1
|
Ufarry Alvarado AJ, Zaidi Pons MA, Plaza Hernández J, Torres Ortiz C. Improvements of Paraquat Treatment in Liquid Media for Behavior and Neurodegenerative Tests. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001273. [PMID: 40226510 PMCID: PMC11993906 DOI: 10.17912/micropub.biology.001273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 01/27/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a disease characterized by misfolded and aggregated proteins that have toxic effects on motor neurons. The missense mutation, G85R, of the sod-1 gene associated with ALS displays locomotor impairments in Caenorhadbitis elegans ( C. elegans ). We treated the sod-1 (G85R) strain with 0 and 2.5 mM Paraquat treatments in a liquid M9 buffer for 4 hours and in solid NGM media for 18 hours. In both methodologies, the locomotion defects and neurodegeneration were significantly increased with 2.5 mM Paraquat. Our work provides evidence of methodology that is more cost effective, rapid and reproducible to perform behavior and neurodegenerative assay in worms.
Collapse
Affiliation(s)
- Axel J. Ufarry Alvarado
- Deparment of Biomedical Sciences , Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico
| | - Malik A. Zaidi Pons
- Deparment of Biomedical Sciences, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico
| | - Jayvier Plaza Hernández
- Deparment of Natural Sciences, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico
| | - Ceidy Torres Ortiz
- Deparment of Natural Sciences, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico
| |
Collapse
|
2
|
Torres AK, Mira RG, Pinto C, Inestrosa NC. Studying the mechanisms of neurodegeneration: C. elegans advantages and opportunities. Front Cell Neurosci 2025; 19:1559151. [PMID: 40207239 PMCID: PMC11979225 DOI: 10.3389/fncel.2025.1559151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Caenorhabditis elegans has been widely used as a model organism in neurodevelopment for several decades due to its simplicity, rapid growth, short life cycle, transparency, and rather simple genetics. It has been useful in modeling neurodegenerative diseases by the heterologous expression of the major proteins that form neurodegenerative-linked aggregates such as amyloid-β peptide, tau protein, and α-synuclein, among others. Furthermore, chemical treatments as well as the existence of several interference RNA libraries, transgenic worm lines, and the possibility of generating new transgenic strains create a magnificent range of possible tools to study the signaling pathways that could confer protection against protein aggregates or, on the contrary, are playing a detrimental role. In this review, we summarize the different C. elegans models of neurodegenerative diseases with a focus on Alzheimer's and Parkinson's diseases and how genetic tools could be used to dissect the signaling pathways involved in their pathogenesis mentioning several examples. Finally, we discuss the use of pharmacological agents in C. elegans models that could help to study these disease-associated signaling pathways and the powerful combinations of experimental designs with genetic tools. This review highlights the advantages of C. elegans as a valuable intermediary between in vitro and mammalian in vivo models in the development of potential new therapies.
Collapse
Affiliation(s)
- Angie K. Torres
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo G. Mira
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Cristina Pinto
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Nibaldo C. Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
3
|
Lescouzères L, Patten SA. Promising animal models for amyotrophic lateral sclerosis drug discovery: a comprehensive update. Expert Opin Drug Discov 2024; 19:1213-1233. [PMID: 39115327 DOI: 10.1080/17460441.2024.2387791] [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: 03/08/2024] [Accepted: 07/30/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons. Several animal models have been generated to understand ALS pathogenesis. They have provided valuable insight into disease mechanisms and the development of therapeutic strategies. AREAS COVERED In this review, the authors provide a concise overview of simple genetic model organisms, including C. elegans, Drosophila, zebrafish, and mouse genetic models that have been generated to study ALS. They emphasize the benefits of each model and their application in translational research for discovering new chemicals, gene therapy approaches, and antibody-based strategies for treating ALS. EXPERT OPINION Significant progress is being made in identifying new therapeutic targets for ALS. This progress is being enabled by promising animal models of the disease using increasingly effective genetic and pharmacological strategies. There are still challenges to be overcome in order to achieve improved success rates for translating drugs from animal models to clinics for treating ALS. Several promising future directions include the establishment of novel preclinical protocol standards, as well as the combination of animal models with human induced pluripotent stem cells (iPSCs).
