51
|
Horio T, Ishikura Y, Ohashi R, Shiina N. Regulation of RNG105/caprin1 dynamics by pathogenic cytoplasmic FUS and TDP-43 in neuronal RNA granules modulates synaptic loss. Heliyon 2023; 9:e17065. [PMID: 37484309 PMCID: PMC10361247 DOI: 10.1016/j.heliyon.2023.e17065] [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: 05/14/2023] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 07/25/2023] Open
Abstract
In neurodegenerative diseases, the condensation of FUS and TDP-43 with RNA granules in neurons is linked to pathology, including synaptic disorders. However, the effects of FUS and TDP-43 on RNA granule factors remain unclear. Here, using primary cultured neurons from the mouse cerebral cortex, we show that excess cytoplasmic FUS and TDP-43 accumulated in dendritic RNA granules, where they increased the dynamics of a scaffold protein RNG105/caprin1 and dissociated it from the granules. This coincided with reduced levels of mRNA and translation around the granules and synaptic loss in dendrites. These defects were suppressed by non-dissociable RNG105, suggesting that RNG105 dissociation mediated the defects. In contrast to the model where FUS and TDP-43 co-aggregate with RNA granule factors to repress their activity, our findings provide a novel pathogenic mechanism whereby FUS and TDP-43 dissociate RNA scaffold proteins from RNA granules which are required for local translation that regulates synapse formation.
Collapse
Affiliation(s)
- Tomoyo Horio
- Laboratory of Neuronal Cell Biology, National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan
- Department of Basic Biology, Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8585, Japan
| | - Yui Ishikura
- Laboratory of Neuronal Cell Biology, National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan
- Department of Basic Biology, Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8585, Japan
| | - Rie Ohashi
- Laboratory of Neuronal Cell Biology, National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan
- Department of Basic Biology, Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8585, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
- Life Science Research Center, University of Toyama, Toyama, Toyama 930-0194, Japan
| | - Nobuyuki Shiina
- Laboratory of Neuronal Cell Biology, National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan
- Department of Basic Biology, Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi 444-8585, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| |
Collapse
|
52
|
Cannon AE, Zürrer WE, Zejlon C, Kulcsar Z, Lewandowski S, Piehl F, Granberg T, Ineichen BV. Neuroimaging findings in preclinical amyotrophic lateral sclerosis models-How well do they mimic the clinical phenotype? A systematic review. Front Vet Sci 2023; 10:1135282. [PMID: 37205225 PMCID: PMC10185801 DOI: 10.3389/fvets.2023.1135282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
Background and objectives Animal models for motor neuron diseases (MND) such as amyotrophic lateral sclerosis (ALS) are commonly used in preclinical research. However, it is insufficiently understood how much findings from these model systems can be translated to humans. Thus, we aimed at systematically assessing the translational value of MND animal models to probe their external validity with regards to magnetic resonance imaging (MRI) features. Methods In a comprehensive literature search in PubMed and Embase, we retrieved 201 unique publications of which 34 were deemed eligible for qualitative synthesis including risk of bias assessment. Results ALS animal models can indeed present with human ALS neuroimaging features: Similar to the human paradigm, (regional) brain and spinal cord atrophy as well as signal changes in motor systems are commonly observed in ALS animal models. Blood-brain barrier breakdown seems to be more specific to ALS models, at least in the imaging domain. It is noteworthy that the G93A-SOD1 model, mimicking a rare clinical genotype, was the most frequently used ALS proxy. Conclusions Our systematic review provides high-grade evidence that preclinical ALS models indeed show imaging features highly reminiscent of human ALS assigning them a high external validity in this domain. This opposes the high attrition of drugs during bench-to-bedside translation and thus raises concerns that phenotypic reproducibility does not necessarily render an animal model appropriate for drug development. These findings emphasize a careful application of these model systems for ALS therapy development thereby benefiting refinement of animal experiments. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier: CRD42022373146.
Collapse
Affiliation(s)
| | | | - Charlotte Zejlon
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Zsolt Kulcsar
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Center of Neurology, Academic Specialist Center, Stockholm Health Services, Stockholm, Sweden
| | - Tobias Granberg
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Benjamin Victor Ineichen
- Center for Reproducible Science, University of Zurich, Zurich, Switzerland
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
53
|
Oliveira NAS, Pinho BR, Oliveira JMA. Swimming against ALS: How to model disease in zebrafish for pathophysiological and behavioral studies. Neurosci Biobehav Rev 2023; 148:105138. [PMID: 36933816 DOI: 10.1016/j.neubiorev.2023.105138] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/02/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease that leads to progressive disability and motor impairment. Existing therapies provide modest improvements in patient survival, raising a need for new treatments for ALS. Zebrafish is a promising model animal for translational and fundamental research in ALS - it is an experimentally tractable vertebrate, with high homology to humans and an ample experimental toolbox. These advantages allow high-throughput study of behavioral and pathophysiological phenotypes. The last decade saw an increased interest in modelling ALS in zebrafish, leading to the current abundance and variety of available methods and models. Additionally, the rise of gene editing techniques and toxin combination studies has created novel opportunities for ALS studies in zebrafish. In this review, we address the relevance of zebrafish as a model animal for ALS studies, the strategies for model induction and key phenotypical evaluation. Furthermore, we discuss established and emerging zebrafish models of ALS, analyzing their validity, including their potential for drug testing, and highlighting research opportunities in this area.
Collapse
Affiliation(s)
- Nuno A S Oliveira
- UCIBIO-REQUIMTE, Applied Molecular Biosciences Unit, Mitochondria and Neurobiology Lab, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, Department of Drug Sciences, Pharmacology Lab, University of Porto, 4050-313 Porto, Portugal
| | - Brígida R Pinho
- UCIBIO-REQUIMTE, Applied Molecular Biosciences Unit, Mitochondria and Neurobiology Lab, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, Department of Drug Sciences, Pharmacology Lab, University of Porto, 4050-313 Porto, Portugal
| | - Jorge M A Oliveira
- UCIBIO-REQUIMTE, Applied Molecular Biosciences Unit, Mitochondria and Neurobiology Lab, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, Department of Drug Sciences, Pharmacology Lab, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
54
|
Gulino R. Synaptic Dysfunction and Plasticity in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:ijms24054613. [PMID: 36902042 PMCID: PMC10003601 DOI: 10.3390/ijms24054613] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Recent evidence has supported the hypothesis that amyotrophic lateral sclerosis (ALS) is a multi-step disease, as the onset of symptoms occurs after sequential exposure to a defined number of risk factors. Despite the lack of precise identification of these disease determinants, it is known that genetic mutations may contribute to one or more of the steps leading to ALS onset, the remaining being linked to environmental factors and lifestyle. It also appears evident that compensatory plastic changes taking place at all levels of the nervous system during ALS etiopathogenesis may likely counteract the functional effects of neurodegeneration and affect the timing of disease onset and progression. Functional and structural events of synaptic plasticity probably represent the main mechanisms underlying this adaptive capability, causing a significant, although partial and transient, resiliency of the nervous system affected by a neurodegenerative disease. On the other hand, the failure of synaptic functions and plasticity may be part of the pathological process. The aim of this review was to summarize what it is known today about the controversial involvement of synapses in ALS etiopathogenesis, and an analysis of the literature, although not exhaustive, confirmed that synaptic dysfunction is an early pathogenetic process in ALS. Moreover, it appears that adequate modulation of structural and functional synaptic plasticity may likely support function sparing and delay disease progression.
Collapse
Affiliation(s)
- Rosario Gulino
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95123 Catania, Italy
| |
Collapse
|
55
|
Wada H, Hikiami R, Kusui M, Minamiyama S, Asada-Utsugi M, Shodai A, Muramatsu SI, Morimura T, Urushitani M. In vivo analysis of aggregation propensity of low levels of mislocalized TDP-43 on cytopathological and behavioral phenotype of ALS/FTLD. Neurosci Res 2023:S0168-0102(23)00040-8. [PMID: 36804599 DOI: 10.1016/j.neures.2023.02.006] [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: 11/30/2022] [Revised: 01/26/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Mislocalization and aggregate formation of TAR DNA-biding protein of 43kD (TDP-43) in the cytoplasm are signatures of amyotrophic lateral sclerosis(ALS) and frontotemporal lobar degeneration (FTLD). However, the role of two cytopathologies in ALS/FTLD pathogenesis is unclear. This study aims to elucidate the difference in their causality of TDP-43 in ALS/FTLD in vivo, using transgenic mice expressing human TDP-43 with defective nuclear localizing signals in neurons (Cyto-TDP) and those with aggregation propensity (Cyto-aggTDP). The expression levels of both proteins are less than half of endogenous TDP-43. Despite the low amount of Cyto-aggTDP, the TDP-43 phosphorylation is more evident than Cyto-TDP. Histopathological study showed accelerated astrogliosis in the anterior cerebral cortex of both mice. Cyto-aggTDP mice demonstrated significant but faint loss of neurons in the perirhinal(PERI) and ectorhinal(ECT) areas and higher Iba1-staining in the spinal cord than aged control. Despite the lack of locomotor dysfunctions in both mice, the open-field test showed enhanced exploratory behavior, indicating that the perpetual mislocalization of TDP-43 may suffice to trigger FTLD behavior. Besides, the aggregation propensity of TDP-43 promotes phosphorylation, but its role in the clinicopathological phenotype may not be primary.
Collapse
Affiliation(s)
- Hideki Wada
- Department of Neurology, Shiga University of Medical Science, Japan; Molecular Neuroscience Research Center, Shiga University of Medical Science, Japan
| | - Ryota Hikiami
- Department of Therapeutics for Protein Misfolding Diseases, Shiga University of Medical Science, Japan.
| | - Makiko Kusui
- Department of Neurology, Shiga University of Medical Science, Japan
| | - Sumio Minamiyama
- Department of Neurology, Shiga University of Medical Science, Japan; Department of Neurology, Kyoto City Hospital, Japan
| | - Megumi Asada-Utsugi
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Japan
| | - Akemi Shodai
- Department of Neurology, Shiga University of Medical Science, Japan
| | - Shin-Ichi Muramatsu
- Division of Neurological Gene Therapy, Center for Open Innovation, Jichi Medical University, Japan
| | - Toshifumi Morimura
- Research Center for Animal Life Science, Shiga University of Medical Science, Japan
| | - Makoto Urushitani
- Department of Neurology, Shiga University of Medical Science, Japan; Molecular Neuroscience Research Center, Shiga University of Medical Science, Japan; Department of Therapeutics for Protein Misfolding Diseases, Shiga University of Medical Science, Japan.
| |
Collapse
|
56
|
Role of Triggers on the Structural and Functional Facets of TAR DNA-binding Protein 43. Neuroscience 2023; 511:110-130. [PMID: 36442745 DOI: 10.1016/j.neuroscience.2022.11.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/15/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
Nuclear TAR DNA-binding protein 43 (TDP-43) mitigates cellular function, but the dynamic nucleus-cytoplasm shuttling of TDP-43 is disrupted in diseases, such as Amyotrophic Lateral Sclerosis (ALS). The polymorphic nature of the TDP-43 structures in vitro and in vivo is a result of environmental factors leading to the protein pathogenesis. Once the triggers which mitigate TDP-43 biochemistry are identified, new therapies can be developed. This review aims to illustrate recent discoveries in the diversity of TDP-43 structures (amyloidogenic and non-amyloidogenic) and highlight the triggers which result in their formation.
