of human SOD1 in which medicinal chemistry and conjugation to an accessory oligonucleotide (ACO) enable activity in CNS tissues. Local delivery via intracerebroventricular or intrathecal injection into SOD1G93A mice delayed disease progression and extended animal survival with superior efficacy compared with an ASO resembling tofersen in sequence and chemistry. Treatment also prevented disease-related declines in motor function, including improvements in animal mobility, muscle strength, and coordination. The ACO itself does not target any specific complementary nucleic acid sequence; rather, it imparts benefits conducive to bioavailability and delivery through its chemistry. The complete conjugate (i.e., siRNA-ACO) represents a novel modality for delivery of duplex RNA (e.g., siRNA) to the CNS that is currently being tested in the clinic for treatment of ALS.
Collapse
Affiliation(s)
- Chunling Duan
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | - Moorim Kang
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | - Xiaojie Pan
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | - Zubao Gan
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | - Vera Huang
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | - Guanlin Li
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
| | | | - Long-Cheng Li
- Ractigen Therapeutics, Nantong, Jiangsu Province, China
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| |
Collapse
|
10
|
Zhu H, Dalvi U, Cazenave W, Cattaert D, Branchereau P. Excitatory action of low frequency depolarizing GABA/glycine synaptic inputs is prevalent in prenatal spinal SOD1 G93A motoneurons. J Physiol 2024; 602:913-932. [PMID: 38345477 DOI: 10.1113/jp285105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 01/24/2024] [Indexed: 03/09/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neurodegenerative disease characterized by progressive motor neuron degeneration and muscle paralysis. Recent evidence suggests the dysfunction of inhibitory signalling in ALS motor neurons. We have shown that embryonic day (E)17.5 spinal motoneurons (MNs) of the SOD1G93A mouse model of ALS exhibit an altered chloride homeostasis. At this prenatal stage, inhibition of spinal motoneurons (MNs) is mediated by depolarizing GABAergic/glycinergic postsynaptic potentials (dGPSPs). Here, using an ex vivo preparation and patch clamp recording from MNs with a chloride equilibrium set below spike threshold, we report that low input resistance (Rin ) E17.5 MNs from the SOD1G93A ALS mouse model do not correctly integrate dGPSPs evoked by electrical stimulations of GABA/glycine inputs at different frequencies. Indeed, firing activity of most wild-type (WT) MNs with low Rin was inhibited by incoming dGPSPs, whereas low Rin SOD1G93A MNs were excited or exhibited a dual response (excited by low frequency dGPSPs and inhibited by high frequency dGPSPs). Simulation highlighted the importance of the GABA/glycine input density and showed that pure excitation could be obtained in SOD-like MNs by moving GABA/glycine input away from the cell body to dendrites. This was in agreement with confocal imaging showing a lack of peri-somatic inhibitory terminals in SOD1G93A MNs compared to WT littermates. Putative fast ALS-vulnerable MNs with low Rin are therefore lacking functional inhibition at the near-term prenatal stage. KEY POINTS: We analysed the integration of GABAergic/glycinergic synaptic events by embryonic spinal motoneurons (MNs) in a mouse model of the amyotrophic lateral sclerosis (ALS) neurodegenerative disease. We found that GABAergic/glycinergic synaptic events do not properly inhibit ALS MNs with low input resistance, most probably corresponding to future vulnerable MNs. We used a neuron model to highlight the importance of the GABA/glycine terminal location and density in the integration of the GABAergic/glycinergic synaptic events. Confocal imaging showed a lack of GABA/glycine terminals on the cell body of ALS MNs. The present study suggests that putative ALS vulnerable MNs with low Rin lack functional inhibition at the near-term stage.
Collapse
Affiliation(s)
- Hongmei Zhu
- University Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
| | - Urvashi Dalvi
- University Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
| | | | - Daniel Cattaert
- University Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
| | | |
Collapse
|
11
|
Liu ML, Ma S, Tai W, Zhong X, Ni H, Zou Y, Wang J, Zhang CL. Screens in aging-relevant human ALS-motor neurons identify MAP4Ks as therapeutic targets for the disease. Cell Death Dis 2024; 15:4. [PMID: 38177100 PMCID: PMC10766628 DOI: 10.1038/s41419-023-06395-7] [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: 09/01/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/06/2024]
Abstract
Effective therapeutics is much needed for amyotrophic lateral sclerosis (ALS), an adult-onset neurodegenerative disease mainly affecting motor neurons. By screening chemical compounds in human patient-derived and aging-relevant motor neurons, we identify a neuroprotective compound and show that MAP4Ks may serve as therapeutic targets for treating ALS. The lead compound broadly improves survival and function of motor neurons directly converted from human ALS patients. Mechanistically, it works as an inhibitor of MAP4Ks, regulates the MAP4Ks-HDAC6-TUBA4A-RANGAP1 pathway, and normalizes subcellular distribution of RANGAP1 and TDP-43. Finally, in an ALS mouse model we show that inhibiting MAP4Ks preserves motor neurons and significantly extends animal lifespan.
Collapse
Affiliation(s)
- Meng-Lu Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shuaipeng Ma
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Wenjiao Tai
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xiaoling Zhong
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Haoqi Ni
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yuhua Zou
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jingcheng Wang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
12
|
Tsioras K, Smith KC, Edassery SL, Garjani M, Li Y, Williams C, McKenna ED, Guo W, Wilen AP, Hark TJ, Marklund SL, Ostrow LW, Gilthorpe JD, Ichida JK, Kalb RG, Savas JN, Kiskinis E. Analysis of proteome-wide degradation dynamics in ALS SOD1 iPSC-derived patient neurons reveals disrupted VCP homeostasis. Cell Rep 2023; 42:113160. [PMID: 37776851 PMCID: PMC10785776 DOI: 10.1016/j.celrep.2023.113160] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/18/2023] [Accepted: 09/06/2023] [Indexed: 10/02/2023] Open
Abstract
Mutations in SOD1 cause amyotrophic lateral sclerosis (ALS) through gain-of-function effects, yet the mechanisms by which misfolded mutant SOD1 (mutSOD1) protein impairs human motor neurons (MNs) remain unclear. Here, we use induced-pluripotent-stem-cell-derived MNs coupled to metabolic stable isotope labeling and mass spectrometry to investigate proteome-wide degradation dynamics. We find several proteins, including the ALS-causal valosin-containing protein (VCP), which predominantly acts in proteasome degradation and autophagy, that degrade slower in mutSOD1 relative to isogenic control MNs. The interactome of VCP is altered in mutSOD1 MNs in vitro, while VCP selectively accumulates in the affected motor cortex of ALS-SOD1 patients. Overexpression of VCP rescues mutSOD1 toxicity in MNs in vitro and in a C. elegans model in vivo, in part due to its ability to modulate the degradation of insoluble mutSOD1. Our results demonstrate that VCP contributes to mutSOD1-dependent degeneration, link two distinct ALS-causal genes, and highlight selective protein degradation impairment in ALS pathophysiology.
Collapse
Affiliation(s)
- Konstantinos Tsioras
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kevin C Smith
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Seby L Edassery
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mehraveh Garjani
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yichen Li
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Chloe Williams
- Department of Integrative Medical Biology, Umeå University, 90187 Umeå, Sweden
| | - Elizabeth D McKenna
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Wenxuan Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Anika P Wilen
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Timothy J Hark
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Stefan L Marklund
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Lyle W Ostrow
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | | | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Robert G Kalb
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jeffrey N Savas
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
13
|
Hernández S, Salvany S, Casanovas A, Piedrafita L, Soto-Bernardini MC, Tarabal O, Blasco A, Gras S, Gatius A, Schwab MH, Calderó J, Esquerda JE. Persistent NRG1 Type III Overexpression in Spinal Motor Neurons Has No Therapeutic Effect on ALS-Related Pathology in SOD1 G93A Mice. Neurotherapeutics 2023; 20:1820-1834. [PMID: 37733208 PMCID: PMC10684470 DOI: 10.1007/s13311-023-01424-x] [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] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease affecting upper and lower motor neurons (MNs). Neuregulin-1 (NRG1) is a pleiotropic growth factor that has been shown to be potentially valuable for ALS when supplemented by means of viral-mediated gene therapy. However, these results are inconsistent with other reports. An alternative approach for investigating the therapeutic impact of NRG1 on ALS is the use of transgenic mouse lines with genetically defined NRG1 overexpression. Here, we took advantage of a mouse line with NRG1 type III overexpression in spinal cord α motor neurons (MN) to determine the impact of steadily enhanced NRG1 signalling on mutant superoxide dismutase 1 (SOD1)-induced disease. The phenotype of SOD1G93A-NRG1 double transgenic mice was analysed in detail, including neuropathology and extensive behavioural testing. At least 3 animals per condition and sex were histopathologically assessed, and a minimum of 10 mice per condition and sex were clinically evaluated. The accumulation of misfolded SOD1 (mfSOD1), MN degeneration, and a glia-mediated neuroinflammatory response are pathological hallmarks of ALS progression in SOD1G93A mice. None of these aspects was significantly improved when examined in double transgenic NRG1-SOD1G93A mice. In addition, behavioural testing revealed that NRG1 type III overexpression did not affect the survival of SOD1G93A mice but accelerated disease onset and worsened the motor phenotype.
Collapse
Affiliation(s)
- Sara Hernández
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Sara Salvany
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - M Clara Soto-Bernardini
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
- Center for Research in Biotechnology (CIB), Costa Rica Institute of Technology (TEC), Cartago, Costa Rica
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Sílvia Gras
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Alaó Gatius
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Markus H Schwab
- Paul Flechsig Institute of Neuropathology, University Hospital Leipzig, Leipzig, Germany
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Josep E Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain.
| |
Collapse
|
14
|
Teli P, Kale V, Vaidya A. Beyond animal models: revolutionizing neurodegenerative disease modeling using 3D in vitro organoids, microfluidic chips, and bioprinting. Cell Tissue Res 2023; 394:75-91. [PMID: 37572163 DOI: 10.1007/s00441-023-03821-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/27/2023] [Indexed: 08/14/2023]
Abstract
Neurodegenerative diseases (NDs) are characterized by uncontrolled loss of neuronal cells leading to a progressive deterioration of brain functions. The transition rate of numerous neuroprotective drugs against Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease, leading to FDA approval, is only 8-14% in the last two decades. Thus, in spite of encouraging preclinical results, these drugs have failed in human clinical trials, demonstrating that traditional cell cultures and animal models cannot accurately replicate human pathophysiology. Hence, in vitro three-dimensional (3D) models have been developed to bridge the gap between human and animal studies. Such technological advancements in 3D culture systems, such as human-induced pluripotent stem cell (iPSC)-derived cells/organoids, organ-on-a-chip technique, and 3D bioprinting, have aided our understanding of the pathophysiology and underlying mechanisms of human NDs. Despite these recent advances, we still lack a 3D model that recapitulates all the key aspects of NDs, thus making it difficult to study the ND's etiology in-depth. Hence in this review, we propose developing a combinatorial approach that allows the integration of patient-derived iPSCs/organoids with 3D bioprinting and organ-on-a-chip technique as it would encompass the neuronal cells along with their niche. Such a 3D combinatorial approach would characterize pathological processes thoroughly, making them better suited for high-throughput drug screening and developing effective novel therapeutics targeting NDs.