Collapse
Affiliation(s)
- Léa Lescouzères
- INRS - Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Shunmoogum A Patten
- INRS - Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
- Departement de Neurosciences, Université de Montréal, Montreal, Canada
| |
Collapse
|
4
|
Elsayyid M, Tanis JE, Yu Y. In-cell processing enables rapid and in-depth proteome analysis of low-input Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613705. [PMID: 39345438 PMCID: PMC11429863 DOI: 10.1101/2024.09.18.613705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Caenorhabditis elegans is a widely used genetic model organism, however, the worm cuticle complicates extraction of intracellular proteins, a prerequisite for typical bottom-up proteomics. Conventional physical disruption procedures are not only time-consuming, but can also cause significant sample loss, making it difficult to perform proteomics with low-input samples. Here, for the first time, we present an on-filter in-cell (OFIC) processing approach, which can digest C. elegans proteins directly in the cells of the organism after methanol fixation. With OFIC processing and single-shot LCMS analysis, we identified over 9,400 proteins from a sample of only 200 worms, the largest C. elegans proteome reported to date that did not require fractionation or enrichment. We systematically evaluated the performance of the OFIC approach by comparing it with conventional lysis-based methods. Our data suggest equivalent and unbiased performance of OFIC processing for C. elegans proteome identification and quantitation. We further evaluated the OFIC approach with even lower input samples, then used this method to determine how the proteome is impacted by loss of superoxide dismutase sod-1, the ortholog of human SOD-1, a gene associated with amyotrophic lateral sclerosis (ALS). Analysis of 8,800 proteins from only 50 worms as the initial input showed that loss of sod-1 affects the abundance of proteins required for stress response, ribosome biogenesis, and metabolism. In conclusion, our streamlined OFIC approach, which can be broadly applied to other systems, minimizes sample loss while offering the simplest workflow reported to date for C. elegans proteomics analysis.
Collapse
Affiliation(s)
- Malek Elsayyid
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Jessica E. Tanis
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Yanbao Yu
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, USA
| |
Collapse
|
5
|
Romussi S, Giunti S, Andersen N, De Rosa MJ. C. elegans: a prominent platform for modeling and drug screening in neurological disorders. Expert Opin Drug Discov 2024; 19:565-585. [PMID: 38509691 DOI: 10.1080/17460441.2024.2329103] [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: 11/13/2023] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Human neurodevelopmental and neurodegenerative diseases (NDevDs and NDegDs, respectively) encompass a broad spectrum of disorders affecting the nervous system with an increasing incidence. In this context, the nematode C. elegans, has emerged as a benchmark model for biological research, especially in the field of neuroscience. AREAS COVERED The authors highlight the numerous advantages of this tiny worm as a model for exploring nervous system pathologies and as a platform for drug discovery. There is a particular focus given to describing the existing models of C. elegans for the study of NDevDs and NDegDs. Specifically, the authors underscore their strong applicability in preclinical drug development. Furthermore, they place particular emphasis on detailing the common techniques employed to explore the nervous system in both healthy and diseased states. EXPERT OPINION Drug discovery constitutes a long and expensive process. The incorporation of invertebrate models, such as C. elegans, stands as an exemplary strategy for mitigating costs and expediting timelines. The utilization of C. elegans as a platform to replicate nervous system pathologies and conduct high-throughput automated assays in the initial phases of drug discovery is pivotal for rendering therapeutic options more attainable and cost-effective.
Collapse
Affiliation(s)
- Stefano Romussi
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
| | - Sebastián Giunti
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Natalia Andersen
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - María José De Rosa
- Laboratorio de Neurobiología de Invertebrados, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), UNS-CONICET, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| |
Collapse
|
6
|
Zhou L, Xu R. Invertebrate genetic models of amyotrophic lateral sclerosis. Front Mol Neurosci 2024; 17:1328578. [PMID: 38500677 PMCID: PMC10944931 DOI: 10.3389/fnmol.2024.1328578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/24/2024] [Indexed: 03/20/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a common adult-onset neurodegenerative disease characterized by the progressive death of motor neurons in the cerebral cortex, brain stem, and spinal cord. The exact mechanisms underlying the pathogenesis of ALS remain unclear. The current consensus regarding the pathogenesis of ALS suggests that the interaction between genetic susceptibility and harmful environmental factors is a promising cause of ALS onset. The investigation of putative harmful environmental factors has been the subject of several ongoing studies, but the use of transgenic animal models to study ALS has provided valuable information on the onset of ALS. Here, we review the current common invertebrate genetic models used to study the pathology, pathophysiology, and pathogenesis of ALS. The considerations of the usage, advantages, disadvantages, costs, and availability of each invertebrate model will also be discussed.