Collapse
|
57
|
Corcia P, Lejeune P, Vourc'h P, Beltran S, Piegay AS, Blasco H, Meininger V. Comparison between PFN1 and SOD1 mutations in amyotrophic lateral sclerosis. Eur J Neurol 2023; 30:552-554. [PMID: 36176198 DOI: 10.1111/ene.15583] [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: 03/25/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND The objective of this study was to characterize the prototypical phenotype of patients with amyotrophic lateral sclerosis (ALS) associated with PFN1 mutations in profilin 1 (PFN1) and to determine clinical indications to test for mutations in this gene. MATERIAL AND METHODS The phenotype of three relatives carrying the M114V PFN1 mutation are detailed here and are compared with those of patients with ALS linked to PFN1 previously reported in the literature. RESULTS In this pedigree and in the literature, the main clinical findings which best describe familial ALS linked to PFN1 might be the following characteristics: pedigrees over five cases, age of onset around 50 years, site of onset systematically lower limbs and the absence of cognitive impairment. CONCLUSION First, the infrequent incidence of patients with ALS linked to PFN1 mutation supports the pursuit of a precise characterization of the phenotype linked to PFN1 mutations. Then, the numerous similarities between the phenotype amongst patients linked to SOD1 and PFN1 mutations and between histological features amongst both mice models prompts a review of the current ALS classifications, taking into consideration both phenotype and genotype.
Collapse
Affiliation(s)
- Philippe Corcia
- CRMR SLA, CHU Bretonneau, Tours, France
- UMR 1253 iBrain, Inserm, Université de Tours, Tours, France
| | | | - Patrick Vourc'h
- UMR 1253 iBrain, Inserm, Université de Tours, Tours, France
- Service de Biochimie et Biologie moléculaire, Tours, France
| | | | | | - Helene Blasco
- UMR 1253 iBrain, Inserm, Université de Tours, Tours, France
- Service de Biochimie et Biologie moléculaire, Tours, France
| | | |
Collapse
|
58
|
Matlock AD, Vaibhav V, Holewinski R, Venkatraman V, Dardov V, Manalo DM, Shelley B, Ornelas L, Banuelos M, Mandefro B, Escalante-Chong R, Li J, Finkbeiner S, Fraenkel E, Rothstein J, Thompson L, Sareen D, Svendsen CN, Van Eyk JE. NeuroLINCS Proteomics: Defining human-derived iPSC proteomes and protein signatures of pluripotency. Sci Data 2023; 10:24. [PMID: 36631473 PMCID: PMC9834231 DOI: 10.1038/s41597-022-01687-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 09/07/2022] [Indexed: 01/13/2023] Open
Abstract
The National Institute of Health (NIH) Library of integrated network-based cellular signatures (LINCS) program is premised on the generation of a publicly available data resource of cell-based biochemical responses or "signatures" to genetic or environmental perturbations. NeuroLINCS uses human inducible pluripotent stem cells (hiPSCs), derived from patients and healthy controls, and differentiated into motor neuron cell cultures. This multi-laboratory effort strives to establish i) robust multi-omic workflows for hiPSC and differentiated neuronal cultures, ii) public annotated data sets and iii) relevant and targetable biological pathways of spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS). Here, we focus on the proteomics and the quality of the developed workflow of hiPSC lines from 6 individuals, though epigenomics and transcriptomics data are also publicly available. Known and commonly used markers representing 73 proteins were reproducibly quantified with consistent expression levels across all hiPSC lines. Data quality assessments, data levels and metadata of all 6 genetically diverse human iPSCs analysed by DIA-MS are parsable and available as a high-quality resource to the public.
Collapse
Affiliation(s)
- Andrea D Matlock
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Vineet Vaibhav
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ronald Holewinski
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Vidya Venkatraman
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Victoria Dardov
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Danica-Mae Manalo
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Brandon Shelley
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Loren Ornelas
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Maria Banuelos
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Berhan Mandefro
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | | | - Jonathan Li
- NeuroLINCS, Department of Biological Engineering, MIT, Cambridge, MA, 02142, USA
| | - Steve Finkbeiner
- NeuroLINCS, Gladstone Institute of Neurological Disease and the Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Ernest Fraenkel
- NeuroLINCS, Department of Biological Engineering, MIT, Cambridge, MA, 02142, USA
| | - Jeffrey Rothstein
- NeuroLINCS, Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Leslie Thompson
- NeuroLINCS, Departments of Psychiatry and Human Behaviour, Neurobiology and Behaviour and UCI MIND, University of California Irvine, Irvine, CA, 92697, USA
| | - Dhruv Sareen
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Clive N Svendsen
- NeuroLINCS, Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Jennifer E Van Eyk
- NeuroLINCS, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
59
|
Nainu F, Mamada SS, Harapan H, Emran TB. Inflammation-Mediated Responses in the Development of Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:39-70. [PMID: 36949305 DOI: 10.1007/978-981-19-7376-5_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Since its first description over a century ago, neurodegenerative diseases (NDDs) have impaired the lives of millions of people worldwide. As one of the major threats to human health, NDDs are characterized by progressive loss of neuronal structure and function, leading to the impaired function of the CNS. While the precise mechanisms underlying the emergence of NDDs remains elusive, association of neuroinflammation with the emergence of NDDs has been suggested. The immune system is tightly controlled to maintain homeostatic milieu and failure in doing so has been shown catastrophic. Here, we review current concepts on the cellular and molecular drivers responsible in the induction of neuroinflammation and how such event further promotes neuronal damage leading to neurodegeneration. Experimental data generated from cell culture and animal studies, gross and molecular pathologies of human CNS samples, and genome-wide association study are discussed to provide deeper insights into the mechanistic details of neuroinflammation and its roles in the emergence of NDDs.
Collapse
Affiliation(s)
- Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Sukamto S Mamada
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Harapan Harapan
- School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| |
Collapse
|
60
|
Tsitsipatis D, Mazan-Mamczarz K, Si Y, Herman AB, Yang JH, Guha A, Piao Y, Fan J, Martindale JL, Munk R, Yang X, De S, Singh BK, Ho R, Gorospe M, King PH. Transcriptomic analysis of human ALS skeletal muscle reveals a disease-specific pattern of dysregulated circRNAs. Aging (Albany NY) 2022; 14:9832-9859. [PMID: 36585921 PMCID: PMC9831722 DOI: 10.18632/aging.204450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/13/2022] [Indexed: 01/01/2023]
Abstract
Circular RNAs are abundant, covalently closed transcripts that arise in cells through back-splicing and display distinct expression patterns across cells and developmental stages. While their functions are largely unknown, their intrinsic stability has made them valuable biomarkers in many diseases. Here, we set out to examine circRNA patterns in amyotrophic lateral sclerosis (ALS). By RNA-sequencing analysis, we first identified circRNAs and linear RNAs that were differentially abundant in skeletal muscle biopsies from ALS compared to normal individuals. By RT-qPCR analysis, we confirmed that 8 circRNAs were significantly elevated and 10 were significantly reduced in ALS, while the linear mRNA counterparts, arising from shared precursor RNAs, generally did not change. Several of these circRNAs were also differentially abundant in motor neurons derived from human induced pluripotent stem cells (iPSCs) bearing ALS mutations, and across different disease stages in skeletal muscle from a mouse model of ALS (SOD1G93A). Interestingly, a subset of the circRNAs significantly elevated in ALS muscle biopsies were significantly reduced in the spinal cord samples from ALS patients and ALS (SOD1G93A) mice. In sum, we have identified differentially abundant circRNAs in ALS-relevant tissues (muscle and spinal cord) that could inform about neuromuscular molecular programs in ALS and guide the development of therapies.
Collapse
Affiliation(s)
- Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Krystyna Mazan-Mamczarz
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ying Si
- Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA
| | - Allison B. Herman
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jen-Hao Yang
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Abhishek Guha
- Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yulan Piao
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jinshui Fan
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jennifer L. Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Xiaoling Yang
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Supriyo De
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Brijesh K. Singh
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ritchie Ho
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Peter H. King
- Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
61
|
Pellegrini F, Padovano V, Biscarini S, Santini T, Setti A, Galfrè SG, Silenzi V, Vitiello E, Mariani D, Nicoletti C, Torromino G, De Leonibus E, Martone J, Bozzoni I. A KO mouse model for the lncRNA Lhx1os produces motor neuron alterations and locomotor impairment. iScience 2022; 26:105891. [PMID: 36647387 PMCID: PMC9840152 DOI: 10.1016/j.isci.2022.105891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/22/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Here, we describe a conserved motor neuron-specific long non-coding RNA, Lhx1os, whose knockout in mice produces motor impairment and postnatal reduction of mature motor neurons (MNs). The ER stress-response pathway result specifically altered with the downregulation of factors involved in the unfolded protein response (UPR). Lhx1os was found to bind the ER-associated PDIA3 disulfide isomerase and to affect the expression of the same set of genes controlled by this protein, indicating that the two factors act in conjunction to modulate the UPR. Altogether, the observed phenotype and function of Lhx1os indicate its important role in the control of MN homeostasis and function.
Collapse
Affiliation(s)
- Flaminia Pellegrini
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy,Center for Life Nano- & Neuro-Science@Sapienza of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Vittorio Padovano
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy,Center for Life Nano- & Neuro-Science@Sapienza of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Silvia Biscarini
- Center for Life Nano- & Neuro-Science@Sapienza of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Tiziana Santini
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy,Center for Life Nano- & Neuro-Science@Sapienza of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Adriano Setti
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Silvia Giulia Galfrè
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Valentina Silenzi
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy,Center for Life Nano- & Neuro-Science@Sapienza of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Erika Vitiello
- Center for Human Technologies (CHT) Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Davide Mariani
- Center for Human Technologies (CHT) Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Carmine Nicoletti
- DAHFMO - Section of Histology and Medical Embryology, Sapienza University of Rome, 00185 Rome, Italy
| | - Giulia Torromino
- Institute of Cellular Biology and Neurobiology "ABT", CNR, Monterotondo, 00015 Rome, Italy
| | - Elvira De Leonibus
- Institute of Cellular Biology and Neurobiology "ABT", CNR, Monterotondo, 00015 Rome, Italy,Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, 80078 Naples, Italy
| | - Julie Martone
- Institute of Molecular Biology and Pathology, CNR, 00185 Rome, Italy,Corresponding author
| | - Irene Bozzoni
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy,Center for Life Nano- & Neuro-Science@Sapienza of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy,Center for Human Technologies (CHT) Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy,Corresponding author
| |
Collapse
|
62
|
Ciuro M, Sangiorgio M, Leanza G, Gulino R. A Meta-Analysis Study of SOD1-Mutant Mouse Models of ALS to Analyse the Determinants of Disease Onset and Progression. Int J Mol Sci 2022; 24:ijms24010216. [PMID: 36613659 PMCID: PMC9820332 DOI: 10.3390/ijms24010216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 12/25/2022] Open
Abstract
A complex interaction between genetic and external factors determines the development of amyotrophic lateral sclerosis (ALS). Epidemiological studies on large patient cohorts have suggested that ALS is a multi-step disease, as symptom onset occurs only after exposure to a sequence of risk factors. Although the exact nature of these determinants remains to be clarified, it seems clear that: (i) genetic mutations may be responsible for one or more of these steps; (ii) other risk factors are probably linked to environment and/or to lifestyle, and (iii) compensatory plastic changes taking place during the ALS etiopathogenesis probably affect the timing of onset and progression of disease. Current knowledge on ALS mechanisms and therapeutic targets, derives mainly from studies involving superoxide dismutase 1 (SOD1) transgenic mice; therefore, it would be fundamental to verify whether a multi-step disease concept can also be applied to these animal models. With this aim, a meta-analysis study has been performed using a collection of primary studies (n = 137), selected according to the following criteria: (1) the studies should employ SOD1 transgenic mice; (2) the studies should entail the presence of a disease-modifying experimental manipulation; (3) the studies should make use of Kaplan-Meier plots showing the distribution of symptom onset and lifespan. Then, using a subset of this study collection (n = 94), the effects of treatments on key molecular mechanisms, as well as on the onset and progression of disease have been analysed in a large population of mice. The results are consistent with a multi-step etiopathogenesis of disease in ALS mice (including two to six steps, depending on the particular SOD1 mutation), closely resembling that observed in patient cohorts, and revealed an interesting relationship between molecular mechanisms and disease manifestation. Thus, SOD1 mouse models may be considered of high predictive value to understand the determinants of disease onset and progression, as well as to identify targets for therapeutic interventions.