Collapse
Affiliation(s)
- Prajakta Teli
- Symbiosis International (Deemed University), Symbiosis School of Biological Sciences, Pune, 412115, India
- Symbiosis International (Deemed University), Symbiosis Center for Stem Cell Research, Pune, 412115, India
| | - Vaijayanti Kale
- Symbiosis International (Deemed University), Symbiosis School of Biological Sciences, Pune, 412115, India
- Symbiosis International (Deemed University), Symbiosis Center for Stem Cell Research, Pune, 412115, India
| | - Anuradha Vaidya
- Symbiosis International (Deemed University), Symbiosis School of Biological Sciences, Pune, 412115, India.
- Symbiosis International (Deemed University), Symbiosis Center for Stem Cell Research, Pune, 412115, India.
| |
Collapse
|
15
|
Liu ML, Ma S, Tai W, Zhong X, Ni H, Zou Y, Wang J, Zhang CL. Chemical screens in aging-relevant human motor neurons identify MAP4Ks as therapeutic targets for amyotrophic lateral sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.24.538014. [PMID: 37162962 PMCID: PMC10168247 DOI: 10.1101/2023.04.24.538014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Effective therapeutics is much needed for amyotrophic lateral sclerosis (ALS), an adult-onset neurodegenerative disease mainly affecting motor neurons. By screening chemical compounds in human patient-derived and aging-relevant motor neurons, we identify a neuroprotective compound and show that MAP4Ks may serve as therapeutic targets for treating ALS. The lead compound broadly improves survival and function of motor neurons directly converted from human ALS patients. Mechanistically, it works as an inhibitor of MAP4Ks, regulates the MAP4Ks-HDAC6-TUBA4A-RANGAP1 pathway, and normalizes subcellular distribution of RANGAP1 and TDP-43. Finally, in an ALS mouse model we show that inhibiting MAP4Ks preserves motor neurons and significantly extends animal lifespan.
Collapse
Affiliation(s)
- Meng-Lu Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shuaipeng Ma
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wenjiao Tai
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaoling Zhong
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Haoqi Ni
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yuhua Zou
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jingcheng Wang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
16
|
Cho E, Woo Y, Suh Y, Suh BK, Kim SJ, Nhung TTM, Yoo JY, Nghi TD, Lee SB, Mun DJ, Park SK. Ratiometric measurement of MAM Ca 2+ dynamics using a modified CalfluxVTN. Nat Commun 2023; 14:3586. [PMID: 37328454 PMCID: PMC10276021 DOI: 10.1038/s41467-023-39343-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/06/2023] [Indexed: 06/18/2023] Open
Abstract
Mitochondria-associated ER membrane (MAM) is a structure where these calcium-regulating organelles form close physical contact sites for efficient Ca2+ crosstalk. Despite the central importance of MAM Ca2+ dynamics in diverse biological processes, directly and specifically measuring Ca2+ concentrations inside MAM is technically challenging. Here, we develop MAM-Calflux, a MAM-specific BRET-based Ca2+ indicator. The successful application of the bimolecular fluorescence complementation (BiFC) concept highlights Ca2+-responsive BRET signals in MAM. The BiFC strategy imparts dual functionality as a Ca2+ indicator and quantitative structural marker specific for MAM. As a ratiometric Ca2+ indicator, MAM-Calflux estimates steady-state MAM Ca2+ levels. Finally, it enables the visualization of uneven intracellular distribution of MAM Ca2+ and the elucidation of abnormally accumulated MAM Ca2+ from the neurons of Parkinson's disease mouse model in both steady-state and stimulated conditions. Therefore, we propose that MAM-Calflux can be a versatile tool for ratiometrically measuring dynamic inter-organellar Ca2+ communication.
Collapse
Affiliation(s)
- Eunbyul Cho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Youngsik Woo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| | - Yeongjun Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Bo Kyoung Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Soo Jeong Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Truong Thi My Nhung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Jin Yeong Yoo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Tran Diem Nghi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Su Been Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Dong Jin Mun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| |
Collapse
|
17
|
Dominov JA, Madigan LA, Whitt JP, Rademacher KL, Webster KM, Zhang H, Banno H, Tang S, Zhang Y, Wightman N, Shychuck EM, Page J, Weiss A, Kelly K, Kucukural A, Brodsky MH, Jaworski A, Fallon JR, Lipscombe D, Brown RH. Up-regulation of cholesterol synthesis pathways and limited neurodegeneration in a knock-in Sod1 mutant mouse model of ALS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539444. [PMID: 37205335 PMCID: PMC10187330 DOI: 10.1101/2023.05.05.539444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disorder affecting brain and spinal cord motor neurons. Mutations in the copper/zinc superoxide dismutase gene ( SOD1 ) are associated with ∼20% of inherited and 1-2% of sporadic ALS cases. Much has been learned from mice expressing transgenic copies of mutant SOD1, which typically involve high-level transgene expression, thereby differing from ALS patients expressing one mutant gene copy. To generate a model that more closely represents patient gene expression, we created a knock-in point mutation (G85R, a human ALS-causing mutation) in the endogenous mouse Sod1 gene, leading to mutant SOD1 G85R protein expression. Heterozygous Sod1 G85R mutant mice resemble wild type, whereas homozygous mutants have reduced body weight and lifespan, a mild neurodegenerative phenotype, and express very low mutant SOD1 protein levels with no detectable SOD1 activity. Homozygous mutants exhibit partial neuromuscular junction denervation at 3-4 months of age. Spinal cord motor neuron transcriptome analyses of homozygous Sod1 G85R mice revealed up-regulation of cholesterol synthesis pathway genes compared to wild type. Transcriptome and phenotypic features of these mice are similar to Sod1 knock-out mice, suggesting the Sod1 G85R phenotype is largely driven by loss of SOD1 function. By contrast, cholesterol synthesis genes are down-regulated in severely affected human TgSOD1 G93A transgenic mice at 4 months. Our analyses implicate dysregulation of cholesterol or related lipid pathway genes in ALS pathogenesis. The Sod1 G85R knock-in mouse is a useful ALS model to examine the importance of SOD1 activity in control of cholesterol homeostasis and motor neuron survival. SIGNIFICANCE STATEMENT Amyotrophic lateral sclerosis is a devastating disease involving the progressive loss of motor neurons and motor function for which there is currently no cure. Understanding biological mechanisms leading to motor neuron death is critical for developing new treatments. Using a new knock-in mutant mouse model carrying a Sod1 mutation that causes ALS in patients, and in the mouse, causes a limited neurodegenerative phenotype similar to Sod1 loss-of-function, we show that cholesterol synthesis pathway genes are up-regulated in mutant motor neurons, whereas the same genes are down-regulated in transgenic SOD1 mice with a severe phenotype. Our data implicate dysregulation of cholesterol or other related lipid genes in ALS pathogenesis and provide new insights that could contribute to strategies for disease intervention.
Collapse
|
18
|
Bak AN, Djukic S, Kadlecova M, Braunstein TH, Jensen DB, Meehan CF. Cytoplasmic TDP-43 accumulation drives changes in C-bouton number and size in a mouse model of sporadic Amyotrophic Lateral Sclerosis. Mol Cell Neurosci 2023; 125:103840. [PMID: 36921783 DOI: 10.1016/j.mcn.2023.103840] [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: 10/15/2022] [Revised: 02/11/2023] [Accepted: 03/04/2023] [Indexed: 03/14/2023] Open
Abstract
An altered neuronal excitability of spinal motoneurones has consistently been implicated in Amyotrophic Lateral Sclerosis (ALS) leading to several investigations of synaptic input to these motoneurones. One such input that has repeatedly been shown to be affected is a population of large cholinergic synapses terminating mainly on the soma of the motoneurones referred to as C-boutons. Most research on these synapses during disease progression has used transgenic Superoxide Dismutase 1 (SOD1) mouse models of the disease which have not only produced conflicting findings, but also fail to recapitulate the key pathological feature seen in ALS; cytoplasmic accumulations of TAR DNA-binding protein 43 (TDP-43). Additionally, they fail to distinguish between slow and fast motoneurones, the latter of which have more C-boutons, but are lost earlier in the disease. To circumvent these issues, we quantified the frequency and volume of C-boutons on traced soleus and gastrocnemius motoneurones, representing predominantly slow and fast motor pools respectively. Experiments were performed using the TDP-43ΔNLS mouse model that carries a transgenic construct of TDP-43 devoid of its nuclear localization signal, preventing its nuclear import. This results in the emergence of pathological TDP-43 inclusions in the cytoplasm, modelling the main pathology seen in this disorder, accompanied by a severe and lethal ALS phenotype. Our results confirmed changes in both the number and volume of C-boutons with a decrease in number on the more vulnerable, predominantly fast gastrocnemius motoneurones and an increase in number on the less vulnerable, predominantly slow soleus motoneurones. Importantly, these changes were only found in male mice. However, both sexes and motor pools showed a decrease in C-bouton volume. Our experiments confirm that cytoplasmic TDP-43 accumulation is sufficient to drive C-bouton changes.
Collapse
Affiliation(s)
| | - Svetlana Djukic
- Department of Neuroscience, University of Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
19
|
Role of the Intermediate Filament Protein Peripherin in Health and Disease. Int J Mol Sci 2022; 23:ijms232315416. [PMID: 36499746 PMCID: PMC9740141 DOI: 10.3390/ijms232315416] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/23/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Intermediate filaments are the most heterogeneous class among cytoskeletal elements. While some of them have been well-characterized, little is known about peripherin. Peripherin is a class III intermediate filament protein with a specific expression in the peripheral nervous system. Epigenetic modifications are involved in this cell-type-specific expression. Peripherin has important roles in neurite outgrowth and stability, axonal transport, and axonal myelination. Moreover, peripherin interacts with proteins involved in vesicular trafficking, signal transduction, DNA/RNA processing, protein folding, and mitochondrial metabolism, suggesting a role in all these processes. This review collects information regarding peripherin gene regulation, post-translational modifications, and functions and its involvement in the onset of a number of diseases.