Collapse
Affiliation(s)
- LiJun Zhou
- Department of Neurology, National Regional Center for Neurological Diseases, Clinical College of Nanchang Medical College, Jiangxi Provincial People's Hospital, First Affiliated Hospital of Nanchang Medical College, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, Jiangxi, China
- Medical College of Nanchang University, Nanchang, China
| | - RenShi Xu
- Department of Neurology, National Regional Center for Neurological Diseases, Clinical College of Nanchang Medical College, Jiangxi Provincial People's Hospital, First Affiliated Hospital of Nanchang Medical College, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, Jiangxi, China
- Medical College of Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Eck RJ, Stair JG, Kraemer BC, Liachko NF. Simple models to understand complex disease: 10 years of progress from Caenorhabditis elegans models of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Front Neurosci 2024; 17:1300705. [PMID: 38239833 PMCID: PMC10794587 DOI: 10.3389/fnins.2023.1300705] [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: 09/23/2023] [Accepted: 11/28/2023] [Indexed: 01/22/2024] Open
Abstract
The nematode Caenorhabditis elegans are a powerful model system to study human disease, with numerous experimental advantages including significant genetic and cellular homology to vertebrate animals, a short lifespan, and tractable behavioral, molecular biology and imaging assays. Beginning with the identification of SOD1 as a genetic cause of amyotrophic lateral sclerosis (ALS), C. elegans have contributed to a deeper understanding of the mechanistic underpinnings of this devastating neurodegenerative disease. More recently this work has expanded to encompass models of other types of ALS and the related disease frontotemporal lobar degeneration (FTLD-TDP), including those characterized by mutation or accumulation of the proteins TDP-43, C9orf72, FUS, HnRNPA2B1, ALS2, DCTN1, CHCHD10, ELP3, TUBA4A, CAV1, UBQLN2, ATXN3, TIA1, KIF5A, VAPB, GRN, and RAB38. In this review we summarize these models and the progress and insights from the last ten years of using C. elegans to study the neurodegenerative diseases ALS and FTLD-TDP.
Collapse
Affiliation(s)
- Randall J. Eck
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Jade G. Stair
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Brian C. Kraemer
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Nicole F. Liachko
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| |
Collapse
|
8
|
Kim H, Kim GS, Hyun SH, Kim E. Advancements in 2D and 3D In Vitro Models for Studying Neuromuscular Diseases. Int J Mol Sci 2023; 24:17006. [PMID: 38069329 PMCID: PMC10707046 DOI: 10.3390/ijms242317006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Neuromuscular diseases (NMDs) are a genetically or clinically heterogeneous group of diseases that involve injury or dysfunction of neuromuscular tissue components, including peripheral motor neurons, skeletal muscles, and neuromuscular junctions. To study NMDs and develop potential therapies, remarkable progress has been made in generating in vitro neuromuscular models using engineering approaches to recapitulate the complex physical and biochemical microenvironments of 3D human neuromuscular tissues. In this review, we discuss recent studies focusing on the development of in vitro co-culture models of human motor neurons and skeletal muscles, with the pros and cons of each approach. Furthermore, we explain how neuromuscular in vitro models recapitulate certain aspects of specific NMDs, including amyotrophic lateral sclerosis and muscular dystrophy. Research on neuromuscular organoids (NMO) will continue to co-develop to better mimic tissues in vivo and will provide a better understanding of the development of the neuromuscular tissue, mechanisms of NMD action, and tools applicable to preclinical studies, including drug screening and toxicity tests.