Collapse
Affiliation(s)
- Maria Ciuro
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95123 Catania, Italy
| | - Maria Sangiorgio
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95123 Catania, Italy
| | - Giampiero Leanza
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
- Molecular Preclinical and Translational Imaging Research Centre—IMPRonTE, University of Catania, 95125 Catania, Italy
| | - Rosario Gulino
- Department of Biomedical and Biotechnological Sciences, Physiology Section, University of Catania, 95123 Catania, Italy
- Molecular Preclinical and Translational Imaging Research Centre—IMPRonTE, University of Catania, 95125 Catania, Italy
- Correspondence:
| |
Collapse
|
63
|
Provenzano F, Nyberg S, Giunti D, Torazza C, Parodi B, Bonifacino T, Usai C, Kerlero de Rosbo N, Milanese M, Uccelli A, Shaw PJ, Ferraiuolo L, Bonanno G. Micro-RNAs Shuttled by Extracellular Vesicles Secreted from Mesenchymal Stem Cells Dampen Astrocyte Pathological Activation and Support Neuroprotection in In-Vitro Models of ALS. Cells 2022; 11:cells11233923. [PMID: 36497181 PMCID: PMC9741322 DOI: 10.3390/cells11233923] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease with no effective cure. Astrocytes display a toxic phenotype in ALS and contribute to motoneuron (MN) degeneration. Modulating astrocytes' neurotoxicity can reduce MN death. Our previous studies showed the beneficial effect of mesenchymal stem cell (MSC) administration in SOD1G93A ALS mice, but the mechanisms are still unclear. We postulated that the effects could be mediated by extracellular vesicles (EVs) secreted by MSCs. We investigated, by immunohistochemical, molecular, and in vitro functional analyses, the activity of MSC-derived EVs on the pathological phenotype and neurotoxicity of astrocytes isolated from the spinal cord of symptomatic SOD1G93A mice and human astrocytes (iAstrocytes) differentiated from inducible neural progenitor cells (iNPCs) of ALS patients. In vitro EV exposure rescued mouse and human ALS astrocytes' neurotoxicity towards MNs. EVs significantly dampened the pathological phenotype and neuroinflammation in SOD1G93A astrocytes. In iAstrocytes, exposure to EVs increased the antioxidant factor Nrf2 and reduced reactive oxygen species. We previously found nine miRNAs upregulated in MSC-derived EVs. Here, the transfection of SOD1G93A astrocytes with single miRNA mimics reduced astrocytes' activation and the expression of neuroinflammatory factors. Moreover, miR-466q and miR-467f mimics downregulate Mapk11, while miR-466m-5p and miR-466i-3p mimics promote the nuclear translocation of Nrf2. In iAstrocytes, transfection with miR-29b-3p mimic upregulated NQO1 antioxidant activity and reduced neurotoxicity towards MNs. MSC-derived EVs modulate astrocytes' reactive phenotype and neurotoxicity through anti-inflammatory and antioxidant-shuttled miRNAs, thus representing a therapeutic strategy in ALS.
Collapse
Affiliation(s)
- Francesca Provenzano
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
| | - Sophie Nyberg
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Debora Giunti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo, 316132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Carola Torazza
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
| | - Benedetta Parodi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo, 316132 Genoa, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council (CNR), Via De Marini 6, 16149 Genoa, Italy
| | - Nicole Kerlero de Rosbo
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
- TomaLab, Institute of Nanotechnology, National Research Council (CNR), Piazzale Aldo Moro 5, 0018 Rome, Italy
| | - Marco Milanese
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
- Correspondence: (M.M.); (L.F.); Tel.: +39-01-0335-2046 (M.M.); +44-(0)114-222-2257 (L.F.)
| | - Antonio Uccelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo, 316132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield S10 2HQ, UK
- Correspondence: (M.M.); (L.F.); Tel.: +39-01-0335-2046 (M.M.); +44-(0)114-222-2257 (L.F.)
| | - Giambattista Bonanno
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
64
|
Mazumder S, Kiernan MC, Halliday GM, Timmins HC, Mahoney CJ. The contribution of brain banks to knowledge discovery in amyotrophic lateral sclerosis: A systematic review. Neuropathol Appl Neurobiol 2022; 48:e12845. [PMID: 35921237 PMCID: PMC9804699 DOI: 10.1111/nan.12845] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/17/2022] [Accepted: 07/23/2022] [Indexed: 01/09/2023]
Abstract
Over the past decade, considerable efforts have been made to accelerate pathophysiological understanding of fatal neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) with brain banks at the forefront. In addition to exploratory disease mechanisms, brain banks have aided our understanding with regard to clinical diagnosis, genetics and cell biology. Across neurodegenerative disorders, the impact of brain tissue in ALS research has yet to be quantified. This review aims to outline (i) how postmortem tissues from brain banks have influenced our understanding of ALS over the last 15 years, (ii) correlate the location of dedicated brain banks with the geographical prevalence of ALS, (iii) identify the frequency of features reported from postmortem studies and (iv) propose common reporting standards for materials obtained from dedicated brain banks. A systematic review was conducted using PubMed and Web of Science databases using key words. From a total of 1439 articles, 73 articles were included in the final review, following PRISMA guidelines. Following a thematic analysis, articles were categorised into five themes; clinico-pathological (13), genetic (20), transactive response DNA binding protein 43 (TDP-43) pathology (12), non-TDP-43 neuronal pathology (nine) and extraneuronal pathology (19). Research primarily focused on the genetics of ALS, followed by protein pathology. About 63% of the brain banks were in the United States of America and United Kingdom. The location of brain banks overall aligned with the incidence of ALS worldwide with 88% of brain banks situated in Europe and North America. An overwhelming lack of consistency in reporting and replicability was observed, strengthening the need for a standardised reporting system. Overall, postmortem material from brain banks generated substantial new knowledge in areas of genetics and proteomics and supports their ongoing role as an important research tool.
Collapse
Affiliation(s)
- Srestha Mazumder
- ForeFront Clinic, Brain and Mind CentreUniversity of SydneySydneyNew South WalesAustralia
| | - Matthew C. Kiernan
- ForeFront Clinic, Brain and Mind CentreUniversity of SydneySydneyNew South WalesAustralia
| | - Glenda M. Halliday
- Frontier, Brain and Mind CentreUniversity of SydneySydneyNew South WalesAustralia
| | - Hannah C. Timmins
- ForeFront Clinic, Brain and Mind CentreUniversity of SydneySydneyNew South WalesAustralia
| | - Colin J. Mahoney
- ForeFront Clinic, Brain and Mind CentreUniversity of SydneySydneyNew South WalesAustralia
| |
Collapse
|
65
|
Butler R, Bradford D, Rodgers KE. Analysis of shared underlying mechanism in neurodegenerative disease. Front Aging Neurosci 2022; 14:1006089. [PMID: 36523957 PMCID: PMC9745190 DOI: 10.3389/fnagi.2022.1006089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/08/2022] [Indexed: 08/27/2023] Open
Abstract
In this review, the relationship between bioenergetics, mitochondrial dysfunction, and inflammation will be and how they contribute to neurodegeneration, specifically in Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS) will be reviewed. Long-term changes in mitochondrial function, autophagy dysfunction, and immune activation are commonalities shared across these age-related disorders. Genetic risk factors for these diseases support an autophagy-immune connection in the underlying pathophysiology. Critical areas of deeper evaluation in these bioenergetic processes may lead to potential therapeutics with efficacy across multiple neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | - Kathleen E. Rodgers
- Department of Medical Pharmacology, Center for Innovation in Brain Science, University of Arizona College of Medicine, Tucson, AZ, United States
| |
Collapse
|
66
|
4-Phenylbutyric Acid (4-PBA) Derivatives Prevent SOD1 Amyloid Aggregation In Vitro with No Effect on Disease Progression in SOD1-ALS Mice. Int J Mol Sci 2022; 23:ijms23169403. [PMID: 36012668 PMCID: PMC9409193 DOI: 10.3390/ijms23169403] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 12/21/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the degeneration of motor neurons. Mutations in the superoxide dismutase (SOD1) gene, causing protein misfolding and aggregation, were suggested as the pathogenic mechanisms involved in familial ALS cases. In the present study, we investigated the potential therapeutic effect of C4 and C5, two derivatives of the chemical chaperone 4-phenylbutyric acid (4-PBA). By combining in vivo and in vitro techniques, we show that, although C4 and C5 successfully inhibited amyloid aggregation of recombinant mutant SOD1 in a dose-dependent manner, they failed to suppress the accumulation of misfolded SOD1. Moreover, C4 or C5 daily injections to SOD1G93A mice following onset had no effect on either the accumulation of misfolded SOD1 or the neuroinflammatory response in the spinal cord and, consequently, failed to extend the survival of SOD1G93A mice or to improve their motor symptoms. Finally, pharmacokinetic (PK) studies demonstrated that high concentrations of C4 and C5 reached the brain and spinal cord but only for a short period of time. Thus, our findings suggest that use of such chemical chaperones for ALS drug development may need to be optimized for more effective results.
Collapse
|
67
|
Boros BD, Schoch KM, Kreple CJ, Miller TM. Antisense Oligonucleotides for the Study and Treatment of ALS. Neurotherapeutics 2022; 19:1145-1158. [PMID: 35653060 PMCID: PMC9587169 DOI: 10.1007/s13311-022-01247-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2022] [Indexed: 10/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by motor neuron loss. ALS is now associated with mutations in numerous genes, many of which cause disease in part through toxic gain-of-function mechanisms. Antisense oligonucleotides (ASOs) are small sequences of DNA that can reduce expression of a target gene at the post-transcriptional level, making them attractive for neutralizing mutant or toxic gene products. Advancements in the medicinal chemistries of ASOs have improved their pharmacodynamic profile to allow safe and effective delivery to the central nervous system. ASO therapies for ALS have rapidly developed over the last two decades, and ASOs that target SOD1, C9orf72, FUS, and ATXN2 are now in clinical trials for familial or sporadic forms of ALS. This review discusses the current state of ASO therapies for ALS, outlining their successes from preclinical development to early clinical trials.
Collapse
Affiliation(s)
- Benjamin D Boros
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, Box 8111, 115 Biotechnology Bldg, 660 S. Euclid Ave, MO, 63110, St. Louis, USA
| | - Kathleen M Schoch
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, Box 8111, 115 Biotechnology Bldg, 660 S. Euclid Ave, MO, 63110, St. Louis, USA
| | - Collin J Kreple
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, Box 8111, 115 Biotechnology Bldg, 660 S. Euclid Ave, MO, 63110, St. Louis, USA
| | - Timothy M Miller
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, Box 8111, 115 Biotechnology Bldg, 660 S. Euclid Ave, MO, 63110, St. Louis, USA.
| |
Collapse
|
68
|
Soares MBP, Gonçalves RGJ, Vasques JF, da Silva-Junior AJ, Gubert F, Santos GC, de Santana TA, Almeida Sampaio GL, Silva DN, Dominici M, Mendez-Otero R. Current Status of Mesenchymal Stem/Stromal Cells for Treatment of Neurological Diseases. Front Mol Neurosci 2022; 15:883378. [PMID: 35782379 PMCID: PMC9244712 DOI: 10.3389/fnmol.2022.883378] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Neurological disorders include a wide spectrum of clinical conditions affecting the central and peripheral nervous systems. For these conditions, which affect hundreds of millions of people worldwide, generally limited or no treatments are available, and cell-based therapies have been intensively investigated in preclinical and clinical studies. Among the available cell types, mesenchymal stem/stromal cells (MSCs) have been widely studied but as yet no cell-based treatment exists for neurological disease. We review current knowledge of the therapeutic potential of MSC-based therapies for neurological diseases, as well as possible mechanisms of action that may be explored to hasten the development of new and effective treatments. We also discuss the challenges for culture conditions, quality control, and the development of potency tests, aiming to generate more efficient cell therapy products for neurological disorders.