Collapse
|
20
|
Campisi L, Chizari S, Ho JSY, Gromova A, Arnold FJ, Mosca L, Mei X, Fstkchyan Y, Torre D, Beharry C, Garcia-Forn M, Jiménez-Alcázar M, Korobeynikov VA, Prazich J, Fayad ZA, Seldin MM, De Rubeis S, Bennett CL, Ostrow LW, Lunetta C, Squatrito M, Byun M, Shneider NA, Jiang N, La Spada AR, Marazzi I. Clonally expanded CD8 T cells characterize amyotrophic lateral sclerosis-4. Nature 2022; 606:945-952. [PMID: 35732742 PMCID: PMC10089623 DOI: 10.1038/s41586-022-04844-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 05/09/2022] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a heterogenous neurodegenerative disorder that affects motor neurons and voluntary muscle control1. ALS heterogeneity includes the age of manifestation, the rate of progression and the anatomical sites of symptom onset. Disease-causing mutations in specific genes have been identified and define different subtypes of ALS1. Although several ALS-associated genes have been shown to affect immune functions2, whether specific immune features account for ALS heterogeneity is poorly understood. Amyotrophic lateral sclerosis-4 (ALS4) is characterized by juvenile onset and slow progression3. Patients with ALS4 show motor difficulties by the time that they are in their thirties, and most of them require devices to assist with walking by their fifties. ALS4 is caused by mutations in the senataxin gene (SETX). Here, using Setx knock-in mice that carry the ALS4-causative L389S mutation, we describe an immunological signature that consists of clonally expanded, terminally differentiated effector memory (TEMRA) CD8 T cells in the central nervous system and the blood of knock-in mice. Increased frequencies of antigen-specific CD8 T cells in knock-in mice mirror the progression of motor neuron disease and correlate with anti-glioma immunity. Furthermore, bone marrow transplantation experiments indicate that the immune system has a key role in ALS4 neurodegeneration. In patients with ALS4, clonally expanded TEMRA CD8 T cells circulate in the peripheral blood. Our results provide evidence of an antigen-specific CD8 T cell response in ALS4, which could be used to unravel disease mechanisms and as a potential biomarker of disease state.
Collapse
Affiliation(s)
- Laura Campisi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Shahab Chizari
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Jessica S Y Ho
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anastasia Gromova
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
- UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA, USA
| | - Frederick J Arnold
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
- UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA, USA
| | - Lorena Mosca
- Medical Genetics Unit, Department of Laboratory Medicine, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Xueyan Mei
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yesai Fstkchyan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Denis Torre
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cindy Beharry
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marta Garcia-Forn
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miguel Jiménez-Alcázar
- Seve Ballesteros Foundation Brain Tumor Group, Molecular Oncology Program, Spanish National Cancer Research Centre, Madrid, Spain
| | | | - Jack Prazich
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, University of California, Irvine, Irvine, CA, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Craig L Bennett
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
- UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA, USA
| | - Lyle W Ostrow
- Neuromuscular Division of the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christian Lunetta
- NEMO Clinical Center, Fondazione Serena Onlus, Milan, Italy
- Neurorehabilitation Department, Istituti Clinici Scientifici Maugeri, IRCCS, Milan, Italy
| | - Massimo Squatrito
- Seve Ballesteros Foundation Brain Tumor Group, Molecular Oncology Program, Spanish National Cancer Research Centre, Madrid, Spain
| | - Minji Byun
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Neil A Shneider
- Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA
| | - Ning Jiang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Albert R La Spada
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA.
- Department of Neurology, University of California, Irvine, Irvine, CA, USA.
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA.
- UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA, USA.
| | - Ivan Marazzi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
21
|
Kim S, Kim DK, Jeong S, Lee J. The Common Cellular Events in the Neurodegenerative Diseases and the Associated Role of Endoplasmic Reticulum Stress. Int J Mol Sci 2022; 23:5894. [PMID: 35682574 PMCID: PMC9180188 DOI: 10.3390/ijms23115894] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022] Open
Abstract
Neurodegenerative diseases are inseparably linked with aging and increase as life expectancy extends. There are common dysfunctions in various cellular events shared among neurogenerative diseases, such as calcium dyshomeostasis, neuroinflammation, and age-associated decline in the autophagy-lysosome system. However, most of all, the prominent pathological feature of neurodegenerative diseases is the toxic buildup of misfolded protein aggregates and inclusion bodies accompanied by an impairment in proteostasis. Recent studies have suggested a close association between endoplasmic reticulum (ER) stress and neurodegenerative pathology in cellular and animal models as well as in human patients. The contribution of mutant or misfolded protein-triggered ER stress and its associated signaling events, such as unfolded protein response (UPR), to the pathophysiology of various neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's disease, amyotrophic lateral sclerosis, and prion disease, is described here. Impaired UPR action is commonly attributed to exacerbated ER stress, pathogenic protein aggregate accumulation, and deteriorating neurodegenerative pathologies. Thus, activating certain UPR components has been shown to alleviate ER stress and its associated neurodegeneration. However, uncontrolled activation of some UPR factors has also been demonstrated to worsen neurodegenerative phenotypes, suggesting that detailed molecular mechanisms around ER stress and its related neurodegenerations should be understood to develop effective therapeutics against aging-associated neurological syndromes. We also discuss current therapeutic endeavors, such as the development of small molecules that selectively target individual UPR components and address ER stress in general.
Collapse
Affiliation(s)
- Soojeong Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
| | - Doo Kyung Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
| | - Seho Jeong
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| |
Collapse
|
22
|
Chin-Chan M, Montes S, Blanco-Álvarez VM, Aguirre-Alarcón HA, Hernández-Rodríguez I, Bautista E. Relevance of biometals during neuronal differentiation and myelination: in vitro and in vivo studies. Biometals 2022; 35:395-427. [DOI: 10.1007/s10534-022-00380-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 02/27/2022] [Indexed: 12/20/2022]
|
23
|
Yu CY, Chang HC. Glutamate signaling mediates C. elegans behavioral plasticity to pathogens. iScience 2022; 25:103919. [PMID: 35252815 PMCID: PMC8889136 DOI: 10.1016/j.isci.2022.103919] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Chun-Ying Yu
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, 62102, Taiwan
| | - Howard C. Chang
- Department of Cell Biology and Neuroscience, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
- Corresponding author
| |
Collapse
|
24
|
Petro TM, Agarkova IV, Esmael A, Dunigan DD, Van Etten JL, Pattee GL. Chlorovirus ATCV-1 Accelerates Motor Deterioration in SOD1-G93A Transgenic Mice and Its SOD1 Augments Induction of Inflammatory Factors From Murine Macrophages. Front Neurol 2022; 13:821166. [PMID: 35280283 PMCID: PMC8908019 DOI: 10.3389/fneur.2022.821166] [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: 11/23/2021] [Accepted: 01/31/2022] [Indexed: 11/25/2022] Open
Abstract
Background Genetically polymorphic Superoxide Dismutase 1 G93A (SOD1-G93A) underlies one form of familial Amyotrophic Lateral Sclerosis (ALS). Exposures from viruses may also contribute to ALS, possibly by stimulating immune factors, such as IL-6, Interferon Stimulated Genes, and Nitric Oxide. Recently, chlorovirus ATCV-1, which encodes a SOD1, was shown to replicate in macrophages and induce inflammatory factors. Objective This study aimed to determine if ATCV-1 influences development of motor degeneration in an ALS mouse model and to assess whether SOD1 of ATCV-1 influences production of inflammatory factors from macrophages. Methods Sera from sporadic ALS patients were screened for antibody to ATCV-1. Active or inactivated ATCV-1, saline, or a viral mimetic, polyinosinic:polycytidylic acid (poly I:C) were injected intracranially into transgenic mice expressing human SOD1-G93A- or C57Bl/6 mice. RAW264.7 mouse macrophage cells were transfected with a plasmid vector expressing ATCV-1 SOD1 or an empty vector prior to stimulation with poly I:C with or without Interferon-gamma (IFN-γ). Results Serum from sporadic ALS patients had significantly more IgG1 antibody directed against ATCV-1 than healthy controls. Infection of SOD1-G93A mice with active ATCV-1 significantly accelerated onset of motor loss, as measured by tail paralysis, hind limb tucking, righting reflex, and latency to fall in a hanging cage-lid test, but did not significantly affect mortality when compared to saline-treated transgenics. By contrast, poly I:C treatment significantly lengthened survival time but only minimally slowed onset of motor loss, while heat-inactivated ATCV-1 did not affect motor loss or survival. ATCV-1 SOD1 significantly increased expression of IL-6, IL-10, ISG promoter activity, and production of Nitric Oxide from RAW264.7 cells. Conclusion ATCV-1 chlorovirus encoding an endogenous SOD1 accelerates pathogenesis but not mortality, while poly I:C that stimulates antiviral immune responses delays mortality in an ALS mouse model. ATCV-1 SOD1 enhances induction of inflammatory factors from macrophages.
Collapse
Affiliation(s)
- Thomas M. Petro
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, United States
- Nebraska Center for Virology, University of Nebraska Lincoln, Lincoln, NE, United States
- *Correspondence: Thomas M. Petro
| | - Irina V. Agarkova
- Nebraska Center for Virology, University of Nebraska Lincoln, Lincoln, NE, United States
- Department Plant Pathology, University of Nebraska Lincoln, Lincoln, NE, United States
| | - Ahmed Esmael
- Nebraska Center for Virology, University of Nebraska Lincoln, Lincoln, NE, United States
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, Egypt
| | - David D. Dunigan
- Nebraska Center for Virology, University of Nebraska Lincoln, Lincoln, NE, United States
- Department Plant Pathology, University of Nebraska Lincoln, Lincoln, NE, United States
| | - James L. Van Etten
- Nebraska Center for Virology, University of Nebraska Lincoln, Lincoln, NE, United States
- Department Plant Pathology, University of Nebraska Lincoln, Lincoln, NE, United States
| | | |
Collapse
|
25
|
Tanaka M, Homma K, Soejima A. Histopathological changes of the spinal cord and motor neuron dynamics in SOD1 Tg mice. J Toxicol Pathol 2022; 35:129-133. [PMID: 35221507 PMCID: PMC8828614 DOI: 10.1293/tox.2021-0056] [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: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 11/23/2022] Open
Abstract
We analyzed the histopathological changes and the number of motor neurons (MNs) in the
lumbar spinal cord of Cu/Zn superoxide dismutase transgenic (SOD1G93ATg) mice,
which are frequently used as a disease model of amyotrophic lateral sclerosis (ALS). In
SOD1G93ATg mice, hyaline inclusions and foamy vacuoles in the neuronal cell
body were observed at 7 weeks of age before neurologic symptoms, and large vacuoles,
spheroid formation, and nerve cell aggregation became prominent after 13 weeks of age. The
number of healthy MNs was 28.7 to 37.1 cells/animal in wild-type mice and 9.3 to 13.6
cells/animal in transgenic (Tg) mice. Furthermore, the number of MNs, including
degenerative neurons, in Tg mice was 27.3–36.1 cells/animal at 18 weeks of age and
17.8–19.6 cells/animal at 21 weeks of age. The present results provide useful information
for the development of drugs in ALS treatment.