Collapse
Affiliation(s)
- Haneul Kim
- Laboratory of Molecular Diagnostics and Cell Biology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Gon Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea;
- Institute for Stem Cell & Regenerative Medicine, Chungbuk National University, Chengju 28644, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Eunhye Kim
- Laboratory of Molecular Diagnostics and Cell Biology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea;
| |
Collapse
|
9
|
Tsioras K, Smith KC, Edassery SL, Garjani M, Li Y, Williams C, McKenna ED, Guo W, Wilen AP, Hark TJ, Marklund SL, Ostrow LW, Gilthorpe JD, Ichida JK, Kalb RG, Savas JN, Kiskinis E. Analysis of proteome-wide degradation dynamics in ALS SOD1 iPSC-derived patient neurons reveals disrupted VCP homeostasis. Cell Rep 2023; 42:113160. [PMID: 37776851 PMCID: PMC10785776 DOI: 10.1016/j.celrep.2023.113160] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/18/2023] [Accepted: 09/06/2023] [Indexed: 10/02/2023] Open
Abstract
Mutations in SOD1 cause amyotrophic lateral sclerosis (ALS) through gain-of-function effects, yet the mechanisms by which misfolded mutant SOD1 (mutSOD1) protein impairs human motor neurons (MNs) remain unclear. Here, we use induced-pluripotent-stem-cell-derived MNs coupled to metabolic stable isotope labeling and mass spectrometry to investigate proteome-wide degradation dynamics. We find several proteins, including the ALS-causal valosin-containing protein (VCP), which predominantly acts in proteasome degradation and autophagy, that degrade slower in mutSOD1 relative to isogenic control MNs. The interactome of VCP is altered in mutSOD1 MNs in vitro, while VCP selectively accumulates in the affected motor cortex of ALS-SOD1 patients. Overexpression of VCP rescues mutSOD1 toxicity in MNs in vitro and in a C. elegans model in vivo, in part due to its ability to modulate the degradation of insoluble mutSOD1. Our results demonstrate that VCP contributes to mutSOD1-dependent degeneration, link two distinct ALS-causal genes, and highlight selective protein degradation impairment in ALS pathophysiology.
Collapse
Affiliation(s)
- Konstantinos Tsioras
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kevin C Smith
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Seby L Edassery
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mehraveh Garjani
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yichen Li
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Chloe Williams
- Department of Integrative Medical Biology, Umeå University, 90187 Umeå, Sweden
| | - Elizabeth D McKenna
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Wenxuan Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Anika P Wilen
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Timothy J Hark
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Stefan L Marklund
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Lyle W Ostrow
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | | | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Robert G Kalb
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jeffrey N Savas
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
10
|
Zhu L, Li S, Li XJ, Yin P. Pathological insights from amyotrophic lateral sclerosis animal models: comparisons, limitations, and challenges. Transl Neurodegener 2023; 12:46. [PMID: 37730668 PMCID: PMC10510301 DOI: 10.1186/s40035-023-00377-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/01/2023] [Indexed: 09/22/2023] Open
Abstract
In order to dissect amyotrophic lateral sclerosis (ALS), a multigenic, multifactorial, and progressive neurodegenerative disease with heterogeneous clinical presentations, researchers have generated numerous animal models to mimic the genetic defects. Concurrent and comparative analysis of these various models allows identification of the causes and mechanisms of ALS in order to finally obtain effective therapeutics. However, most genetically modified rodent models lack overt pathological features, imposing challenges and limitations in utilizing them to rigorously test the potential mechanisms. Recent studies using large animals, including pigs and non-human primates, have uncovered important events that resemble neurodegeneration in patients' brains but could not be produced in small animals. Here we describe common features as well as discrepancies among these models, highlighting new insights from these models. Furthermore, we will discuss how to make rodent models more capable of recapitulating important pathological features based on the important pathogenic insights from large animal models.