Collapse
Affiliation(s)
- Milena B. P. Soares
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Renata G. J. Gonçalves
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana F. Vasques
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Almir J. da Silva-Junior
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Nanotecnologia no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Gubert
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Girlaine Café Santos
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Thaís Alves de Santana
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | - Gabriela Louise Almeida Sampaio
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (IGM-FIOCRUZ/BA), Salvador, Brazil
- Instituto SENAI de Sistemas Avançados de Saúde (CIMATEC ISI-SAS), Centro Universitário SENAI/CIMATEC, Salvador, Brazil
| | | | - Massimo Dominici
- Laboratory of Cellular Therapy, Division of Oncology, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Rosalia Mendez-Otero
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Saúde no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa Redes de Pesquisa em Nanotecnologia no Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
69
|
Bauer US, Fiskum V, Nair RR, van de Wijdeven R, Kentros C, Sandvig I, Sandvig A. Validation of Functional Connectivity of Engineered Neuromuscular Junction With Recombinant Monosynaptic Pseudotyped ΔG-Rabies Virus Tracing. Front Integr Neurosci 2022; 16:855071. [PMID: 35669734 PMCID: PMC9163662 DOI: 10.3389/fnint.2022.855071] [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: 01/14/2022] [Accepted: 05/02/2022] [Indexed: 11/15/2022] Open
Abstract
Current preclinical models of neurodegenerative disease, such as amyotrophic lateral sclerosis (ALS), can significantly benefit from in vitro neuroengineering approaches that enable the selective study and manipulation of neurons, networks, and functional units of interest. Custom-designed compartmentalized microfluidic culture systems enable the co-culture of different relevant cell types in interconnected but fluidically isolated microenvironments. Such systems can thus be applied for ALS disease modeling, as they enable the recapitulation and study of neuromuscular junctions (NMJ) through co-culturing of motor neurons and muscle cells in separate, but interconnected compartments. These in vitro systems are particularly relevant for investigations of mechanistic aspects of the ALS pathological cascade in engineered NMJ, as progressive loss of NMJ functionality may constitute one of the hallmarks of disease related pathology at early onset, in line with the dying back hypothesis. In such models, ability to test whether motor neuron degeneration in ALS starts at the nerve terminal or at the NMJ and retrogradely progresses to the motor neuron cell body largely relies on robust methods for verification of engineered NMJ functionality. In this study, we demonstrate the functionality of engineered NMJs within a microfluidic chip with a differentially perturbable microenvironment using a designer pseudotyped ΔG-rabies virus for retrograde monosynaptic tracing.
Collapse
Affiliation(s)
- Ulrich Stefan Bauer
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Vegard Fiskum
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Rajeevkumar Raveendran Nair
- Centre for Neural Computation, Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Rosanne van de Wijdeven
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Clifford Kentros
- Centre for Neural Computation, Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Institute of Neuroscience, University of Oregon, Eugene, OR, United States
| | - Ioanna Sandvig
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Axel Sandvig
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Clinical Neuroscience, Umeå University Hospital, Umeå, Sweden
- Department of Community Medicine and Rehabilitation, Umeå University, Umeå, Sweden
- *Correspondence: Axel Sandvig,
| |
Collapse
|
70
|
Patients' Stem Cells Differentiation in a 3D Environment as a Promising Experimental Tool for the Study of Amyotrophic Lateral Sclerosis. Int J Mol Sci 2022; 23:ijms23105344. [PMID: 35628156 PMCID: PMC9141644 DOI: 10.3390/ijms23105344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 12/17/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease (NDD) that affects motor neurons, causing weakness, muscle atrophy and spasticity. Unfortunately, there are only symptomatic treatments available. Two important innovations in recent years are three-dimensional (3D) bioprinting and induced pluripotent stem cells (iPSCs). The aim of this work was to demonstrate the robustness of 3D cultures for the differentiation of stem cells for the study of ALS. We reprogrammed healthy and sALS peripheral blood mononuclear cells (PBMCs) in iPSCs and differentiated them in neural stem cells (NSCs) in 2D. NSCs were printed in 3D hydrogel-based constructs and subsequently differentiated first in motor neuron progenitors and finally in motor neurons. Every step of differentiation was tested for cell viability and characterized by confocal microscopy and RT-qPCR. Finally, we tested the electrophysiological characteristics of included NSC34. We found that NSCs maintained good viability during the 3D differentiation. Our results suggest that the hydrogel does not interfere with the correct differentiation process or with the electrophysiological features of the included cells. Such evidence confirmed that 3D bioprinting can be considered a good model for the study of ALS pathogenesis.
Collapse
|
71
|
Majkutewicz I. Dimethyl fumarate: A review of preclinical efficacy in models of neurodegenerative diseases. Eur J Pharmacol 2022; 926:175025. [DOI: 10.1016/j.ejphar.2022.175025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/27/2022] [Accepted: 05/09/2022] [Indexed: 11/03/2022]
|
72
|
Li S, Zhu Y, Wei C, Li C, Chen W, Jiang S, Yuan D, Xu R. Identification of Molecular Correlations Between DHRS4 and Progressive Neurodegeneration in Amyotrophic Lateral Sclerosis By Gene Co-Expression Network Analysis. Front Immunol 2022; 13:874978. [PMID: 35479082 PMCID: PMC9035787 DOI: 10.3389/fimmu.2022.874978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/15/2022] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease, and its candidate biomarkers have not yet been fully elucidated in previous studies. Therefore, with the present study, we aim to define and verify effective biomarkers of ALS by bioinformatics. Here, we employed differentially expressed gene (DEG) analysis, weighted gene co-expression network analysis (WGCNA), enrichment analysis, immune infiltration analysis, and protein-protein interaction (PPI) to identify biomarkers of ALS. To validate the biomarkers, we isolated the lumbar spinal cord from mice and characterized them using Western blotting and immunofluorescence. The results showed that Dhrs4 expression in the spinal cord was upregulated with the progression of SOD1G93A mice, and the upregulation of DHRS4 and its synergistic DHRS3 might be primarily associated with the activation of the complement cascade in the immune system (C1QA, C1QB, C1QC, C3, and ITGB2), which might be a novel mechanism that induces spinal neurodegeneration in ALS. We propose that DHRS4 and its synergistic DHRS3 are promising molecular markers for detecting ALS progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People’s Hospital, Affiliated People’s Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
73
|
Development of a Novel Technique for the Measurement of Neuromuscular Junction Functionality in Isotonic Conditions. Cell Mol Bioeng 2022; 15:255-265. [PMID: 35611165 PMCID: PMC9124252 DOI: 10.1007/s12195-022-00721-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/09/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction The neuromuscular junction (NMJ) is a chemical synapse responsible for converting electrical pulses generated by the motor neuron into electrical activity in muscle fibers, and is severely impaired in various diseases, such as Amyotrophic Lateral Sclerosis (ALS). Here, we proposed a novel technique to measure, for the first time, NMJ functionality in isotonic conditions, which better reflect muscle physiological activity. Methods We employed the in-situ testing technique, studied a proper placing of two pairs of wire electrodes for nerve and muscle stimulation, developed an extensive testing protocol, and proposed a novel parameter, the Isotonic Neurotransmission Failure (INF), to properly capture the impairments in neurotransmission during isotonic fatigue. We employed wild-type mice to assess the feasibility of the proposed technique, and the ALS model SOD1G93A mice to demonstrate the validity of the INF. Results Results confirmed the measurement accuracy in term of average value and coefficient of variation of the parameters measured through nerve stimulation in comparison with the corresponding values obtained for membrane stimulation. The INF values computed for the SOD1G93A tibialis anterior muscles pointed out an impairment of ALS mice during the isotonic fatigue test, whereas, as expected, their resistance to fatigue was higher. Conclusions In this work we devised a novel technique and a new parameter for a deep assessment of NMJ functionality in isotonic conditions, including fatigue, which is the most crucial condition for the neuronal signal transmission. This technique may be applied to other animal models, to unravel the mechanisms behind muscle-nerve impairments in other neurodegenerative pathologies.
Collapse
|
74
|
Zschüntzsch J, Meyer S, Shahriyari M, Kummer K, Schmidt M, Kummer S, Tiburcy M. The Evolution of Complex Muscle Cell In Vitro Models to Study Pathomechanisms and Drug Development of Neuromuscular Disease. Cells 2022; 11:1233. [PMID: 35406795 PMCID: PMC8997482 DOI: 10.3390/cells11071233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
Many neuromuscular disease entities possess a significant disease burden and therapeutic options remain limited. Innovative human preclinical models may help to uncover relevant disease mechanisms and enhance the translation of therapeutic findings to strengthen neuromuscular disease precision medicine. By concentrating on idiopathic inflammatory muscle disorders, we summarize the recent evolution of the novel in vitro models to study disease mechanisms and therapeutic strategies. A particular focus is laid on the integration and simulation of multicellular interactions of muscle tissue in disease phenotypes in vitro. Finally, the requirements of a neuromuscular disease drug development workflow are discussed with a particular emphasis on cell sources, co-culture systems (including organoids), functionality, and throughput.
Collapse
Affiliation(s)
- Jana Zschüntzsch
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Stefanie Meyer
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Mina Shahriyari
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| | - Karsten Kummer
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Matthias Schmidt
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| | - Susann Kummer
- Risk Group 4 Pathogens–Stability and Persistence, Biosafety Level-4 Laboratory, Center for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany;
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| |
Collapse
|
75
|
Martinez B, Peplow PV. MicroRNA expression in animal models of amyotrophic lateral sclerosis and potential therapeutic approaches. Neural Regen Res 2022; 17:728-740. [PMID: 34472458 PMCID: PMC8530133 DOI: 10.4103/1673-5374.322431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/03/2021] [Accepted: 03/27/2021] [Indexed: 12/11/2022] Open
Abstract
A review of recent animal models of amyotrophic lateral sclerosis showed a large number of miRNAs had altered levels of expression in the brain and spinal cord, motor neurons of spinal cord and brainstem, and hypoglossal, facial, and red motor nuclei and were mostly upregulated. Among the miRNAs found to be upregulated in two of the studies were miR-21, miR-155, miR-125b, miR-146a, miR-124, miR-9, and miR-19b, while those downregulated in two of the studies included miR-146a, miR-29, miR-9, and miR-125b. A change of direction in miRNA expression occurred in some tissues when compared (e.g., miR-29b-3p in cerebellum and spinal cord of wobbler mice at 40 days), or at different disease stages (e.g., miR-200a in spinal cord of SOD1(G93A) mice at 95 days vs. 108 and 112 days). In the animal models, suppression of miR-129-5p resulted in increased lifespan, improved muscle strength, reduced neuromuscular junction degeneration, and tended to improve motor neuron survival in the SOD1(G93A) mouse model. Suppression of miR-155 was also associated with increased lifespan, while lowering of miR-29a tended to improve lifespan in males and increase muscle strength in SOD1(G93A) mice. Overexpression of members of miR-17~92 cluster improved motor neuron survival in SOD1(G93A) mice. Treatment with an artificial miRNA designed to target hSOD1 increased lifespan and improved muscle strength in SOD1(G93A) animals. Further studies with animal models of amyotrophic lateral sclerosis are warranted to validate these findings and identify specific miRNAs whose suppression or directed against hSOD1 results in increased lifespan, improved muscle strength, reduced neuromuscular junction degeneration, and improved motor neuron survival in SOD1(G93A) animals.
Collapse
Affiliation(s)
- Bridget Martinez
- Physical Chemistry and Applied Spectroscopy, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
- Department of Medicine, St. Georges University School of Medicine, Grenada
| | - Philip V. Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
76
|
New pathogenic insights from large animal models of neurodegenerative diseases. Protein Cell 2022; 13:707-720. [PMID: 35334073 PMCID: PMC9233730 DOI: 10.1007/s13238-022-00912-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Animal models are essential for investigating the pathogenesis and developing the treatment of human diseases. Identification of genetic mutations responsible for neurodegenerative diseases has enabled the creation of a large number of small animal models that mimic genetic defects found in the affected individuals. Of the current animal models, rodents with genetic modifications are the most commonly used animal models and provided important insights into pathogenesis. However, most of genetically modified rodent models lack overt neurodegeneration, imposing challenges and obstacles in utilizing them to rigorously test the therapeutic effects on neurodegeneration. Recent studies that used CRISPR/Cas9-targeted large animal (pigs and monkeys) have uncovered important pathological events that resemble neurodegeneration in the patient’s brain but could not be produced in small animal models. Here we highlight the unique nature of large animals to model neurodegenerative diseases as well as the limitations and challenges in establishing large animal models of neurodegenerative diseases, with focus on Huntington disease, Amyotrophic lateral sclerosis, and Parkinson diseases. We also discuss how to use the important pathogenic insights from large animal models to make rodent models more capable of recapitulating important pathological features of neurodegenerative diseases.