Collapse
Affiliation(s)
- Masaharu Tanaka
- Research Unit/Neuroscience, Sohyaku. Innovation Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama-shi 227-0033, Japan
| | - Kengo Homma
- Research Unit/Neuroscience, Sohyaku. Innovation Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama-shi 227-0033, Japan
| | - Aki Soejima
- Research Unit/Neuroscience, Sohyaku. Innovation Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama-shi 227-0033, Japan
| |
Collapse
|
26
|
Woo TG, Yoon MH, Kang SM, Park S, Cho JH, Hwang YJ, Ahn J, Jang H, Shin YJ, Jung EM, Ha NC, Kim BH, Kwon Y, Park BJ. Novel chemical inhibitor against SOD1 misfolding and aggregation protects neuron-loss and ameliorates disease symptoms in ALS mouse model. Commun Biol 2021; 4:1397. [PMID: 34912047 PMCID: PMC8674338 DOI: 10.1038/s42003-021-02862-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease characterized by selective death of motor neurons. Mutations in Cu, Zn-superoxide dismutase (SOD1) causing the gain of its toxic property are the major culprit of familial ALS (fALS). The abnormal SOD1 aggregation in the motor neurons has been suggested as the major pathological hallmark of ALS patients. However, the development of pharmacological interventions against SOD1 still needs further investigation. In this study, using ELISA-based chemical screening with wild and mutant SOD1 proteins, we screened a new small molecule, PRG-A01, which could block the misfolding/aggregation of SOD1 or TDP-43. The drug rescued the cell death induced by mutant SOD1 in human neuroblastoma cell line. Administration of PRG-A01 into the ALS model mouse resulted in significant improvement of muscle strength, motor neuron viability and mobility with extended lifespan. These results suggest that SOD1 misfolding/aggregation is a potent therapeutic target for SOD1 related ALS.
Collapse
Affiliation(s)
- Tae-Gyun Woo
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Min-Ho Yoon
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - So-Mi Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Soyoung Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Jung-Hyun Cho
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Young Jun Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Jinsook Ahn
- Department of Food Science, College of Agricultural and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyewon Jang
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Yun-Jeong Shin
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Eui-Man Jung
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Nam-Chul Ha
- Department of Food Science, College of Agricultural and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Bae-Hoon Kim
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea
| | - Yonghoon Kwon
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea.
| | - Bum-Joon Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea.
- Rare Disease R&D Center, PRG S&T Co., Ltd, Busan, Republic of Korea.
| |
Collapse
|
27
|
Shiraishi W, Yamasaki R, Hashimoto Y, Ko S, Kobayakawa Y, Isobe N, Matsushita T, Kira JI. Clearance of peripheral nerve misfolded mutant protein by infiltrated macrophages correlates with motor neuron disease progression. Sci Rep 2021; 11:16438. [PMID: 34385589 PMCID: PMC8360983 DOI: 10.1038/s41598-021-96064-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
Macrophages expressing C-C chemokine receptor type 2 (CCR2) infiltrate the central and peripheral neural tissues of amyotrophic lateral sclerosis (ALS) patients. To identify the functional role of CCR2+ macrophages in the pathomechanisms of ALS, we used an ALS animal model, mutant Cu/Zn superoxide dismutase 1G93A (mSOD1)-transgenic (Tg) mice. To clarify the CCR2 function in the model, we generated SOD1G93A/CCR2Red fluorescence protein (RFP)/Wild type (WT)/CX3CR1Green fluorescence protein (GFP)/WT-Tg mice, which heterozygously express CCR2-RFP and CX3CR1-GFP, and SOD1G93A/CCR2RFP/RFP-Tg mice, which lack CCR2 protein expression and present with a CCR2-deficient phenotype. In mSOD1-Tg mice, mSOD1 accumulated in the sciatic nerve earlier than in the spinal cord. Furthermore, spinal cords of SOD1G93A/CCR2RFP/WT/CX3CR1GFP/WT mice showed peripheral macrophage infiltration that emerged at the end-stage, whereas in peripheral nerves, macrophage infiltration started from the pre-symptomatic stage. Before disease onset, CCR2+ macrophages harboring mSOD1 infiltrated sciatic nerves earlier than the lumbar cord. CCR2-deficient mSOD1-Tg mice showed an earlier onset and axonal derangement in the sciatic nerve than CCR2-positive mSOD1-Tg mice. CCR2-deficient mSOD1-Tg mice showed an increase in deposited mSOD1 in the sciatic nerve compared with CCR2-positive mice. These findings suggest that CCR2+ and CX3CR1+ macrophages exert neuroprotective functions in mSOD1 ALS via mSOD1 clearance from the peripheral nerves.
Collapse
Affiliation(s)
- Wataru Shiraishi
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan ,grid.415432.50000 0004 0377 9814Department of Neurology, Kokura Memorial Hospital, Fukuoka, 802-8555 Japan
| | - Ryo Yamasaki
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Yu Hashimoto
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Senri Ko
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Yuko Kobayakawa
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Noriko Isobe
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Takuya Matsushita
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Jun-ichi Kira
- grid.177174.30000 0001 2242 4849Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan ,grid.411731.10000 0004 0531 3030Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy At Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Ookawa, Fukuoka 831-8501 Japan ,grid.411731.10000 0004 0531 3030Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, 2-6-11 Yakuin, Chuo-ku, Fukuoka, 810-0022 Japan
| |
Collapse
|
28
|
Shadrach JL, Stansberry WM, Milen AM, Ives RE, Fogarty EA, Antonellis A, Pierchala BA. Translatomic analysis of regenerating and degenerating spinal motor neurons in injury and ALS. iScience 2021; 24:102700. [PMID: 34235408 PMCID: PMC8246596 DOI: 10.1016/j.isci.2021.102700] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/09/2020] [Accepted: 06/07/2021] [Indexed: 12/23/2022] Open
Abstract
The neuromuscular junction is a synapse critical for muscle strength and coordinated motor function. Unlike CNS injuries, motor neurons mount robust regenerative responses after peripheral nerve injuries. Conversely, motor neurons selectively degenerate in diseases such as amyotrophic lateral sclerosis (ALS). To assess how these insults affect motor neurons in vivo, we performed ribosomal profiling of mouse motor neurons. Motor neuron-specific transcripts were isolated from spinal cords following sciatic nerve crush, a model of acute injury and regeneration, and in the SOD1G93A ALS model. Of the 267 transcripts upregulated after nerve crush, 38% were also upregulated in SOD1G93A motor neurons. However, most upregulated genes in injured and ALS motor neurons were context specific. Some of the most significantly upregulated transcripts in both paradigms were chemokines such as Ccl2 and Ccl7, suggesting an important role for neuroimmune modulation. Collectively these data will aid in defining pro-regenerative and pro-degenerative mechanisms in motor neurons.
Collapse
Affiliation(s)
- Jennifer L. Shadrach
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Wesley M. Stansberry
- Department of Anatomy, Cell Biology & Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Allison M. Milen
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Rachel E. Ives
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, USA
| | | | - Anthony Antonellis
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Brian A. Pierchala
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Anatomy, Cell Biology & Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
29
|
Kim HN, Kim JY. A Systematic Review of Oropharyngeal Dysphagia Models in Rodents. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:4987. [PMID: 34067192 PMCID: PMC8125817 DOI: 10.3390/ijerph18094987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/24/2021] [Accepted: 05/04/2021] [Indexed: 11/16/2022]
Abstract
Oropharyngeal dysphagia is a condition characterized by swallowing difficulty in the mouth and pharynx, which can be due to various factors. Animal models of oropharyngeal dysphagia are essential to confirm the cause-specific symptoms, pathological findings, and the effect of treatment. Recently, various animal models of dysphagia have been reported. The purpose of this review is to organize the rodent models of oropharyngeal dysphagia reported to date. The articles were obtained from Medline, Embase, and the Cochrane library, and selected following the PRISMA guideline. The animal models in which oropharyngeal dysphagia was induced in rats or mice were selected and classified based on the diseases causing oropharyngeal dysphagia. The animal used, method of inducing dysphagia, and screening methods and results were collected from the selected 37 articles. Various rodent models of oropharyngeal dysphagia provide distinctive information on atypical swallowing. Applying and analyzing the treatment in rodent models of dysphagia induced from various causes is an essential process to develop symptom-specific treatments. Therefore, the results of this study provide fundamental and important data for selecting appropriate animal models to study dysphagia.
Collapse
Affiliation(s)
- Han-Na Kim
- Department of Dental Hygiene, College of Health and Medical Sciences, Cheongju University, Cheongju 28503, Korea;
| | - Ji-Youn Kim
- Department of Dental Hygiene, College of Health Science, Gachon University, Incheon 21936, Korea
| |
Collapse
|
30
|
Proietti D, Giordani L, De Bardi M, D’Ercole C, Lozanoska-Ochser B, Amadio S, Volonté C, Marinelli S, Muchir A, Bouché M, Borsellino G, Sacco A, Puri PL, Madaro L. Activation of skeletal muscle-resident glial cells upon nerve injury. JCI Insight 2021; 6:143469. [PMID: 33661767 PMCID: PMC8119188 DOI: 10.1172/jci.insight.143469] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/25/2021] [Indexed: 01/06/2023] Open
Abstract
Here, we report on the identification of Itga7-expressing muscle-resident glial cells activated by loss of neuromuscular junction (NMJ) integrity. Gene expression analysis at the bulk and single-cell level revealed that these cells are distinct from Itga7-expressing muscle satellite cells. We show that a selective activation and expansion of Itga7+ glial cells occur in response to muscle nerve lesion. Upon activation, muscle glial-derived progenies expressed neurotrophic genes, including nerve growth factor receptor, which enables their isolation by FACS. We show that activated muscle glial cells also expressed genes potentially implicated in extracellular matrix remodeling at NMJs. We found that tenascin C, which was highly expressed by muscle glial cells, activated upon nerve injury and preferentially localized to NMJ. Interestingly, we observed that the activation of muscle glial cells by acute nerve injury was reversible upon NMJ repair. By contrast, in a mouse model of ALS, in which NMJ degeneration is progressive, muscle glial cells steadily increased over the course of the disease. However, they exhibited an impaired neurotrophic activity, suggesting that pathogenic activation of glial cells may be implicated in ALS progression.