Collapse
Affiliation(s)
- Longhong Zhu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Peng Yin
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
11
|
Kalia M, Miotto M, Ness D, Opie-Martin S, Spargo TP, Di Rienzo L, Biagini T, Petrizzelli F, Al Khleifat A, Kabiljo R, Project MinE ALS Sequencing Consortium, SOD1-ALS clinical and genetic data collection group, Mazza T, Ruocco G, Milanetti E, Dobson RJB, Al-Chalabi A, Iacoangeli A. Molecular dynamics analysis of superoxide dismutase 1 mutations suggests decoupling between mechanisms underlying ALS onset and progression. Comput Struct Biotechnol J 2023; 21:5296-5308. [PMID: 37954145 PMCID: PMC10637862 DOI: 10.1016/j.csbj.2023.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 11/14/2023] Open
Abstract
Mutations in the superoxide dismutase 1 (SOD1) gene are the second most common known cause of ALS. SOD1 variants express high phenotypic variability and over 200 have been reported in people with ALS. It was previously proposed that variants can be broadly classified in two groups, 'wild-type like' (WTL) and 'metal binding region' (MBR) variants, based on their structural location and biophysical properties. MBR variants, but not WTL variants, were associated with a reduction of SOD1 enzymatic activity. In this study we used molecular dynamics and large clinical datasets to characterise the differences in the structural and dynamic behaviour of WTL and MBR variants with respect to the wild-type SOD1, and how such differences influence the ALS clinical phenotype. Our study identified marked structural differences, some of which are observed in both variant groups, while others are group specific. Moreover, collecting clinical data of approximately 500 SOD1 ALS patients carrying variants, we showed that the survival time of patients carrying an MBR variant is generally longer (∼6 years median difference, p < 0.001) with respect to patients with a WTL variant. In conclusion, our study highlighted key differences in the dynamic behaviour between WTL and MBR SOD1 variants, and between variants and wild-type SOD1 at an atomic and molecular level, that could be further investigated to explain the associated phenotypic variability. Our results support the hypothesis of a decoupling between mechanisms of onset and progression of SOD1 ALS, and an involvement of loss-of-function of SOD1 with the disease progression.
Collapse
Affiliation(s)
- Munishikha Kalia
- Department of Biostatistics and Health Informatics, King’s College London, London, UK
- Department of Basic and Clinical Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Mattia Miotto
- Center for Life Nano & Neuro Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Deborah Ness
- Department of Biostatistics and Health Informatics, King’s College London, London, UK
- Department of Basic and Clinical Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Sarah Opie-Martin
- Department of Basic and Clinical Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Thomas P. Spargo
- Department of Biostatistics and Health Informatics, King’s College London, London, UK
- Department of Basic and Clinical Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Lorenzo Di Rienzo
- Center for Life Nano & Neuro Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Tommaso Biagini
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, S. Giovanni Rotondo, Italy
| | - Francesco Petrizzelli
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, S. Giovanni Rotondo, Italy
| | - Ahmad Al Khleifat
- Department of Basic and Clinical Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Renata Kabiljo
- Department of Biostatistics and Health Informatics, King’s College London, London, UK
- Department of Basic and Clinical Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | | | | | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, S. Giovanni Rotondo, Italy
| | - Giancarlo Ruocco
- Center for Life Nano & Neuro Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
- Department of Physics, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Edoardo Milanetti
- Center for Life Nano & Neuro Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
- Department of Physics, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Richard JB Dobson
- Department of Biostatistics and Health Informatics, King’s College London, London, UK
- Institute of Health Informatics, University College London, London, UK
- National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust King’s College London, London, United Kingdom
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
- Clinical Neurosciences, King’s College Hospital, Denmark Hill, London, UK
| | - Alfredo Iacoangeli
- Department of Biostatistics and Health Informatics, King’s College London, London, UK
- Department of Basic and Clinical Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
- National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust King’s College London, London, United Kingdom
| |
Collapse
|
12
|
Shi Y, Zhao Y, Lu L, Gao Q, Yu D, Sun M. CRISPR/Cas9: implication for modeling and therapy of amyotrophic lateral sclerosis. Front Neurosci 2023; 17:1223777. [PMID: 37483353 PMCID: PMC10359984 DOI: 10.3389/fnins.2023.1223777] [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: 05/16/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a deadly neurological disease with a complicated and variable pathophysiology yet to be fully understood. There is currently no effective treatment available to either slow or terminate it. However, recent advances in ALS genomics have linked genes to phenotypes, encouraging the creation of novel therapeutic approaches and giving researchers more tools to create efficient animal models. Genetically engineered rodent models replicating ALS disease pathology have a high predictive value for translational research. This review addresses the history of the evolution of gene editing tools, the most recent ALS disease models, and the application of CRISPR/Cas9 against ALS disease.