Collapse
|
77
|
Ruffo P, Perrone B, Conforti FL. SOD-1 Variants in Amyotrophic Lateral Sclerosis: Systematic Re-Evaluation According to ACMG-AMP Guidelines. Genes (Basel) 2022; 13:genes13030537. [PMID: 35328090 PMCID: PMC8955492 DOI: 10.3390/genes13030537] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 02/01/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common type of motor neuron disease whose causes are unclear. The first ALS gene associated with the autosomal dominant form of the disease was SOD1. This gene has a high rate of rare variants, and an appropriate classification is essential for a correct ALS diagnosis. In this study, we re-evaluated the classification of all previously reported SOD1 variants (n = 202) from ALSoD, project MinE, and in-house databases by applying the ACMG-AMP criteria to ALS. New bioinformatics analysis, frequency rating, and a thorough search for functional studies were performed. We also proposed adjusting criteria strength describing how to apply them to SOD1 variants. Most of the previously reported variants have been reclassified as likely pathogenic and pathogenic based on the modified weight of the PS3 criterion, highlighting how in vivo or in vitro functional studies are determining their interpretation and classification. Furthermore, this study reveals the concordance and discordance of annotations between open databases, indicating the need for expert review to adapt the study of variants to a specific disease. Indeed, in complex diseases, such as ALS, the oligogenic inheritance, the presence of genes that act as risk factors and the reduced penetration must be considered. Overall, the diagnosis of ALS remains clinical, and improving variant classification could support genetic data as diagnostic criteria.
Collapse
|
78
|
Martin S, Battistini C, Sun J. A Gut Feeling in Amyotrophic Lateral Sclerosis: Microbiome of Mice and Men. Front Cell Infect Microbiol 2022; 12:839526. [PMID: 35360111 PMCID: PMC8963415 DOI: 10.3389/fcimb.2022.839526] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a severely debilitating disease characterized by progressive degeneration of motor neurons. ALS etiology and pathophysiology are not well understood. It could be the consequences of complex interactions among host factors, microbiome, and the environmental factors. Recent data suggest the novel roles of intestinal dysfunction and microbiota in ALS etiology and progression. Although microbiome may indeed play a critical role in ALS pathogenesis, studies implicating innate immunity and intestinal changes in early disease pathology are limited. The gastrointestinal symptoms in the ALS patients before their diagnosis are largely ignored in the current medical practice. This review aims to explore existing evidence of gastrointestinal symptoms and progress of microbiome in ALS pathogenesis from human and animal studies. We discuss dietary, metabolites, and possible therapeutic approaches by targeting intestinal function and microbiome. Finally, we evaluate existing evidence and identify gaps in the knowledge for future directions in ALS. It is essential to understanding the microbiome and intestinal pathogenesis that determine when, where, and whether microbiome and metabolites critical to ALS progression. These studies will help us to develop more accurate diagnosis and better treatment not only for this challenging disease, but also for other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarah Martin
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Carolina Battistini
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois at Chicago (UIC) Cancer Center, University of Illinois at Chicago, Chicago, IL, United States
- Jesse Brown VA Medical Center, Chicago, IL, United States
| |
Collapse
|
79
|
Yang C, Qiao T, Yu J, Wang H, Guo Y, Salameh J, Metterville J, Parsi S, Yusuf I, Brown RH, Cai H, Xu Z. Low-level overexpression of wild type TDP-43 causes late-onset, progressive neurodegeneration and paralysis in mice. PLoS One 2022; 17:e0255710. [PMID: 35113871 PMCID: PMC8812852 DOI: 10.1371/journal.pone.0255710] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Modestly increased expression of transactive response DNA binding protein (TDP-43) gene have been reported in amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and other neuromuscular diseases. However, whether this modest elevation triggers neurodegeneration is not known. Although high levels of TDP-43 overexpression have been modeled in mice and shown to cause early death, models with low-level overexpression that mimic the human condition have not been established. In this study, transgenic mice overexpressing wild type TDP-43 at less than 60% above the endogenous CNS levels were constructed, and their phenotypes analyzed by a variety of techniques, including biochemical, molecular, histological, behavioral techniques and electromyography. The TDP-43 transgene was expressed in neurons, astrocytes, and oligodendrocytes in the cortex and predominantly in astrocytes and oligodendrocytes in the spinal cord. The mice developed a reproducible progressive weakness ending in paralysis in mid-life. Detailed analysis showed ~30% loss of large pyramidal neurons in the layer V motor cortex; in the spinal cord, severe demyelination was accompanied by oligodendrocyte injury, protein aggregation, astrogliosis and microgliosis, and elevation of neuroinflammation. Surprisingly, there was no loss of lower motor neurons in the lumbar spinal cord despite the complete paralysis of the hindlimbs. However, denervation was detected at the neuromuscular junction. These results demonstrate that low-level TDP-43 overexpression can cause diverse aspects of ALS, including late-onset and progressive motor dysfunction, neuroinflammation, and neurodegeneration. Our findings suggest that persistent modest elevations in TDP-43 expression can lead to ALS and other neurological disorders involving TDP-43 proteinopathy. Because of the predictable and progressive clinical paralytic phenotype, this transgenic mouse model will be useful in preclinical trial of therapeutics targeting neurological disorders associated with elevated levels of TDP-43.
Collapse
Affiliation(s)
- Chunxing Yang
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Tao Qiao
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Jia Yu
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
| | - Hongyan Wang
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Yansu Guo
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Johnny Salameh
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Jake Metterville
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Sepideh Parsi
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Issa Yusuf
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Robert H. Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- RNA Therapeutic Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Neuroscience Program, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Huaibin Cai
- Transgenics Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States of America
| | - Zuoshang Xu
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- RNA Therapeutic Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Neuroscience Program, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
80
|
Identification of Regulatory Factors and Prognostic Markers in Amyotrophic Lateral Sclerosis. Antioxidants (Basel) 2022; 11:antiox11020303. [PMID: 35204186 PMCID: PMC8868268 DOI: 10.3390/antiox11020303] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 12/10/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive degeneration of motor neurons, leading to muscle atrophy, paralysis and even death. Immune disorder, redox imbalance, autophagy disorder, and iron homeostasis disorder have been shown to play critical roles in the pathogenesis of ALS. However, the exact pathogenic genes and the underlying mechanism of ALS remain unclear. The purpose of this study was to screen for pathogenic regulatory genes and prognostic markers in ALS using bioinformatics methods. We used Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, gene set enrichment analysis (GSEA), and expression regulation network analysis to investigate the function of differentially expressed genes in the nerve tissue, lymphoid tissue, and whole blood of patients with ALS. Our results showed that the up-regulated genes were mainly involved in immune regulation and inflammation, and the down-regulated genes were mainly involved in energy metabolism and redox processes. Eleven up-regulated transcription factors (CEBPB, CEBPD, STAT5A, STAT6, RUNX1, REL, SMAD3, GABPB2, FOXO1, PAX6, and FOXJ1) and one down-regulated transcription factor (NOG) in the nerve tissue of patients with ALS likely play important regulatory roles in the pathogenesis of ALS. Based on construction and evaluation of the ALS biomarker screening model, cluster analysis of the identified characteristic genes, univariate Cox proportional hazards regression analysis, and the random survival forest algorithm, we found that MAEA, TPST1, IFNGR2, and ALAS2 may be prognostic markers regarding the survival of ALS patients. High expression of MAEA, TPST1, and IFNGR2 and low expression of ALAS2 in ALS patients may be closely related to short survival of ALS patients. Taken together, our results indicate that immune disorders, inflammation, energy metabolism, and redox imbalance may be the important pathogenic factors of ALS. CEBPB, CEBPD, STAT5A, STAT6, RUNX1, REL, SMAD3, GABPB2, FOXO1, PAX6, FOXJ1, and NOG may be important regulatory factors linked to the pathogenesis of ALS. MAEA, TPST1, IFNGR2, and ALAS2 are potential important ALS prognostic markers. Our findings provide evidence on the pathogenesis of ALS, potential targets for the development of new drugs for ALS, and important markers for predicting ALS prognosis.
Collapse
|
81
|
Baxi EG, Thompson T, Li J, Kaye JA, Lim RG, Wu J, Ramamoorthy D, Lima L, Vaibhav V, Matlock A, Frank A, Coyne AN, Landin B, Ornelas L, Mosmiller E, Thrower S, Farr SM, Panther L, Gomez E, Galvez E, Perez D, Meepe I, Lei S, Mandefro B, Trost H, Pinedo L, Banuelos MG, Liu C, Moran R, Garcia V, Workman M, Ho R, Wyman S, Roggenbuck J, Harms MB, Stocksdale J, Miramontes R, Wang K, Venkatraman V, Holewenski R, Sundararaman N, Pandey R, Manalo DM, Donde A, Huynh N, Adam M, Wassie BT, Vertudes E, Amirani N, Raja K, Thomas R, Hayes L, Lenail A, Cerezo A, Luppino S, Farrar A, Pothier L, Prina C, Morgan T, Jamil A, Heintzman S, Jockel-Balsarotti J, Karanja E, Markway J, McCallum M, Joslin B, Alibazoglu D, Kolb S, Ajroud-Driss S, Baloh R, Heitzman D, Miller T, Glass JD, Patel-Murray NL, Yu H, Sinani E, Vigneswaran P, Sherman AV, Ahmad O, Roy P, Beavers JC, Zeiler S, Krakauer JW, Agurto C, Cecchi G, Bellard M, Raghav Y, Sachs K, Ehrenberger T, Bruce E, Cudkowicz ME, Maragakis N, Norel R, Van Eyk JE, Finkbeiner S, Berry J, Sareen D, Thompson LM, Fraenkel E, Svendsen CN, et alBaxi EG, Thompson T, Li J, Kaye JA, Lim RG, Wu J, Ramamoorthy D, Lima L, Vaibhav V, Matlock A, Frank A, Coyne AN, Landin B, Ornelas L, Mosmiller E, Thrower S, Farr SM, Panther L, Gomez E, Galvez E, Perez D, Meepe I, Lei S, Mandefro B, Trost H, Pinedo L, Banuelos MG, Liu C, Moran R, Garcia V, Workman M, Ho R, Wyman S, Roggenbuck J, Harms MB, Stocksdale J, Miramontes R, Wang K, Venkatraman V, Holewenski R, Sundararaman N, Pandey R, Manalo DM, Donde A, Huynh N, Adam M, Wassie BT, Vertudes E, Amirani N, Raja K, Thomas R, Hayes L, Lenail A, Cerezo A, Luppino S, Farrar A, Pothier L, Prina C, Morgan T, Jamil A, Heintzman S, Jockel-Balsarotti J, Karanja E, Markway J, McCallum M, Joslin B, Alibazoglu D, Kolb S, Ajroud-Driss S, Baloh R, Heitzman D, Miller T, Glass JD, Patel-Murray NL, Yu H, Sinani E, Vigneswaran P, Sherman AV, Ahmad O, Roy P, Beavers JC, Zeiler S, Krakauer JW, Agurto C, Cecchi G, Bellard M, Raghav Y, Sachs K, Ehrenberger T, Bruce E, Cudkowicz ME, Maragakis N, Norel R, Van Eyk JE, Finkbeiner S, Berry J, Sareen D, Thompson LM, Fraenkel E, Svendsen CN, Rothstein JD. Answer ALS, a large-scale resource for sporadic and familial ALS combining clinical and multi-omics data from induced pluripotent cell lines. Nat Neurosci 2022; 25:226-237. [PMID: 35115730 PMCID: PMC8825283 DOI: 10.1038/s41593-021-01006-0] [Show More Authors] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022]
Abstract
Answer ALS is a biological and clinical resource of patient-derived, induced pluripotent stem (iPS) cell lines, multi-omic data derived from iPS neurons and longitudinal clinical and smartphone data from over 1,000 patients with ALS. This resource provides population-level biological and clinical data that may be employed to identify clinical-molecular-biochemical subtypes of amyotrophic lateral sclerosis (ALS). A unique smartphone-based system was employed to collect deep clinical data, including fine motor activity, speech, breathing and linguistics/cognition. The iPS spinal neurons were blood derived from each patient and these cells underwent multi-omic analytics including whole-genome sequencing, RNA transcriptomics, ATAC-sequencing and proteomics. The intent of these data is for the generation of integrated clinical and biological signatures using bioinformatics, statistics and computational biology to establish patterns that may lead to a better understanding of the underlying mechanisms of disease, including subgroup identification. A web portal for open-source sharing of all data was developed for widespread community-based data analytics.