Collapse
Affiliation(s)
- Daisy Proietti
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | - Lorenzo Giordani
- Sorbonne Université, INSERM UMRS 974, Association Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | | | - Chiara D’Ercole
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | - Biliana Lozanoska-Ochser
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | | | - Cinzia Volonté
- IRCCS Fondazione Santa Lucia, Rome, Italy
- CNR, National Research Council, Institute for Systems Analysis and Computer Science, Rome, Italy
| | - Sara Marinelli
- CNR, National Research Council, Institute of Biochemistry and Cell Biology, Monterotondo Scalo, Rome, Italy
| | - Antoine Muchir
- Sorbonne Université, INSERM UMRS 974, Association Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Marina Bouché
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | | | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Luca Madaro
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| |
Collapse
|
31
|
Ryan ÉB, Yan J, Miller N, Dayanidhi S, Ma YC, Deng HX, Siddique T. Early death of ALS-linked CHCHD10-R15L transgenic mice with central nervous system, skeletal muscle, and cardiac pathology. iScience 2021; 24:102061. [PMID: 33659869 PMCID: PMC7890413 DOI: 10.1016/j.isci.2021.102061] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 08/27/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
Mutations in coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10) have been identified in patients suffering from various degenerative diseases including mitochondrial myopathy, spinal muscular atrophy Jokela type, frontotemporal dementia, and/or amyotrophic lateral sclerosis (ALS). The pathogenic mechanism underlying CHCHD10-linked divergent disorders remains largely unknown. Here we show that transgenic mice overexpressing an ALS-linked CHCHD10 p.R15L mutation leads to an abbreviated lifespan compared with CHCHD10-WT transgenic mice. The occurrence and severity of the phenotype correlates to transgene copy number. Central nervous system (CNS), skeletal muscle, and cardiac pathology is apparent in CHCHD10-R15L transgenic mice. Despite the pathology, CHCHD10-R15L transgenic mice perform comparably to control mice in motor behavioral tasks until very close to death. Although paralysis is not observed, these models provide insight into the pleiotropic nature of CHCHD10 and suggest a contribution of CNS, skeletal muscle, and cardiac pathology to CHCHD10 p.R15L-ALS pathogenesis.
Collapse
Affiliation(s)
- Éanna B. Ryan
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Tarry Building, Room 13-715, 303 East Chicago Avenue, Chicago, IL 60611, USA
- Northwestern University Interdepartmental Neuroscience Program, Chicago, IL, USA
| | - Jianhua Yan
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Tarry Building, Room 13-715, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Nimrod Miller
- Ann and Robert H. Lurie Children's Hospital of Chicago and Departments of Pediatrics, Neurology and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sudarshan Dayanidhi
- Shirley Ryan AbilityLab and Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yongchao C. Ma
- Ann and Robert H. Lurie Children's Hospital of Chicago and Departments of Pediatrics, Neurology and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Han-Xiang Deng
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Tarry Building, Room 13-715, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Teepu Siddique
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Tarry Building, Room 13-715, 303 East Chicago Avenue, Chicago, IL 60611, USA
- Northwestern University Interdepartmental Neuroscience Program, Chicago, IL, USA
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
32
|
Atkinson R, Leung J, Bender J, Kirkcaldie M, Vickers J, King A. TDP-43 mislocalization drives neurofilament changes in a novel model of TDP-43 proteinopathy. Dis Model Mech 2021; 14:dmm.047548. [PMID: 33408125 PMCID: PMC7888715 DOI: 10.1242/dmm.047548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022] Open
Abstract
Mislocalization of the TAR DNA-binding protein 43 (TDP-43) from the nucleus to the cytoplasm is a common feature of neurodegenerative conditions such as amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). The downstream in vivo cellular effects of this mislocalization are not well understood. To investigate the impact of mislocalized TDP-43 on neuronal cell bodies, axons and axonal terminals, we utilized the mouse visual system to create a new model of TDP-43 proteinopathy. Mouse (C57BL/6J) retinal ganglion cells (RGCs) were transduced with GFP-tagged human wildtype TDP-43 (hTDP-WT-GFP) and human TDP-43 with a mutation in the nuclear localization sequence (hTDP-ΔNLS-GFP), to cause TDP-43 mislocalization, with ∼60% transduction efficiency achieved. Expression of both hTDP-WT-GFP and hTDP-ΔNLS-GFP resulted in changes to neurofilament expression, with cytoplasmic TDP-43 being associated with significantly (p<0.05) increased neurofilament heavy expression in the cell soma, and both forms of altered TDP-43 leading to significantly (p<0.05) decreased numbers of neurofilament-positive axons within the optic nerve. Alterations to neurofilament proteins were associated with significantly (p<0.05) increased microglial density in the optic nerve and retina. Furthermore expression of hTDP-WT-GFP was associated with a significant (p<0.05) increase in pre-synaptic input into RGCs in the retina. The current study has developed a new model allowing detailed examination of alterations to TDP-43 and will contribute to the knowledge of TDP-43-mediated neuronal alterations and degeneration.
Collapse
Affiliation(s)
- Rachel Atkinson
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| | - Jacqueline Leung
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| | - James Bender
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| | - Matthew Kirkcaldie
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| | - James Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| | - Anna King
- Wicking Dementia Research and Education Centre, University of Tasmania, Medical Science Precinct, 17, Liverpool Street, Hobart, Tasmania, Australia 7000, Australia
| |
Collapse
|
33
|
Nakata K, Namiki M, Kobatake Y, Nishida H, Sakai H, Yamato O, Urushitani M, Maeda S, Kamishina H. Up-regulated spinal microRNAs induce aggregation of superoxide dismutase 1 protein in canine degenerative myelopathy. Res Vet Sci 2020; 135:479-485. [PMID: 33261827 DOI: 10.1016/j.rvsc.2020.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/01/2020] [Accepted: 11/22/2020] [Indexed: 11/19/2022]
Abstract
Canine degenerative myelopathy (DM) is a fatal progressive neurodegenerative disease. Mutations in the superoxide dismutase 1 (SOD1) gene have been shown to be the major risk factor for DM, and it is hypothesized that neural degeneration is caused by a "gain of toxic function" of mutant SOD1. In this study, the spinal cord microRNA (miRNA) profiles of DM-affected dogs were investigated to elucidate the pathomechanisms of DM. Quantification of 277 miRNAs identified three up-regulated miRNAs and 18 down-regulated miRNAs in the spinal cords of DM-affected dogs. Based on gene ontology analysis, the target cluster of up-regulated miRNAs was associated with protein expression or modification and cellular response, and that of down-regulated miRNAs was associated with tissue development. In these clusters, we focused on the mechanism of protein ubiquitination. Polyubiquitination assay demonstrated that canine SOD1 proteins were polyubiquitinated and degraded by proteasomes. Immunohistochemistry of the spinal cords of DM-affected dogs showed that mutant SOD1 aggregations were not ubiquitin immunopositive. Using cultured cells, co-transfection of canine SOD1 and up-regulated miRNA in DM-affected dogs demonstrated that miR-23a, miR-142 and miR-221 significantly increased the proportion of cells with mutant SOD1 aggregation. These results suggested that up-regulated miRNAs in the spinal cords of DM-affected dogs may inhibit ubiquitination of misfolded SOD1 protein and induce mutant SOD1 aggregations, leading to further progression of degenerative processes in the DM pathology.
Collapse
Affiliation(s)
- Kohei Nakata
- The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Mariko Namiki
- Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yui Kobatake
- Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Hidetaka Nishida
- Department of Veterinary Clinical Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58, Ohraikita, Izumisano, Osaka 598-8531, Japan
| | - Hiroki Sakai
- The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Faculty of Applied Biological Sciences and Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Osamu Yamato
- Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Makoto Urushitani
- Department of Neurology, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Sadatoshi Maeda
- The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Hiroaki Kamishina
- The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; Faculty of Applied Biological Sciences and Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
| |
Collapse
|
34
|
Badawi Y, Nishimune H. Impairment Mechanisms and Intervention Approaches for Aged Human Neuromuscular Junctions. Front Mol Neurosci 2020; 13:568426. [PMID: 33328881 PMCID: PMC7717980 DOI: 10.3389/fnmol.2020.568426] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
The neuromuscular junction (NMJ) is a chemical synapse formed between a presynaptic motor neuron and a postsynaptic muscle cell. NMJs in most vertebrate species share many essential features; however, some differences distinguish human NMJs from others. This review will describe the pre- and postsynaptic structures of human NMJs and compare them to NMJs of laboratory animals. We will focus on age-dependent declines in function and changes in the structure of human NMJs. Furthermore, we will describe insights into the aging process revealed from mouse models of accelerated aging. In addition, we will compare aging phenotypes to other human pathologies that cause impairments of pre- and postsynaptic structures at NMJs. Finally, we will discuss potential intervention approaches for attenuating age-related NMJ dysfunction and sarcopenia in humans.
Collapse
Affiliation(s)
- Yomna Badawi
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS, United States
| | - Hiroshi Nishimune
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS, United States.,Neurobiology of Aging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Japan
| |
Collapse
|
35
|
Castellanos-Montiel MJ, Chaineau M, Durcan TM. The Neglected Genes of ALS: Cytoskeletal Dynamics Impact Synaptic Degeneration in ALS. Front Cell Neurosci 2020; 14:594975. [PMID: 33281562 PMCID: PMC7691654 DOI: 10.3389/fncel.2020.594975] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that selectively affects motor neurons (MNs) of the cortex, brainstem, and spinal cord. Several genes have been linked to both familial (fALS) and sporadic (sALS) cases of ALS. Among all the ALS-related genes, a group of genes known to directly affect cytoskeletal dynamics (ALS2, DCTN1, PFN1, KIF5A, NF-L, NF-H, PRPH, SPAST, and TUBA4A) is of high importance for MN health and survival, considering that MNs are large polarized cells with axons that can reach up to 1 m in length. In particular, cytoskeletal dynamics facilitate the transport of organelles and molecules across the long axonal distances within the cell, playing a key role in synapse maintenance. The majority of ALS-related genes affecting cytoskeletal dynamics were identified within the past two decades, making it a new area to explore for ALS. The purpose of this review is to provide insights into ALS-associated cytoskeletal genes and outline how recent studies have pointed towards novel pathways that might be impacted in ALS. Further studies making use of extensive analysis models to look for true hits, the newest technologies such as CRIPSR/Cas9, human induced pluripotent stem cells (iPSCs) and axon sequencing, as well as the development of more transgenic animal models could potentially help to: differentiate the variants that truly act as a primary cause of the disease from the ones that act as risk factors or disease modifiers, identify potential interactions between two or more ALS-related genes in disease onset and progression and increase our understanding of the molecular mechanisms leading to cytoskeletal defects. Altogether, this information will give us a hint on the real contribution of the cytoskeletal ALS-related genes during this lethal disease.