Collapse
Affiliation(s)
- Yajun Shi
- Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Center for Medical Genetics and Prenatal Diagnosis, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yan Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Likui Lu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qinqin Gao
- Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Center for Medical Genetics and Prenatal Diagnosis, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Dongyi Yu
- Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Center for Medical Genetics and Prenatal Diagnosis, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Miao Sun
- Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Center for Medical Genetics and Prenatal Diagnosis, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
13
|
Lins J, Brock TJ, Hopkins CE, Hart AC. Generation of a C. elegans tdp-1 null allele and humanized TARDBP containing human disease-variants. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000693. [PMID: 37351305 PMCID: PMC10282831 DOI: 10.17912/micropub.biology.000693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/24/2023]
Abstract
Clinical variants of TARDBP are associated with frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS) and other degenerative diseases. The predicted C. elegans ortholog of TARDBP is encoded by tdp-1 , but functional orthology has not been demonstrated in vivo. We undertook CRISPR/Cas9-based genome editing of the tdp-1 locus to create a complete loss of function allele; all tdp-1 exons and introns were deleted, creating tdp-1(tgx58) , which resulted in neurodegeneration after oxidative stress. Next, we undertook CRISPR-based genome editing to replace tdp-1 exons with human TARDBP coding sequences, creating humanized ( hTARDBP ) C. elegans expressing TDP-43 . Based on the efficiency of this genome editing, we suggest that iterative genome editing of the tdp-1 target locus using linked coCRISPR markers, like dpy-10 , would be a more efficient strategy for sequential assembly of the large engineered transgenes. hTARDBP decreased the neurodegeneration defect of tdp-1(tgx58) , demonstrating functional cross-species orthology. To develop C. elegans models of FTD and ALS, we inserted five different patient TARDBP variants in the C. elegans hTARDBP locus. Only one clinical variant increased stress-induced neurodegeneration; other variants caused inconsistent or negligible defects under these conditions. Combined, this work yielded an unambiguous null allele for tdp-1 , a validated, humanized hTARDBP, and multiple ALS/FTD patient-associated variant models that can be used for future studies.
Collapse
Affiliation(s)
- Jeremy Lins
- Department of Neuroscience, Brown University, Providence, RI 02912
| | | | | | - Anne C Hart
- Department of Neuroscience, Brown University, Providence, RI 02912
| |
Collapse
|
14
|
Kassouf T, Shrivastava R, Meszka I, Bailly A, Polanowska J, Trauchessec H, Mandrioli J, Carra S, Xirodimas DP. Targeting the NEDP1 enzyme to ameliorate ALS phenotypes through stress granule disassembly. SCIENCE ADVANCES 2023; 9:eabq7585. [PMID: 37000881 PMCID: PMC10065448 DOI: 10.1126/sciadv.abq7585] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 02/27/2023] [Indexed: 06/19/2023]
Abstract
The elimination of aberrant inclusions is regarded as a therapeutic approach in neurodegeneration. In amyotrophic lateral sclerosis (ALS), mutations in proteins found within cytoplasmic condensates called stress granules (SGs) are linked to the formation of pathological SGs, aberrant protein inclusions, and neuronal toxicity. We found that inhibition of NEDP1, the enzyme that processes/deconjugates the ubiquitin-like molecule NEDD8, promotes the disassembly of physiological and pathological SGs. Reduction in poly(ADP-ribose) polymerase1 activity through hyper-NEDDylation is a key mechanism for the observed phenotype. These effects are related to improved cell survival in human cells, and in C. elegans, nedp1 deletion ameliorates ALS phenotypes related to animal motility. Our studies reveal NEDP1 as potential therapeutic target for ALS, correlated to the disassembly of pathological SGs.