Collapse
Affiliation(s)
- Emily G Baxi
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Jonathan Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Julia A Kaye
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Ryan G Lim
- UCI MIND, University of California, Irvine, CA, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Divya Ramamoorthy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Leandro Lima
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Vineet Vaibhav
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andrea Matlock
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aaron Frank
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alyssa N Coyne
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barry Landin
- Computational Biology Center, IBM T.J. Watson Research Center, Yorktown Heights, NY, USA
| | - Loren Ornelas
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Elizabeth Mosmiller
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sara Thrower
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Lindsey Panther
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emilda Gomez
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Erick Galvez
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Daniel Perez
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Imara Meepe
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susan Lei
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Berhan Mandefro
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hannah Trost
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Louis Pinedo
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maria G Banuelos
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Chunyan Liu
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ruby Moran
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Veronica Garcia
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael Workman
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Richie Ho
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stacia Wyman
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Matthew B Harms
- Department of Neurology and Genetics, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jennifer Stocksdale
- Department of Psychiatry and Human Behavior and Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA
| | | | - Keona Wang
- Department of Psychiatry and Human Behavior and Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ronald Holewenski
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rakhi Pandey
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Danica-Mae Manalo
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aneesh Donde
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nhan Huynh
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Miriam Adam
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brook T Wassie
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Edward Vertudes
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Naufa Amirani
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Krishna Raja
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Reuben Thomas
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Lindsey Hayes
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alex Lenail
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aianna Cerezo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Luppino
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alanna Farrar
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lindsay Pothier
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carolyn Prina
- Department of Neurology and Genetics, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Arish Jamil
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Sarah Heintzman
- Department of Neurology and Genetics, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | | | - Jesse Markway
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Molly McCallum
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Ben Joslin
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Deniz Alibazoglu
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Stephen Kolb
- Department of Neurology and Genetics, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Robert Baloh
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Tim Miller
- Department of Neurology, Washington University, St. Louis, MO, USA
| | | | | | - Hong Yu
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ervin Sinani
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Prasha Vigneswaran
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexander V Sherman
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Omar Ahmad
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Promit Roy
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jay C Beavers
- Microsoft Research, Microsoft Corporation, Redmond, WA, USA
| | - Steven Zeiler
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John W Krakauer
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carla Agurto
- Computational Biology Center, IBM T.J. Watson Research Center, Yorktown Heights, NY, USA
| | - Guillermo Cecchi
- Computational Biology Center, IBM T.J. Watson Research Center, Yorktown Heights, NY, USA
| | - Mary Bellard
- Microsoft University Relations, Microsoft Corporation, Redmond, WA, USA
| | - Yogindra Raghav
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Karen Sachs
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tobias Ehrenberger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Elizabeth Bruce
- Microsoft University Relations, Microsoft Corporation, Redmond, WA, USA
| | - Merit E Cudkowicz
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholas Maragakis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raquel Norel
- Computational Biology Center, IBM T.J. Watson Research Center, Yorktown Heights, NY, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes and the Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - James Berry
- Department of Neurology, Healey Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dhruv Sareen
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Leslie M Thompson
- UCI MIND, University of California, Irvine, CA, USA
- Department of Biological Chemistry, University of California, Irvine, CA, USA
- Department of Psychiatry and Human Behavior and Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Clive N Svendsen
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jeffrey D Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
82
|
Pan Y, Nicolazzo JA. Altered Blood-Brain Barrier and Blood-Spinal Cord Barrier Dynamics in Amyotrophic Lateral Sclerosis: Impact on Medication Efficacy and Safety. Br J Pharmacol 2022; 179:2577-2588. [PMID: 35048358 DOI: 10.1111/bph.15802] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 11/26/2022] Open
Abstract
The access of drugs into the central nervous system (CNS) is regulated by the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB). A large body of evidence supports perturbation of these barriers in neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Modifications to the BBB and BSCB are also reported in amyotrophic lateral sclerosis (ALS), albeit these modifications have received less attention relative to those in other neurodegenerative diseases. Such alterations to the BBB and BSCB have the potential to impact on CNS exposure of drugs in ALS, modulating the effectiveness of drugs intended to reach the brain and the toxicity of drugs that are not intended to reach the brain. Given the clinical importance of these phenomena, this review will summarise reported modifications to the BBB and BSCB in ALS, discuss their impact on CNS drug exposure and suggest further research directions so as to optimise medicine use in people with ALS.
Collapse
Affiliation(s)
- Yijun Pan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
83
|
Arjmand B, Kokabi Hamidpour S, Rabbani Z, Tayanloo-Beik A, Rahim F, Aghayan HR, Larijani B. Organ on a Chip: A Novel in vitro Biomimetic Strategy in Amyotrophic Lateral Sclerosis (ALS) Modeling. Front Neurol 2022; 12:788462. [PMID: 35111126 PMCID: PMC8802668 DOI: 10.3389/fneur.2021.788462] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/20/2021] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis is a pernicious neurodegenerative disorder that is associated with the progressive degeneration of motor neurons, the disruption of impulse transmission from motor neurons to muscle cells, and the development of mobility impairments. Clinically, muscle paralysis can spread to other parts of the body. Hence it may have adverse effects on swallowing, speaking, and even breathing, which serves as major problems facing these patients. According to the available evidence, no definite treatment has been found for amyotrophic lateral sclerosis (ALS) that results in a significant outcome, although some pharmacological and non-pharmacological treatments are currently applied that are accompanied by some positive effects. In other words, available therapies are only used to relieve symptoms without any significant treatment effects that highlight the importance of seeking more novel therapies. Unfortunately, the process of discovering new drugs with high therapeutic potential for ALS treatment is fraught with challenges. The lack of a broad view of the disease process from early to late-stage and insufficiency of preclinical studies for providing validated results prior to conducting clinical trials are other reasons for the ALS drug discovery failure. However, increasing the combined application of different fields of regenerative medicine, especially tissue engineering and stem cell therapy can be considered as a step forward to develop more novel technologies. For instance, organ on a chip is one of these technologies that can provide a platform to promote a comprehensive understanding of neuromuscular junction biology and screen candidate drugs for ALS in combination with pluripotent stem cells (PSCs). The structure of this technology is based on the use of essential components such as iPSC- derived motor neurons and iPSC-derived skeletal muscle cells on a single miniaturized chip for ALS modeling. Accordingly, an organ on a chip not only can mimic ALS complexities but also can be considered as a more cost-effective and time-saving disease modeling platform in comparison with others. Hence, it can be concluded that lab on a chip can make a major contribution as a biomimetic micro-physiological system in the treatment of neurodegenerative disorders such as ALS.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Babak Arjmand
| | - Shayesteh Kokabi Hamidpour
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Rabbani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fakher Rahim
- Health Research Institute, Thalassemia, and Hemoglobinopathies Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Bagher Larijani
| |
Collapse
|
84
|
Hines TJ, Lutz C, Murray SA, Burgess RW. An Integrated Approach to Studying Rare Neuromuscular Diseases Using Animal and Human Cell-Based Models. Front Cell Dev Biol 2022; 9:801819. [PMID: 35047510 PMCID: PMC8762301 DOI: 10.3389/fcell.2021.801819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
As sequencing technology improves, the identification of new disease-associated genes and new alleles of known genes is rapidly increasing our understanding of the genetic underpinnings of rare diseases, including neuromuscular diseases. However, precisely because these disorders are rare and often heterogeneous, they are difficult to study in patient populations. In parallel, our ability to engineer the genomes of model organisms, such as mice or rats, has gotten increasingly efficient through techniques such as CRISPR/Cas9 genome editing, allowing the creation of precision human disease models. Such in vivo model systems provide an efficient means for exploring disease mechanisms and identifying therapeutic strategies. Furthermore, animal models provide a platform for preclinical studies to test the efficacy of those strategies. Determining whether the same mechanisms are involved in the human disease and confirming relevant parameters for treatment ideally involves a human experimental system. One system currently being used is induced pluripotent stem cells (iPSCs), which can then be differentiated into the relevant cell type(s) for in vitro confirmation of disease mechanisms and variables such as target engagement. Here we provide a demonstration of these approaches using the example of tRNA-synthetase-associated inherited peripheral neuropathies, rare forms of Charcot-Marie-Tooth disease (CMT). Mouse models have led to a better understanding of both the genetic and cellular mechanisms underlying the disease. To determine if the mechanisms are similar in human cells, we will use genetically engineered iPSC-based models. This will allow comparisons of different CMT-associated GARS alleles in the same genetic background, reducing the variability found between patient samples and simplifying the availability of cell-based models for a rare disease. The necessity of integrating mouse and human models, strategies for accomplishing this integration, and the challenges of doing it at scale are discussed using recently published work detailing the cellular mechanisms underlying GARS-associated CMT as a framework.
Collapse
|
85
|
Wang M, Kang L, Wang Y, Yang B, Zhang C, Lu Y, Kang L. Microglia in motor neuron disease: Signaling evidence from last 10 years. Dev Neurobiol 2022; 82:625-638. [PMID: 36309345 PMCID: PMC9828749 DOI: 10.1002/dneu.22905] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 07/09/2022] [Accepted: 10/19/2022] [Indexed: 01/30/2023]
Abstract
Motor neuron disease (MND), including amyotrophic lateral sclerosis, spinal muscular atrophy and others, involved the upper or lower motor neurons selective loss, is characterized by neurodegeneration and neuroinflammation, in conjunction with microglia. We summarized that pathways and key mediators are associated with microglia, such as fractalkine signaling, purinergic signaling, NF-κB signaling, p38 MAPK signaling, TREM2-APOE signaling, ROCK signaling, C1q signaling, and Ion channel, which are involved in the activation, proliferation, and inflammation of microglia. This review aims to identify the microglia-related molecular target and explore potential treatment strategies for MND based on that target.
Collapse
Affiliation(s)
- Min‐Jia Wang
- School of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| | - Lu Kang
- School of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| | - Yao‐Zheng Wang
- School of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| | - Bi‐Ru Yang
- Department of Postpartum RehabilitationSichuan Jinxin Women & Children HospitalChengduChina
| | - Chun Zhang
- School of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| | - Yu‐Feng Lu
- School of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| | - Liang Kang
- Institute of Sports Medicine and HealthChengdu Sports UniversityChengduChina
| |
Collapse
|
86
|
Russo K, Wharton KA. BMP/TGF-β signaling as a modulator of neurodegeneration in ALS. Dev Dyn 2022; 251:10-25. [PMID: 33745185 PMCID: PMC11929146 DOI: 10.1002/dvdy.333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 12/19/2022] Open
Abstract
This commentary focuses on the emerging intersection between BMP/TGF-β signaling roles in nervous system function and the amyotrophic lateral sclerosis (ALS) disease state. Future research is critical to elucidate the molecular underpinnings of this intersection of the cellular processes disrupted in ALS and those influenced by BMP/TGF-β signaling, including synapse structure, neurotransmission, plasticity, and neuroinflammation. Such knowledge promises to inform us of ideal entry points for the targeted modulation of dysfunctional cellular processes in an effort to abrogate ALS pathologies. It is likely that different interventions are required, either at discrete points in disease progression, or across multiple dysfunctional processes which together lead to motor neuron degeneration and death. We discuss the challenging, but intriguing idea that modulation of the pleiotropic nature of BMP/TGF-β signaling could be advantageous, as a way to simultaneously treat defects in more than one cell process across different forms of ALS.