Collapse
Affiliation(s)
| | - Mathilde Chaineau
- Early Drug Discovery Unit (EDDU), Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit (EDDU), Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| |
Collapse
|
36
|
Terashima T, Kobashi S, Watanabe Y, Nakanishi M, Honda N, Katagi M, Ohashi N, Kojima H. Enhancing the Therapeutic Efficacy of Bone Marrow-Derived Mononuclear Cells with Growth Factor-Expressing Mesenchymal Stem Cells for ALS in Mice. iScience 2020; 23:101764. [PMID: 33251493 PMCID: PMC7677706 DOI: 10.1016/j.isci.2020.101764] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/16/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022] Open
Abstract
Several treatments have been attempted in amyotrophic lateral sclerosis (ALS) animal models and patients. Recently, transplantation of bone marrow-derived mononuclear cells (MNCs) was investigated as a regenerative therapy for ALS, but satisfactory treatments remain to be established. To develop an effective treatment, we focused on mesenchymal stem cells (MSCs) expressing hepatocyte growth factor, glial cell line-derived neurotrophic factor, and insulin-like growth factor using human artificial chromosome vector (HAC-MSCs). Here, we demonstrated the transplantation of MNCs with HAC-MSCs in ALS mice. As per our results, the progression of motor dysfunction was significantly delayed, and their survival was prolonged dramatically. Additional analysis revealed preservation of motor neurons, suppression of gliosis, engraftment of numerous MNCs, and elevated chemotaxis-related cytokines in the spinal cord of treated mice. Therefore, growth factor-expressing MSCs enhance the therapeutic effects of bone marrow-derived MNCs for ALS and have a high potential as a novel cell therapy for patients with ALS. MNCs with growth factor-expressing MSCs is an effective cell therapy for ALS mice The MSCs enhance therapeutic effects by migration of MNCs into ALS mice spinal cord This cell therapy suppresses neuronal loss and gliosis in ALS mice spinal cord This cell therapy induces several cytokines expression in ALS mice spinal cord
Collapse
Affiliation(s)
- Tomoya Terashima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Shuhei Kobashi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Yasuhiro Watanabe
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8504, Japan
| | - Mami Nakanishi
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8504, Japan
| | - Naoto Honda
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8504, Japan
| | - Miwako Katagi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Natsuko Ohashi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Hideto Kojima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
37
|
Mathew B, Ruiz P, Dutta S, Entrekin JT, Zhang S, Patel KD, Simmons MS, Augelli-Szafran CE, Cowell RM, Suto MJ. Structure-activity relationship (SAR) studies of N-(3-methylpyridin-2-yl)-4-(pyridin-2-yl)thiazol-2-amine (SRI-22819) as NF-ҡB activators for the treatment of ALS. Eur J Med Chem 2020; 210:112952. [PMID: 33139114 DOI: 10.1016/j.ejmech.2020.112952] [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: 08/20/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 11/25/2022]
Abstract
ALS is a rare type of progressive neurological disease with unknown etiology. It results in the gradual degeneration and death of motor neurons responsible for controlling the voluntary muscles. Identification of mutations in the superoxide dismutase (SOD) 1 gene has been the most significant finding in ALS research. SOD1 abnormalities have been associated with both familial as well as sporadic ALS cases. SOD2 is a highly inducible SOD that performs in concurrence with SOD1 to detoxify ROS. Induction of SOD2 can be obtained through activation of NF-ҡBs. We previously reported that SRI-22819 increases NF-ҡB expression and activation in vitro, but it has poor ADME properties in general and has no oral bioavailability. Our initial studies were focused on direct modifications of SRI-22819. There were active compounds identified but no improvement in microsomal stability was observed. In this context, we focused on making more significant structural changes in the core of the molecule. Ataluren, an oxadiazole compound that promotes read-through and expression of dystrophin in patients with Duchenne muscular dystrophy, bears some structural similarity to SRI-22819. Thus, we synthesized a series of SRI-22819 and Ataluren (PTC124) hybrid compounds. Several compounds from this series exhibited improved activity, microsomal stability and lower calculated polar surface area (PSA). This manuscript describes the synthesis and biological evaluation of SRI-22819 analogs and its hybrid combination with Ataluren.
Collapse
Affiliation(s)
- Bini Mathew
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA.
| | - Pedro Ruiz
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA.
| | - Shilpa Dutta
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA.
| | - Jordan T Entrekin
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA
| | - Sixue Zhang
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA
| | - Kaval D Patel
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA
| | - Micah S Simmons
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA
| | - Corinne E Augelli-Szafran
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA.
| | - Rita M Cowell
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA.
| | - Mark J Suto
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL, 35205, USA.
| |
Collapse
|
38
|
Selective neuronal degeneration in MATR3 S85C knock-in mouse model of early-stage ALS. Nat Commun 2020; 11:5304. [PMID: 33082323 PMCID: PMC7576598 DOI: 10.1038/s41467-020-18949-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
A missense mutation, S85C, in the MATR3 gene is a genetic cause for amyotrophic lateral sclerosis (ALS). It is unclear how the S85C mutation affects MATR3 function and contributes to disease. Here, we develop a mouse model that harbors the S85C mutation in the endogenous Matr3 locus using the CRISPR/Cas9 system. MATR3 S85C knock-in mice recapitulate behavioral and neuropathological features of early-stage ALS including motor impairment, muscle atrophy, neuromuscular junction defects, Purkinje cell degeneration and neuroinflammation in the cerebellum and spinal cord. Our neuropathology data reveals a loss of MATR3 S85C protein in the cell bodies of Purkinje cells and motor neurons, suggesting that a decrease in functional MATR3 levels or loss of MATR3 function contributes to neuronal defects. Our findings demonstrate that the MATR3 S85C mouse model mimics aspects of early-stage ALS and would be a promising tool for future basic and preclinical research.
Collapse
|
39
|
ALS-Related Mutant SOD1 Aggregates Interfere with Mitophagy by Sequestering the Autophagy Receptor Optineurin. Int J Mol Sci 2020; 21:ijms21207525. [PMID: 33065963 PMCID: PMC7590160 DOI: 10.3390/ijms21207525] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the progressive demise of motor neurons. One of the causes of familial ALS is the mutation of the gene encoding superoxide dismutase 1 (SOD1), which leads to abnormal protein aggregates. How SOD1 aggregation drives ALS is still poorly understood. Recently, ALS pathogenesis has been functionally implicated in mitophagy, specifically the clearance of damaged mitochondria. Here, to understand this mechanism, we investigated the relationship between the mitophagy receptor optineurin and SOD1 aggregates. We found that mutant SOD1 (mSOD1) proteins associate with and then sequester optineurin, which is required to form the mitophagosomes, to aggregates in N2a cells. Optineurin recruitment into mSOD1 aggregates resulted in a reduced mitophagy flux. Furthermore, we observed that an exogenous augmentation of optineurin alleviated the cellular cytotoxicity induced by mSOD1. Taken together, these studies demonstrate that ALS-linked mutations in SOD1 interfere with the mitophagy process through optineurin sequestration, suggesting that the accumulation of damaged mitochondria may play a crucial role in the pathophysiological mechanisms contributing to ALS.
Collapse
|
40
|
Herrando-Grabulosa M, Gaja-Capdevila N, Vela JM, Navarro X. Sigma 1 receptor as a therapeutic target for amyotrophic lateral sclerosis. Br J Pharmacol 2020; 178:1336-1352. [PMID: 32761823 DOI: 10.1111/bph.15224] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/13/2020] [Accepted: 07/25/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult disease causing a progressive loss of upper and lower motoneurons, muscle paralysis and early death. ALS has a poor prognosis of 3-5 years after diagnosis with no effective cure. The aetiopathogenic mechanisms involved include glutamate excitotoxicity, oxidative stress, protein misfolding, mitochondrial alterations, disrupted axonal transport and inflammation. Sigma non-opioid intracellular receptor 1 (sigma 1 receptor) is a protein expressed in motoneurons, mainly found in the endoplasmic reticulum (ER) on the mitochondria-associated ER membrane (MAM) or in close contact with cholinergic postsynaptic sites. MAMs are sites that allow the assembly of several complexes implicated in essential survival cell functions. The sigma 1 receptor modulates essential mechanisms for motoneuron survival including excitotoxicity, calcium homeostasis, ER stress and mitochondrial dysfunction. This review updates sigma 1 receptor mechanisms and its alterations in ALS, focusing on MAM modulation, which may constitute a novel target for therapeutic strategies. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.
Collapse
Affiliation(s)
- Mireia Herrando-Grabulosa
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Núria Gaja-Capdevila
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - José M Vela
- Esteve Pharmaceuticals S.A., Drug Discovery and Preclinical Development, Barcelona, Spain
| | - Xavier Navarro
- Institute of Neurosciences, Department Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Institut Guttmann de Neurorehabilitació, Badalona, Spain
| |
Collapse
|
41
|
Baek KW, Jung YK, Kim JS, Park JS, Hah YS, Kim SJ, Yoo JI. Rodent Model of Muscular Atrophy for Sarcopenia Study. J Bone Metab 2020; 27:97-110. [PMID: 32572370 PMCID: PMC7297619 DOI: 10.11005/jbm.2020.27.2.97] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/15/2020] [Accepted: 05/16/2020] [Indexed: 12/25/2022] Open
Abstract
The hallmark symptom of sarcopenia is the loss of muscle mass and strength without the loss of overall body weight. Sarcopenia patients are likely to have worse clinical outcomes and higher mortality than do healthy individuals. The sarcopenia population shows an annual increase of ~0.8% in the population after age 50, and the prevalence rate is rapidly increasing with the recent worldwide aging trend. Based on International Classification of Diseases, Tenth Revision, a global classification of disease published by the World Health Organization, issued the disease code (M62.84) given to sarcopenia in 2016. Therefore, it is expected that the study of sarcopenia will be further activated based on the classification of disease codes in the aging society. Several epidemiological studies and meta-analyses have looked at the correlation between the prevalence of sarcopenia and several environmental factors. In addition, studies using cell lines and rodents have been done to understand the biological mechanism of sarcopenia. Laboratory rodent models are widely applicable in sarcopenia studies because of the advantages of time savings, cost saving, and various analytical applications that could not be used for human subjects. The rodent models that can be applied to the sarcopenia research are diverse, but a simple and fast method that can cause atrophy or aging is preferred. Therefore, we will introduce various methods of inducing muscular atrophy in rodent models to be applied to the study of sarcopenia.