Collapse
Affiliation(s)
| | | | - Igor Meszka
- CRBM, Univ. Montpellier, CNRS, Montpellier, France
| | | | | | | | - Jessica Mandrioli
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia,G. Campi 287, 41125 Modena, Italy
| | - Serena Carra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia,G. Campi 287, 41125 Modena, Italy
| | | |
Collapse
|
15
|
Caenorhabditis elegans as a Model System to Study Human Neurodegenerative Disorders. Biomolecules 2023; 13:biom13030478. [PMID: 36979413 PMCID: PMC10046667 DOI: 10.3390/biom13030478] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/18/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
In recent years, advances in science and technology have improved our quality of life, enabling us to tackle diseases and increase human life expectancy. However, longevity is accompanied by an accretion in the frequency of age-related neurodegenerative diseases, creating a growing burden, with pervasive social impact for human societies. The cost of managing such chronic disorders and the lack of effective treatments highlight the need to decipher their molecular and genetic underpinnings, in order to discover new therapeutic targets. In this effort, the nematode Caenorhabditis elegans serves as a powerful tool to recapitulate several disease-related phenotypes and provides a highly malleable genetic model that allows the implementation of multidisciplinary approaches, in addition to large-scale genetic and pharmacological screens. Its anatomical transparency allows the use of co-expressed fluorescent proteins to track the progress of neurodegeneration. Moreover, the functional conservation of neuronal processes, along with the high homology between nematode and human genomes, render C. elegans extremely suitable for the study of human neurodegenerative disorders. This review describes nematode models used to study neurodegeneration and underscores their contribution in the effort to dissect the molecular basis of human diseases and identify novel gene targets with therapeutic potential.
Collapse
|
16
|
Kumar R, Malik Z, Singh M, Rachana R, Mani S, Ponnusamy K, Haider S. Amyotrophic Lateral Sclerosis Risk Genes and Suppressor. Curr Gene Ther 2023; 23:148-162. [PMID: 36366843 DOI: 10.2174/1566523223666221108113330] [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: 04/11/2022] [Revised: 08/24/2022] [Accepted: 09/01/2022] [Indexed: 11/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that leads to death by progressive paralysis and respiratory failure within 2-4 years of onset. About 90-95% of ALS cases are sporadic (sALS), and 5-10% are inherited through family (fALS). Though the mechanisms of the disease are still poorly understood, so far, approximately 40 genes have been reported as ALS causative genes. The mutations in some crucial genes, like SOD1, C9ORF72, FUS, and TDP-43, are majorly associated with ALS, resulting in ROS-associated oxidative stress, excitotoxicity, protein aggregation, altered RNA processing, axonal and vesicular trafficking dysregulation, and mitochondrial dysfunction. Recent studies show that dysfunctional cellular pathways get restored as a result of the repair of a single pathway in ALS. In this review article, our aim is to identify putative targets for therapeutic development and the importance of a single suppressor to reduce multiple symptoms by focusing on important mutations and the phenotypic suppressors of dysfunctional cellular pathways in crucial genes as reported by other studies.
Collapse
Affiliation(s)
- Rupesh Kumar
- Department of Biotechnology, Jaypee Institute of Information Technology, Sec-62, Noida, Uttar Pradesh, India
| | - Zubbair Malik
- School of Computational and Integrative Science, Jawaharlal Nehru University, New Delhi-110067, India
| | - Manisha Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Sec-62, Noida, Uttar Pradesh, India
| | - R Rachana
- Department of Biotechnology, Jaypee Institute of Information Technology, Sec-62, Noida, Uttar Pradesh, India
| | - Shalini Mani
- Department of Biotechnology, Jaypee Institute of Information Technology, Sec-62, Noida, Uttar Pradesh, India
| | | | - Shazia Haider
- Department of Biotechnology, Jaypee Institute of Information Technology, Sec-62, Noida, Uttar Pradesh, India
| |
Collapse
|
17
|
Pasko VI, Churkina AS, Shakhov AS, Kotlobay AA, Alieva IB. Modeling of Neurodegenerative Diseases: 'Step by Step' and 'Network' Organization of the Complexes of Model Systems. Int J Mol Sci 2022; 24:ijms24010604. [PMID: 36614047 PMCID: PMC9820769 DOI: 10.3390/ijms24010604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/17/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022] Open
Abstract
Neurodegenerative diseases have acquired the status of one of the leading causes of death in developed countries, which requires creating new model systems capable of accurately reproducing the mechanisms underlying these pathologies. Here we analyzed modern model systems and their contribution to the solution of unexplored manifestations of neuropathological processes. Each model has unique properties that make it the optimal tool for modeling certain aspects of neurodegenerative disorders. We concluded that to optimize research, it is necessary to combine models into complexes that include organisms and artificial systems of different organizational levels. Such complexes can be organized in two ways. The first method can be described as "step by step", where each model for studying a certain characteristic is a separate step that allows using the information obtained in the modeling process for the gradual study of increasingly complex processes in subsequent models. The second way is a 'network' approach. Studies are carried out with several types of models simultaneously, and experiments with each specific type are adjusted in conformity with the data obtained from other models. In our opinion, the 'network' approach to combining individual model systems seems more promising for fundamental biology as well as diagnostics and therapy.