Collapse
Affiliation(s)
- Kathryn Russo
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island, USA
| | - Kristi A Wharton
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island, USA
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
87
|
Yadav RK, Mehan S, Sahu R, Kumar S, Khan A, Makeen HA, Al Bratty M. Protective effects of apigenin on methylmercury-induced behavioral/neurochemical abnormalities and neurotoxicity in rats. Hum Exp Toxicol 2022; 41:9603271221084276. [PMID: 35373622 DOI: 10.1177/09603271221084276] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Methylmercury (MeHg) is a neurotoxin that induces neurotoxicity and cell death in neurons. MeHg increases oligodendrocyte death, glial cell activation, and motor neuron demyelination in the motor cortex and spinal cord. As a result, MeHg plays an important role in developing neurocomplications similar to amyotrophic lateral sclerosis (ALS). Recent research has implicated c-JNK and p38MAPK overactivation in the pathogenesis of ALS. Apigenin (APG) is a flavonoid having anti-inflammatory, antioxidant, and c-JNK/p38MAPK inhibitory activities. The purpose of this study is to determine whether APG possesses neuroprotective effects in MeHg-induced neurotoxicity in adult rats associated with ALS-like neuropathological alterations. In the current study, the neurotoxin MeHg causes an ALS-like phenotype in Wistar rats after 21 days of oral administration at a dose of 5 mg/kg. Prolonged administration of APG (40 and 80 mg/kg) improved neurobehavioral parameters such as learning memory, cognition, motor coordination, and grip strength. This is mainly associated with the downregulation of c-JNK and p38MAPK signaling as well as the restoration of myelin basic protein within the brain. Furthermore, APG inhibited neuronal apoptotic markers (Bax, Bcl-2, and caspase-3), restored neurotransmitter imbalance, decreased inflammatory markers (TNF- and IL-1), and alleviated oxidative damage. As a result, the current study shows that APG has neuroprotective potential as a c-JNK and p38MAPK signaling inhibitor against MeHg-induced neurotoxicity in adult rats. Based on these promising findings, we suggested that APG could be a potential new therapeutic approach over other conventional therapeutic approaches for MeHg-induced neurotoxicity in neurobehavioral, molecular, and neurochemical abnormalities.
Collapse
Affiliation(s)
- Rajeshwar Kumar Yadav
- Neuropharmacology Division, Department of Pharmacology, 75126ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, 75126ISF College of Pharmacy, Moga, Punjab, India
| | - Rakesh Sahu
- Neuropharmacology Division, Department of Pharmacology, 75126ISF College of Pharmacy, Moga, Punjab, India
| | - Sumit Kumar
- Neuropharmacology Division, Department of Pharmacology, 75126ISF College of Pharmacy, Moga, Punjab, India
| | - Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, 123285Jazan University, Jazan, Saudi Arabia
| | - Hafiz Antar Makeen
- Department of Clinical Pharmacy, College of Pharmacy, 123285Jazan University, Jazan, Saudi Arabia
| | - Mohammed Al Bratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, 123285Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
88
|
Yang W, Chen X, Li S, Li XJ. Genetically modified large animal models for investigating neurodegenerative diseases. Cell Biosci 2021; 11:218. [PMID: 34933675 PMCID: PMC8690884 DOI: 10.1186/s13578-021-00729-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 12/02/2022] Open
Abstract
Neurodegenerative diseases represent a large group of neurological disorders including Alzheimer’s disease, amyotrophic lateral sclerosis, Parkinson’s disease, and Huntington’s disease. Although this group of diseases show heterogeneous clinical and pathological phenotypes, they share important pathological features characterized by the age-dependent and progressive degeneration of nerve cells that is caused by the accumulation of misfolded proteins. The association of genetic mutations with neurodegeneration diseases has enabled the establishment of various types of animal models that mimic genetic defects and have provided important insights into the pathogenesis. However, most of genetically modified rodent models lack the overt and selective neurodegeneration seen in the patient brains, making it difficult to use the small animal models to validate the effective treatment on neurodegeneration. Recent studies of pig and monkey models suggest that large animals can more faithfully recapitulate pathological features of neurodegenerative diseases. In this review, we discuss the important differences in animal models for modeling pathological features of neurodegenerative diseases, aiming to assist the use of animal models to better understand the pathogenesis and to develop effective therapeutic strategies.
Collapse
Affiliation(s)
- Weili Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Xiusheng Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
89
|
Grivennikov IA, Tarantul VZ. Genome Editing Technology for the Study and Correction of Neurodegenerative Diseases. NEUROCHEM J+ 2021. [DOI: 10.1134/s181971242104005x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
90
|
Guida N, Sanguigno L, Mascolo L, Calabrese L, Serani A, Molinaro P, Lau CG, Annunziato L, Formisano L. The Transcriptional Complex Sp1/KMT2A by Up-Regulating Restrictive Element 1 Silencing Transcription Factor Accelerates Methylmercury-Induced Cell Death in Motor Neuron-Like NSC34 Cells Overexpressing SOD1-G93A. Front Neurosci 2021; 15:771580. [PMID: 34899171 PMCID: PMC8662822 DOI: 10.3389/fnins.2021.771580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Methylmercury (MeHg) exposure has been related to amyotrophic lateral sclerosis (ALS) pathogenesis and molecular mechanisms of its neurotoxicity has been associated to an overexpression of the Restrictive Element 1 Silencing Transcription factor (REST). Herein, we evaluated the possibility that MeHg could accelerate neuronal death of the motor neuron-like NSC34 cells transiently overexpressing the human Cu2+/Zn2+superoxide dismutase 1 (SOD1) gene mutated at glycine 93 (SOD1-G93A). Indeed, SOD1-G93A cells exposed to 100 nM MeHg for 24 h showed a reduction in cell viability, as compared to cells transfected with empty vector or with unmutated SOD1 construct. Interestingly, cell survival reduction in SOD1-G93A cells was associated with an increase of REST mRNA and protein levels. Furthermore, MeHg increased the expression of the transcriptional factor Sp1 and promoted its binding to REST gene promoter sequence. Notably, Sp1 knockdown reverted MeHg-induced REST increase. Co-immunoprecipitation experiments demonstrated that Sp1 physically interacted with the epigenetic writer Lysine-Methyltransferase-2A (KMT2A). Moreover, knocking-down of KMT2A reduced MeHg-induced REST mRNA and protein increase in SOD1-G93A cells. Finally, we found that MeHg-induced REST up-regulation triggered necropoptotic cell death, monitored by RIPK1 increased protein expression. Interestingly, REST knockdown or treatment with the necroptosis inhibitor Necrostatin-1 (Nec) decelerated MeH-induced cell death in SOD1-G93A cells. Collectively, this study demonstrated that MeHg hastens necroptotic cell death in SOD1-G93A cells via Sp1/KMT2A complex, that by epigenetic mechanisms increases REST gene expression.
Collapse
Affiliation(s)
| | - Luca Sanguigno
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Luigi Mascolo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Lucrezia Calabrese
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Angelo Serani
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Pasquale Molinaro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - C Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | | | - Luigi Formisano
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| |
Collapse
|
91
|
Sahdeo S, Goldstein DB. Translating amyotrophic lateral sclerosis genes into drug development leads. Nat Genet 2021; 53:1624-1626. [PMID: 34873336 DOI: 10.1038/s41588-021-00981-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
92
|
Bonifacino T, Zerbo RA, Balbi M, Torazza C, Frumento G, Fedele E, Bonanno G, Milanese M. Nearly 30 Years of Animal Models to Study Amyotrophic Lateral Sclerosis: A Historical Overview and Future Perspectives. Int J Mol Sci 2021; 22:ijms222212236. [PMID: 34830115 PMCID: PMC8619465 DOI: 10.3390/ijms222212236] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, multigenic, multifactorial, and non-cell autonomous neurodegenerative disease characterized by upper and lower motor neuron loss. Several genetic mutations lead to ALS development and many emerging gene mutations have been discovered in recent years. Over the decades since 1990, several animal models have been generated to study ALS pathology including both vertebrates and invertebrates such as yeast, worms, flies, zebrafish, mice, rats, guinea pigs, dogs, and non-human primates. Although these models show different peculiarities, they are all useful and complementary to dissect the pathological mechanisms at the basis of motor neuron degeneration and ALS progression, thus contributing to the development of new promising therapeutics. In this review, we describe the up to date and available ALS genetic animal models, classified by the different genetic mutations and divided per species, pointing out their features in modeling, the onset and progression of the pathology, as well as their specific pathological hallmarks. Moreover, we highlight similarities, differences, advantages, and limitations, aimed at helping the researcher to select the most appropriate experimental animal model, when designing a preclinical ALS study.
Collapse
Affiliation(s)
- Tiziana Bonifacino
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Genoa, Italy
| | - Roberta Arianna Zerbo
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Matilde Balbi
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Carola Torazza
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Giulia Frumento
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Ernesto Fedele
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence:
| | - Giambattista Bonanno
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Marco Milanese
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Genoa, Italy
| |
Collapse
|
93
|
Ahmadi A, Gispert JD, Navarro A, Vilor-Tejedor N, Sadeghi I. Single-cell Transcriptional Changes in Neurodegenerative Diseases. Neuroscience 2021; 479:192-205. [PMID: 34748859 DOI: 10.1016/j.neuroscience.2021.10.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 01/25/2023]
Abstract
In recent decades, our understanding of the molecular changes involved in neurodegenerative diseases has been transformed. Single-cell RNA sequencing and single-nucleus RNA sequencing technologies have been applied to provide cellular and molecular details of the brain at the single-cell level. This has expanded our knowledge of the central nervous system and provided insights into the molecular vulnerability of brain cell types and underlying mechanisms in neurodegenerative diseases. In this review, we highlight the recent advances and findings related to neurodegenerative diseases using these cutting-edge technologies.
Collapse
Affiliation(s)
- Amirhossein Ahmadi
- Department of Biology, Faculty of Nano and BioScience and Technology, Persian Gulf University, Bushehr 75169, Iran
| | - Juan D Gispert
- BarcelonaBeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Arcadi Navarro
- BarcelonaBeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Natalia Vilor-Tejedor
- BarcelonaBeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Erasmus MC University Medical Center. Department of Clinical Genetics, Rotterdam, the Netherlands.
| | - Iman Sadeghi
- BarcelonaBeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain.
| |
Collapse
|
94
|
Sumien N, Cunningham JT, Davis DL, Engelland R, Fadeyibi O, Farmer GE, Mabry S, Mensah-Kane P, Trinh OTP, Vann PH, Wilson EN, Cunningham RL. Neurodegenerative Disease: Roles for Sex, Hormones, and Oxidative Stress. Endocrinology 2021; 162:6360925. [PMID: 34467976 PMCID: PMC8462383 DOI: 10.1210/endocr/bqab185] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Indexed: 02/08/2023]
Abstract
Neurodegenerative diseases cause severe impairments in cognitive and motor function. With an increasing aging population and the onset of these diseases between 50 and 70 years, the consequences are bound to be devastating. While age and longevity are the main risk factors for neurodegenerative diseases, sex is also an important risk factor. The characteristic of sex is multifaceted, encompassing sex chromosome complement, sex hormones (estrogens and androgens), and sex hormone receptors. Sex hormone receptors can induce various signaling cascades, ranging from genomic transcription to intracellular signaling pathways that are dependent on the health of the cell. Oxidative stress, associated with aging, can impact the health of the cell. Sex hormones can be neuroprotective under low oxidative stress conditions but not in high oxidative stress conditions. An understudied sex hormone receptor that can induce activation of oxidative stress signaling is the membrane androgen receptor (mAR). mAR can mediate nicotinamide adenine dinucleotide-phosphate (NADPH) oxidase (NOX)-generated oxidative stress that is associated with several neurodegenerative diseases, such as Alzheimer disease. Further complicating this is that aging can alter sex hormone signaling. Prior to menopause, women experience more estrogens than androgens. During menopause, this sex hormone profile switches in women due to the dramatic ovarian loss of 17β-estradiol with maintained ovarian androgen (testosterone, androstenedione) production. Indeed, aging men have higher estrogens than aging women due to aromatization of androgens to estrogens. Therefore, higher activation of mAR-NOX signaling could occur in menopausal women compared with aged men, mediating the observed sex differences. Understanding of these signaling cascades could provide therapeutic targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Nathalie Sumien
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - J Thomas Cunningham
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Delaney L Davis
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rachel Engelland
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Oluwadarasimi Fadeyibi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - George E Farmer
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Steve Mabry
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Paapa Mensah-Kane
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Oanh T P Trinh
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Philip H Vann
- Department of Pharmacology & Neuroscience, Center for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - E Nicole Wilson
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Correspondence: Rebecca L. Cunningham, PhD, Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3400 Camp Bowie Boulevard, Fort Worth, TX, USA, 76107-2699.