Collapse
Affiliation(s)
- Kyung-Wan Baek
- Department of Physical Education, Gyeongsang National University, Jinju, Korea
- Department of Orthopaedic Surgery, Gyoengsang National University Hospital, Gyeongsang National University, Jinju, Korea
| | - Youn-Kwan Jung
- Biomedical Research Institute, Gyoengsang National University Hospital, Gyeongsang National University, Jinju, Korea
| | - Ji-Seok Kim
- Department of Physical Education, Gyeongsang National University, Jinju, Korea
| | - Jin Sung Park
- Department of Orthopaedic Surgery, Gyoengsang National University Hospital, Gyeongsang National University, Jinju, Korea
| | - Young-Sool Hah
- Biomedical Research Institute, Gyoengsang National University Hospital, Gyeongsang National University, Jinju, Korea
| | - So-Jeong Kim
- Department of Convergence of Medical Sciences, Gyeongsang National University, Jinju, Korea
| | - Jun-Il Yoo
- Department of Orthopaedic Surgery, Gyoengsang National University Hospital, Gyeongsang National University, Jinju, Korea
| |
Collapse
|
42
|
D’Amico AG, Maugeri G, Saccone S, Federico C, Cavallaro S, Reglodi D, D’Agata V. PACAP Modulates the Autophagy Process in an In Vitro Model of Amyotrophic Lateral Sclerosis. Int J Mol Sci 2020; 21:ijms21082943. [PMID: 32331311 PMCID: PMC7216177 DOI: 10.3390/ijms21082943] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/15/2020] [Accepted: 04/19/2020] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease of complex etiology leading to motor neuron degeneration. Many gene alterations cause this pathology, including mutation in Cu, Zn superoxide dismutase (SOD1), which leads to its gain of function. Mutant SOD1 proteins are prone to aberrant misfolding and create aggregates that impair autophagy. The hypoxic stress is strictly linked to the disease progression since it induces uncontrolled autophagy activation and the consequent high rates of cell death. Previously, we showed that pituitary adenylate cyclase-activating polypeptide (PACAP) exerts neurotrophic activity in cultured mSOD1 motor neurons exposed to serum deprivation. To date, no studies have examined whether the protective effect of PACAP on mSOD1 cells exposed to hypoxic insult is mediated through the regulation of the autophagy process. In the present study, we used the neuroblastoma-spinal cord-34 (NSC-34) cell line, stably expressing human wild type or mutant SOD1 G93A, to represent a well characterized in vitro model of a familial form of ALS. These cells were exposed to 100-µM desferrioxamine mesylate salt for 24h, to mimic the hypoxic stress affecting motor neurons during the disease progression. Our results showed that PACAP treatment significantly reduced cell death and hypoxia-induced mSOD1 accumulation by modulating the autophagy process in G93A motor neurons, as revealed by the decreased LC3II and the increased p62 levels, two autophagy indicators. These results were also confirmed by evaluating the vacuole formation detected through light chain 3 (LC3) immunofluorescence. Furthermore, the PACAP effects on autophagy seem to be mediated through the activation of the MAPK/ERK signaling pathway. Overall, our data demonstrated that PACAP exerts an ameliorative effect on the mSOD1 motor neuron viability by modulating a hypoxia-induced autophagy process through activation of MAPK/ERK signaling cascade.
Collapse
Affiliation(s)
- Agata Grazia D’Amico
- Department of Human Science and Promotion of quality of Life, San Raffaele Open University of Rome, Via di Val Cannuta, 247, 00166 Roma, Italy;
| | - Grazia Maugeri
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 87, 95123 Catania, Italy;
| | - Salvatore Saccone
- Section of Animal Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, 95123 Catania, Italy; (S.S.); (C.F.)
| | - Concetta Federico
- Section of Animal Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, 95123 Catania, Italy; (S.S.); (C.F.)
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation, Italian National Research Council, 95123 Catania, Italy;
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pecs Medical School, 7622 Pécs, Hungary;
| | - Velia D’Agata
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia, 87, 95123 Catania, Italy;
- Correspondence: ; Tel.: +39-095-378-2039; Fax: +39-095-378-2046
| |
Collapse
|
43
|
Mathew B, Ruiz P, Pathak V, Suto MJ. Development of novel small molecules for the treatment of ALS. Bioorg Med Chem Lett 2020; 30:126950. [PMID: 31928838 DOI: 10.1016/j.bmcl.2020.126950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/19/2019] [Accepted: 01/01/2020] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rare and progressive neurodegenerative disease with unknown etiology. It is caused by the degeneration of motor neurons responsible for controlling voluntary muscles. It has been reported that mutations in the superoxide dismutase (SOD) 1 gene can lead to ALS. SOD1 abnormalities have been identified in both familial, as well as sporadic ALS cases. SOD2 is a highly inducible SOD that works in conjunction with SOD1. SOD2 can be induced through activation of NF-κBs. We previously reported that the novel small molecule, SRI-22818, increases NF-κB expression and activation and SOD2 levels in vitro and has activity in vivo in the SOD1-G93A reference model of ALS. We report herein the synthesis and biological evaluation of SRI-22818 analogs.
Collapse
Affiliation(s)
- Bini Mathew
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205, USA
| | - Pedro Ruiz
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205, USA
| | - Vibha Pathak
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205, USA
| | - Mark J Suto
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205, USA.
| |
Collapse
|
44
|
Marcuzzo S, Terragni B, Bonanno S, Isaia D, Cavalcante P, Cappelletti C, Ciusani E, Rizzo A, Regalia G, Yoshimura N, Ugolini GS, Rasponi M, Bechi G, Mantegazza M, Mantegazza R, Bernasconi P, Minati L. Hyperexcitability in Cultured Cortical Neuron Networks from the G93A-SOD1 Amyotrophic Lateral Sclerosis Model Mouse and its Molecular Correlates. Neuroscience 2019; 416:88-99. [PMID: 31400485 DOI: 10.1016/j.neuroscience.2019.07.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 11/25/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting the corticospinal tract and leading to motor neuron death. According to a recent study, magnetic resonance imaging-visible changes suggestive of neurodegeneration seem absent in the motor cortex of G93A-SOD1 ALS mice. However, it has not yet been ascertained whether the cortical neural activity is intact, or alterations are present, perhaps even from an early stage. Here, cortical neurons from this model were isolated at post-natal day 1 and cultured on multielectrode arrays. Their activity was studied with a comprehensive pool of neurophysiological analyses probing excitability, criticality and network architecture, alongside immunocytochemistry and molecular investigations. Significant hyperexcitability was visible through increased network firing rate and bursting, whereas topological changes in the synchronization patterns were apparently absent. The number of dendritic spines was increased, accompanied by elevated transcriptional levels of the DLG4 gene, NMDA receptor 1 and the early pro-apoptotic APAF1 gene. The extracellular Na+, Ca2+, K+ and Cl- concentrations were elevated, pointing to perturbations in the culture micro-environment. Our findings highlight remarkable early changes in ALS cortical neuron activity and physiology. These changes suggest that the causative factors of hyperexcitability and associated toxicity could become established much earlier than the appearance of disease symptoms, with implications for the discovery of new hypothetical therapeutic targets.
Collapse
Affiliation(s)
- Stefania Marcuzzo
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy.
| | - Benedetta Terragni
- Neurophysiopathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Silvia Bonanno
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Davide Isaia
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS UMR7104, INSERM U964, Université de Strasbourg, 67404 Illkirch CU, Strasbourg, France
| | - Paola Cavalcante
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Cristina Cappelletti
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Emilio Ciusani
- Laboratory of Clinical Pathology and Medical Genetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Ambra Rizzo
- Laboratory of Clinical Pathology and Medical Genetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Giulia Regalia
- Neuroengineering and Medical Robotics Laboratory, Politecnico di Milano, Milan 20133, Italy; Currently working at Empatica srl, Milan 20144, Italy
| | - Natsue Yoshimura
- World Research Hub Initiative (WRHI), Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Giovanni Stefano Ugolini
- Department of Electronics, Information & Bioengineering, Politecnico di Milano, Milan 20133, Italy
| | - Marco Rasponi
- Department of Electronics, Information & Bioengineering, Politecnico di Milano, Milan 20133, Italy
| | - Giulia Bechi
- Neurophysiopathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Massimo Mantegazza
- Neurophysiopathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy; Université Côte d'Azur, CNRS UMR7275, LabEx ICST, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne-Sophia Antipolis, France
| | - Renato Mantegazza
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Pia Bernasconi
- Neurology IV -Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Ludovico Minati
- World Research Hub Initiative (WRHI), Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan; Complex Systems Theory Department, Institute of Nuclear Physics, Polish Academy of Sciences (IFJ-PAN), 31-342 Kraków, Poland; Center for Mind/Brain Sciences (CIMeC), University of Trento, 38123 Trento, Italy
| |
Collapse
|
45
|
Park JH, Elpers C, Reunert J, McCormick ML, Mohr J, Biskup S, Schwartz O, Rust S, Grüneberg M, Seelhöfer A, Schara U, Boltshauser E, Spitz DR, Marquardt T. SOD1 deficiency: a novel syndrome distinct from amyotrophic lateral sclerosis. Brain 2019; 142:2230-2237. [PMID: 31332433 PMCID: PMC6658856 DOI: 10.1093/brain/awz182] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/22/2019] [Accepted: 04/28/2019] [Indexed: 11/13/2022] Open
Abstract
Superoxide dismutase 1 (SOD1) is the principal cytoplasmic superoxide dismutase in humans and plays a major role in redox potential regulation. It catalyses the transformation of the superoxide anion (O2•-) into hydrogen peroxide. Heterozygous variants in SOD1 are a common cause of familial amyotrophic lateral sclerosis. In this study we describe the homozygous truncating variant c.335dupG (p.C112Wfs*11) in SOD1 that leads to total absence of enzyme activity. The resulting phenotype is severe and marked by progressive loss of motor abilities, tetraspasticity with predominance in the lower extremities, mild cerebellar atrophy, and hyperekplexia-like symptoms. Heterozygous carriers have a markedly reduced enzyme activity when compared to wild-type controls but show no overt neurologic phenotype. These results are in contrast with the previously proposed theory that a loss of function is the underlying mechanism in SOD1-related motor neuron disease and should be considered before application of previously proposed SOD1 silencing as a treatment option for amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Julien H Park
- Department of General Paediatrics, University of Münster, Münster, Germany
| | - Christiane Elpers
- Department of General Paediatrics, University of Münster, Münster, Germany
| | - Janine Reunert
- Department of General Paediatrics, University of Münster, Münster, Germany
| | - Michael L McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa, USA
| | - Julia Mohr
- CeGaT GmbH und Praxis für Humangenetik Tübingen, Tübingen, Germany
| | - Saskia Biskup
- CeGaT GmbH und Praxis für Humangenetik Tübingen, Tübingen, Germany
| | - Oliver Schwartz
- Department of General Paediatrics, University of Münster, Münster, Germany
| | - Stephan Rust
- Department of General Paediatrics, University of Münster, Münster, Germany
| | - Marianne Grüneberg
- Department of General Paediatrics, University of Münster, Münster, Germany
| | - Anja Seelhöfer
- Department of General Paediatrics, University of Münster, Münster, Germany
| | - Ulrike Schara
- Department of Paediatric Neurology, University Hospital Essen, Essen, Germany
| | - Eugen Boltshauser
- Department of Paediatric Neurology, University Children’s Hospital, Zürich, Switzerland
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa, USA
| | - Thorsten Marquardt
- Department of General Paediatrics, University of Münster, Münster, Germany
| |
Collapse
|
46
|
Osman KL, Kohlberg S, Mok A, Brooks R, Lind LA, McCormack K, Ferreira A, Kadosh M, Fagan MK, Bearce E, Nichols NL, Coates JR, Lever TE. Optimizing the Translational Value of Mouse Models of ALS for Dysphagia Therapeutic Discovery. Dysphagia 2019; 35:343-359. [PMID: 31300881 DOI: 10.1007/s00455-019-10034-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/10/2019] [Accepted: 07/02/2019] [Indexed: 01/03/2023]
Abstract
The goal of this study was to compare dysphagia phenotypes in low and high copy number (LCN and HCN) transgenic superoxide dismutase 1 (SOD1) mouse models of ALS to accelerate the discovery of novel and effective treatments for dysphagia and early amyotrophic lateral sclerosis (ALS) diagnosis. Clinicopathological features of dysphagia were characterized in individual transgenic mice and age-matched controls utilizing videofluoroscopy in conjunction with postmortem assays of the tongue and hypoglossal nucleus. Quantitative PCR accurately differentiated HCN-SOD1 and LCN-SOD1 mice and nontransgenic controls. All HCN-SOD1 mice developed stereotypical paralysis in both hindlimbs. In contrast, LCN-SOD1 mice displayed wide variability in fore- and hindlimb involvement. Lick rate, swallow rate, inter-swallow interval, and pharyngeal transit time were significantly altered in both HCN-SOD1 and LCN-SOD1 mice compared to controls. Tongue weight, tongue dorsum surface area, total tongue length, and caudal tongue length were significantly reduced only in the LCN-SOD1 mice compared to age-matched controls. LCN-SOD1 mice with lower body weights had smaller/lighter weight tongues, and those with forelimb paralysis and slower lick rates died at a younger age. LCN-SOD1 mice had a 32% loss of hypoglossal neurons, which differed significantly when compared to age-matched control mice. These novel findings for LCN-SOD1 mice are congruent with reported dysphagia and associated tongue atrophy and hypoglossal nucleus pathology in human ALS patients, thus highlighting the translational potential of this mouse model in ALS research.