| |
Collapse
|
95
|
Sun J, Huang T, Debelius JW, Fang F. Gut microbiome and amyotrophic lateral sclerosis: A systematic review of current evidence. J Intern Med 2021; 290:758-788. [PMID: 34080741 DOI: 10.1111/joim.13336] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS), characterized by a loss of motor neurons in the brain and spinal cord, is a relatively rare but currently incurable neurodegenerative disease. The global incidence of ALS is estimated as 1.75 per 100,000 person-years and the global prevalence is estimated as 4.1-8.4 per 100,000 individuals. Contributions from outside the central nervous system to the etiology of ALS have been increasingly recognized. Gut microbiome is one of the most quickly growing fields of research for ALS. In this article, we performed a comprehensive review of the results from existing animal and human studies, to provide an up-to-date summary of the current research on gut microbiome and ALS. In brief, we found relatively consistent results from animal studies, suggesting an altered gut microbiome composition in experimental ALS. Publication bias might however be a concern. Findings from human studies are largely inconclusive. A few animal and human studies demonstrated the usefulness of intervention with microbial-derived metabolites in modulating the disease progression of ALS. We discussed potential methodological concerns in these studies, including study design, statistical power, handling process of biospecimens and sequencing data, as well as statistical methods and interpretation of results. Finally, we made a few proposals for continued microbiome research in ALS, with the aim to provide valid, reproducible, and translatable findings.
Collapse
Affiliation(s)
- Jiangwei Sun
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tingting Huang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Justine W Debelius
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Fang Fang
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
96
|
Moreno-Jiménez L, Benito-Martín M, Sanclemente-Alamán I, Matías-Guiu J, Sancho-Bielsa F, Canales-Aguirre A, Mateos-Díaz J, Matías-Guiu J, Aguilar J, Gómez-Pinedo U. Modelos experimentales murinos en la esclerosis lateral amiotrófica. Puesta al día. Neurologia 2021. [DOI: 10.1016/j.nrl.2021.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
97
|
Laneve P, Tollis P, Caffarelli E. RNA Deregulation in Amyotrophic Lateral Sclerosis: The Noncoding Perspective. Int J Mol Sci 2021; 22:10285. [PMID: 34638636 PMCID: PMC8508793 DOI: 10.3390/ijms221910285] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 12/18/2022] Open
Abstract
RNA metabolism is central to cellular physiopathology. Almost all the molecular pathways underpinning biological processes are affected by the events governing the RNA life cycle, ranging from transcription to degradation. The deregulation of these processes contributes to the onset and progression of human diseases. In recent decades, considerable efforts have been devoted to the characterization of noncoding RNAs (ncRNAs) and to the study of their role in the homeostasis of the nervous system (NS), where they are highly enriched. Acting as major regulators of gene expression, ncRNAs orchestrate all the steps of the differentiation programs, participate in the mechanisms underlying neural functions, and are crucially implicated in the development of neuronal pathologies, among which are neurodegenerative diseases. This review aims to explore the link between ncRNA dysregulation and amyotrophic lateral sclerosis (ALS), the most frequent motoneuron (MN) disorder in adults. Notably, defective RNA metabolism is known to be largely associated with this pathology, which is often regarded as an RNA disease. We also discuss the potential role that these transcripts may play as diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Pietro Laneve
- Institute of Molecular Biology and Pathology, National Research Council, 00185 Rome, Italy
| | - Paolo Tollis
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy;
| | - Elisa Caffarelli
- Institute of Molecular Biology and Pathology, National Research Council, 00185 Rome, Italy
| |
Collapse
|
98
|
Santoso JW, Li X, Gupta D, Suh GC, Hendricks E, Lin S, Perry S, Ichida JK, Dickman D, McCain ML. Engineering skeletal muscle tissues with advanced maturity improves synapse formation with human induced pluripotent stem cell-derived motor neurons. APL Bioeng 2021; 5:036101. [PMID: 34286174 PMCID: PMC8282350 DOI: 10.1063/5.0054984] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
To develop effective cures for neuromuscular diseases, human-relevant in vitro models of neuromuscular tissues are critically needed to probe disease mechanisms on a cellular and molecular level. However, previous attempts to co-culture motor neurons and skeletal muscle have resulted in relatively immature neuromuscular junctions (NMJs). In this study, NMJs formed by human induced pluripotent stem cell (hiPSC)-derived motor neurons were improved by optimizing the maturity of the co-cultured muscle tissue. First, muscle tissues engineered from the C2C12 mouse myoblast cell line, cryopreserved primary human myoblasts, and freshly isolated primary chick myoblasts on micromolded gelatin hydrogels were compared. After three weeks, only chick muscle tissues remained stably adhered to hydrogels and exhibited progressive increases in myogenic index and stress generation, approaching values generated by native muscle tissue. After three weeks of co-culture with hiPSC-derived motor neurons, engineered chick muscle tissues formed NMJs with increasing co-localization of pre- and postsynaptic markers as well as increased frequency and magnitude of synaptic activity, surpassing structural and functional maturity of previous in vitro models. Engineered chick muscle tissues also demonstrated increased expression of genes related to sarcomere maturation and innervation over time, revealing new insights into the molecular pathways that likely contribute to enhanced NMJ formation. These approaches for engineering advanced neuromuscular tissues with relatively mature NMJs and interrogating their structure and function have many applications in neuromuscular disease modeling and drug development.
Collapse
Affiliation(s)
- Jeffrey W. Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Xiling Li
- Department of Biological Sciences, Dornsife College of Arts and Letters, University of Southern California, Los Angeles, California 90089, USA
| | - Divya Gupta
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Gio C. Suh
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Eric Hendricks
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| | - Shaoyu Lin
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| | - Sarah Perry
- Department of Biological Sciences, Dornsife College of Arts and Letters, University of Southern California, Los Angeles, California 90089, USA
| | - Justin K. Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| | - Dion Dickman
- Department of Biological Sciences, Dornsife College of Arts and Letters, University of Southern California, Los Angeles, California 90089, USA
| | - Megan L. McCain
- Author to whom correspondence should be addressed:. Tel: +1 2138210791. URL:https://livingsystemsengineering.usc.edu
| |
Collapse
|
99
|
Roudeau S, Trist BG, Carmona A, Davies KM, Halliday GM, Rufin Y, Claverol S, Van Malderen SJM, Falkenberg G, Double KL, Ortega R. Native Separation and Metallation Analysis of SOD1 Protein from the Human Central Nervous System: a Methodological Workflow. Anal Chem 2021; 93:11108-11115. [PMID: 34348022 DOI: 10.1021/acs.analchem.1c01128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Studies of the metal content of metalloproteins in tissues from the human central nervous system (CNS) can be compromised by preparative techniques which alter levels of, or interactions between, metals and the protein of interest within a complex mixture. We developed a methodological workflow combining size exclusion chromatography, native isoelectric focusing, and either proton or synchrotron X-ray fluorescence within electrophoresis gels to analyze the endogenous metal content of copper-zinc superoxide dismutase (SOD1) purified from minimal amounts (<20 mg) of post-mortem human brain and spinal cord tissue. Abnormal metallation and aggregation of SOD1 are suspected to play a role in amyotrophic lateral sclerosis and Parkinson's disease, but data describing SOD1 metal occupancy in human tissues have not previously been reported. Validating our novel approach, we demonstrated step-by-step metal preservation, preserved SOD1 activity, and substantial enrichment of SOD1 protein versus confounding metalloproteins. We analyzed tissues from nine healthy individuals and five CNS regions (occipital cortex, substantia nigra, locus coeruleus, dorsal spinal cord, and ventral spinal cord). We found that Cu and Zn were bound to SOD1 in a ratio of 1.12 ± 0.28, a ratio very close to the expected value of 1. Our methodological workflow can be applied to the study of endogenous native SOD1 in a pathological context and adapted to a range of metalloproteins from human tissues and other sources.
Collapse
Affiliation(s)
- Stéphane Roudeau
- Univ. Bordeaux, CNRS, CENBG, UMR-5797, F-33170 Gradignan, France
| | - Benjamin G Trist
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Camperdown, Sydney, New South Wales 2050, Australia
| | | | - Katherine M Davies
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Camperdown, Sydney, New South Wales 2050, Australia
| | - Glenda M Halliday
- Brain and Mind Centre and Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Camperdown, Sydney, New South Wales 2050, Australia
| | - Yann Rufin
- Plateforme Biochimie et Biophysique (BioProt), Univ. Bordeaux, F-33077 Bordeaux, France
| | - Stéphane Claverol
- Plateforme Proteome, Univ. Bordeaux, Camperdown, F-33076 Bordeaux, France
| | | | | | - Kay L Double
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Camperdown, Sydney, New South Wales 2050, Australia
| | - Richard Ortega
- Univ. Bordeaux, CNRS, CENBG, UMR-5797, F-33170 Gradignan, France
| |
Collapse
|
100
|
Pikatza-Menoio O, Elicegui A, Bengoetxea X, Naldaiz-Gastesi N, López de Munain A, Gerenu G, Gil-Bea FJ, Alonso-Martín S. The Skeletal Muscle Emerges as a New Disease Target in Amyotrophic Lateral Sclerosis. J Pers Med 2021; 11:671. [PMID: 34357138 PMCID: PMC8307751 DOI: 10.3390/jpm11070671] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 01/02/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons (MNs) and severe muscle atrophy without effective treatment. Most research on ALS has been focused on the study of MNs and supporting cells of the central nervous system. Strikingly, the recent observations of pathological changes in muscle occurring before disease onset and independent from MN degeneration have bolstered the interest for the study of muscle tissue as a potential target for delivery of therapies for ALS. Skeletal muscle has just been described as a tissue with an important secretory function that is toxic to MNs in the context of ALS. Moreover, a fine-tuning balance between biosynthetic and atrophic pathways is necessary to induce myogenesis for muscle tissue repair. Compromising this response due to primary metabolic abnormalities in the muscle could trigger defective muscle regeneration and neuromuscular junction restoration, with deleterious consequences for MNs and thereby hastening the development of ALS. However, it remains puzzling how backward signaling from the muscle could impinge on MN death. This review provides a comprehensive analysis on the current state-of-the-art of the role of the skeletal muscle in ALS, highlighting its contribution to the neurodegeneration in ALS through backward-signaling processes as a newly uncovered mechanism for a peripheral etiopathogenesis of the disease.
Collapse
Affiliation(s)
- Oihane Pikatza-Menoio
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Amaia Elicegui
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Xabier Bengoetxea
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
| | - Neia Naldaiz-Gastesi
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Adolfo López de Munain
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
- Department of Neurology, Donostialdea Integrated Health Organization, Osakidetza Basque Health Service, 20014 Donostia/San Sebastián, Spain
- Department of Neurosciences, Faculty of Medicine and Nursery, University of the Basque Country UPV-EHU, 20014 Donostia/San Sebastián, Spain
| | - Gorka Gerenu
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
- Department of Physiology, University of the Basque Country UPV-EHU, 48940 Leioa, Spain
| | - Francisco Javier Gil-Bea
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Sonia Alonso-Martín
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| |
Collapse
|