Collapse
Affiliation(s)
- Kate L Osman
- Department of Otolaryngology - Head and Neck Surgery, University of Missouri School of Medicine, One Hospital Dr. MA314, Columbia, MO, 65212, USA
| | - Sabrina Kohlberg
- Department of Otolaryngology - Head and Neck Surgery, University of Missouri School of Medicine, One Hospital Dr. MA314, Columbia, MO, 65212, USA
| | - Alexis Mok
- Department of Otolaryngology - Head and Neck Surgery, University of Missouri School of Medicine, One Hospital Dr. MA314, Columbia, MO, 65212, USA
| | - Ryan Brooks
- Department of Otolaryngology - Head and Neck Surgery, University of Missouri School of Medicine, One Hospital Dr. MA314, Columbia, MO, 65212, USA
| | - Lori A Lind
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| | - Katelyn McCormack
- Department of Otolaryngology - Head and Neck Surgery, University of Missouri School of Medicine, One Hospital Dr. MA314, Columbia, MO, 65212, USA
| | - Andries Ferreira
- Department of Otolaryngology - Head and Neck Surgery, University of Missouri School of Medicine, One Hospital Dr. MA314, Columbia, MO, 65212, USA
| | - Matan Kadosh
- Department of Otolaryngology - Head and Neck Surgery, University of Missouri School of Medicine, One Hospital Dr. MA314, Columbia, MO, 65212, USA
| | - Mary K Fagan
- Department of Communication Science and Disorders, University of Missouri School of Health Professions, Columbia, MO, USA
| | - Elizabeth Bearce
- Department of Otolaryngology - Head and Neck Surgery, University of Missouri School of Medicine, One Hospital Dr. MA314, Columbia, MO, 65212, USA
| | - Nicole L Nichols
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| | - Joan R Coates
- Department of Veterinary Medicine and Surgery, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| | - Teresa E Lever
- Department of Otolaryngology - Head and Neck Surgery, University of Missouri School of Medicine, One Hospital Dr. MA314, Columbia, MO, 65212, USA. .,Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, MO, USA. .,Department of Communication Science and Disorders, University of Missouri School of Health Professions, Columbia, MO, USA.
| |
Collapse
|
47
|
Lee SH, Kim S, Lee N, Lee J, Yu SS, Kim JH, Kim S. Intrathecal delivery of recombinant AAV1 encoding hepatocyte growth factor improves motor functions and protects neuromuscular system in the nerve crush and SOD1-G93A transgenic mouse models. Acta Neuropathol Commun 2019; 7:96. [PMID: 31189468 PMCID: PMC6563368 DOI: 10.1186/s40478-019-0737-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 05/13/2019] [Indexed: 11/19/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disease resulting from motor neuron degeneration that causes muscle weakness, paralysis, and eventually respiratory failure. We investigated whether recombinant adeno-associated virus encoding human hepatocyte growth factor (rAAV-HGF) could generate beneficial effects in two mouse models with neuromuscular problems when intrathecally delivered to the subarachnoid space. We chose AAV serotype 1 (rAAV1) based on the expression levels and distribution of HGF protein in the lumbar spinal cord (LSC). After a single intrathecal (IT) injection of rAAV1-HGF, the protein level of HGF in the LSC peaked on day 14 and thereafter gradually decreased over the next 14 weeks. rAAV1-HGF was initially tested in the mouse nerve crush model. IT injection of rAAV1-HGF improved mouse hindlimb strength and rotarod performance, while histological analyses showed that the length of regenerated axons was increased and the structure of the neuromuscular junction (NMJ) was restored. rAAV1-HGF was also evaluated in the SOD1-G93A transgenic (TG) mouse model. Again, rAAV1-HGF not only improved motor performance but also increased the survival rate. Moreover, the number and diameter of spinal motor neurons (SMNs) were increased, and the shape of the NMJs restored. Data from in vitro motor cortical culture experiments indicated that treatment with recombinant HGF protein (rHGF) increased the axon length of corticospinal motor neurons (CSMNs). When cultures were treated with an ERK inhibitor, the effects of HGF on axon elongation, protein aggregation, and oxidative stress were suppressed, indicating that ERK phosphorylation played an important role(s). Taken together, our results suggested that HGF might play an important role(s) in delaying disease progression in the SOD1-G93A TG mouse model by reducing oxidative stress through the control of ERK phosphorylation.
Collapse
|
48
|
Just-Borràs L, Hurtado E, Cilleros-Mañé V, Biondi O, Charbonnier F, Tomàs M, Garcia N, Lanuza MA, Tomàs J. Overview of Impaired BDNF Signaling, Their Coupled Downstream Serine-Threonine Kinases and SNARE/SM Complex in the Neuromuscular Junction of the Amyotrophic Lateral Sclerosis Model SOD1-G93A Mice. Mol Neurobiol 2019; 56:6856-6872. [PMID: 30929165 DOI: 10.1007/s12035-019-1550-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/13/2019] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a chronic neurodegenerative disease characterized by progressive motor weakness. It is accepted that it is caused by motoneuron degeneration leading to a decrease in muscle stimulation. However, ALS is being redefined as a distal axonopathy, in that neuromuscular junction dysfunction precedes and may even influence motoneuron loss. In this synapse, several metabotropic receptor-mediated signaling pathways converge on effector kinases that phosphorylate targets that are crucial for synaptic stability and neurotransmission quality. We have previously shown that, in physiological conditions, nerve-induced muscle contraction regulates the brain-derived neurotrophic factor/tropomyosin-related kinase B (BDNF/TrkB) signaling to retrogradely modulate presynaptic protein kinases PKC and PKA, which are directly involved in the modulation of acetylcholine release. In ALS patients, the alteration of this signaling may significantly contribute to a motor impairment. Here, we investigate whether BDNF/TrkB signaling, the downstream PKC (cPKCβI, cPKCα, and nPKCε isoforms), and PKA (regulatory and catalytic subunits) and some SNARE/SM exocytotic machinery proteins (Munc18-1 and SNAP-25) are altered in the skeletal muscle of pre- and symptomatic SOD1-G93A mice. We found that this pathway is strongly affected in symptomatic ALS mice muscles including an unbalance between (I) BDNF and TrkB isoforms, (II) PKC isoforms and PKA subunits, and (III) Munc18-1 and SNAP-25 phosphorylation ratios. Changes in TrkB.T1 and cPKCβI are precociously observed in presymptomatic mice. Altogether, several of these molecular alterations can be partly associated with the known fast-to-slow motor unit transition during the disease process but others can be related with the initial disease pathogenesis.
Collapse
Affiliation(s)
- Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Víctor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Olivier Biondi
- INSERM UMRS 1124 and Université Paris Descartes, 45 rue des Saints-Pères, 75270, Paris Cedex 06, France
| | - Frédéric Charbonnier
- INSERM UMRS 1124 and Université Paris Descartes, 45 rue des Saints-Pères, 75270, Paris Cedex 06, France
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain.
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer St Llorenç num 21, 43201, Reus, Spain.
| |
Collapse
|
49
|
FM19G11-Loaded Gold Nanoparticles Enhance the Proliferation and Self-Renewal of Ependymal Stem Progenitor Cells Derived from ALS Mice. Cells 2019; 8:cells8030279. [PMID: 30909571 PMCID: PMC6468696 DOI: 10.3390/cells8030279] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/18/2019] [Accepted: 03/20/2019] [Indexed: 11/17/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease affecting motor neurons. In ALS mice, neurodegeneration is associated with the proliferative restorative attempts of ependymal stem progenitor cells (epSPCs) that normally lie in a quiescent in the spinal cord. Thus, modulation of the proliferation of epSPCs may represent a potential strategy to counteract neurodegeneration. Recent studies demonstrated that FM19G11, a hypoxia-inducible factor modulator, induces epSPC self-renewal and proliferation. The aim of the study was to investigate whether FM19G11-loaded gold nanoparticles (NPs) can affect self-renewal and proliferation processes in epSPCs isolated from G93A-SOD1 mice at disease onset. We discovered elevated levels of SOX2, OCT4, AKT1, and AKT3, key genes associated with pluripotency, self-renewal, and proliferation, in G93A-SOD1 epSPCs at the transcriptional and protein levels after treatment with FM19G11-loaded NPs. We also observed an increase in the levels of the mitochondrial uncoupling protein (UCP) gene in treated cells. FM19G11-loaded NPs treatment also affected the expression of the cell cycle-related microRNA (miR)-19a, along with its target gene PTEN, in G93A-SOD1 epSPCs. Overall our findings establish the significant impact of FM19G11-loaded NPs on the cellular pathways involved in self-renewal and proliferation in G93A-SOD1 epSPCs, thus providing an impetus to the design of novel tailored approaches to delay ALS disease progression.
Collapse
|
50
|
Kounakis K, Tavernarakis N. The Cytoskeleton as a Modulator of Aging and Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1178:227-245. [PMID: 31493230 DOI: 10.1007/978-3-030-25650-0_12] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cytoskeleton consists of filamentous protein polymers that form organized structures, contributing to a multitude of cell life aspects. It includes three types of polymers: the actin microfilaments, the microtubules and the intermediate filaments. Decades of research have implicated the cytoskeleton in processes that regulate cellular and organismal aging, as well as neurodegeneration associated with injury or neurodegenerative disease, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic Lateral Sclerosis, or Charcot Marie Tooth disease. Here, we provide a brief overview of cytoskeletal structure and function, and discuss experimental evidence linking cytoskeletal function and dynamics with aging and neurodegeneration.
Collapse
Affiliation(s)
- Konstantinos Kounakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece.,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece. .,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece.
| |
Collapse
|