Collapse
Affiliation(s)
| | - Aleksandra Sergeevna Churkina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 1–73, Leninskye Gory, 119992 Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1–40, Leninskye Gory, 119992 Moscow, Russia
| | - Anton Sergeevich Shakhov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1–40, Leninskye Gory, 119992 Moscow, Russia
| | - Anatoly Alexeevich Kotlobay
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
| | - Irina Borisovna Alieva
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1–40, Leninskye Gory, 119992 Moscow, Russia
- Correspondence:
| |
Collapse
|
18
|
Chauhan P, Wadhwa K, Singh G. Caenorhabditis elegans as a model system to evaluate neuroprotective potential of nano formulations. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.1018754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The impact of neurodegenerative illnesses on society is significant, but the mechanisms leading to neuronal malfunction and death in these conditions remain largely unknown despite identifying essential disease genes. To pinpoint the mechanisms behind the pathophysiology of neurodegenerative diseases, several researchers have turned to nematode C. elegans instead of using mammals. Since C. elegans is transparent, free-living, and amenable to culture, it has several benefits. As a result, all the neurons in C. elegans can be easily identified, and their connections are understood. Human proteins linked to Neurodegeneration can be made to express in them. It is also possible to analyze how C. elegans orthologs of the genes responsible for human neurodegenerative diseases function. In this article, we focused at some of the most important C. elegans neurodegeneration models that accurately represent many elements of human neurodegenerative illness. It has been observed that studies using the adaptable C. elegans have helped us in better understanding of human diseases. These studies have used it to replicate several aspects of human neurodegeneration. A nanotech approach involves engineering materials or equipments interacting with biological systems at the molecular level to trigger physiological responses by increasing stimulation, responding, and interacting with target sites while minimizing side effects, thus revolutionizing the treatment and diagnosis of neurodegenerative diseases. Nanotechnologies are being used to treat neurological disorders and deliver nanoscale drugs. This review explores the current and future uses of these nanotechnologies as innovative therapeutic modalities in treatment of neurodegenerative diseases using C elegans as an experimental model.
Collapse
|
19
|
Lins J, Hopkins CE, Brock T, Hart AC. The use of CRISPR to generate a whole-gene humanized MAPT and the examination of P301L and G272V clinical variants, along with the creation of deletion null alleles of ptl-1, pgrn-1 and alfa-1 loci. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000615. [PMID: 36204657 PMCID: PMC9530923 DOI: 10.17912/micropub.biology.000615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 01/01/1970] [Accepted: 09/02/2022] [Indexed: 11/06/2022]
Abstract
To study important genes involved in Frontotemporal Dementia ( MAPT , GRN and C9orf72 ), we created deletion alleles in the three orthologous genes ( ptl-1 , pgrn-1 , and alfa-1 ). Simultaneously, we replaced the C. elegans ptl-1 gene with the predicted orthologous human MAPT gene, often called whole-gene humanization, which allows direct assessment of conserved gene function, as well as the opportunity to examine consequences of clinical disease-associated patient variations. Each gene was manipulated using a different selection strategy, including a novel strategy using an unc-18 mutation rescue technique. Clinical MAPT ALS/FTD missense variants G272V and P301L were successfully inserted in hMAPT . Neither ptl-1 loss or clinical variants caused neuronal defects in young adult or aged C. elegans , based on examination of glutamatergic phasmid neurons. Yet, we noted decreased survival to day 9 in the P301L hMAPT strain, compared to control strains. Based on these results, we comment on strategies for humanization, including the importance of confirming C. elegans gene predictions and identifying loss of function defects for each gene before embarking on humanization, and we report the creation of strains and a new gene-editing selection strategy that will be useful for future studies.
Collapse
Affiliation(s)
- Jeremy Lins
- Department of Neuroscience, Brown University, Providence, RI 02912
| | | | | | - Anne C. Hart
- Department of Neuroscience, Brown University, Providence, RI 02912
- Robert J. & Nancy D. Carney Institute for Brain Science, Brown University, Providence RI 02912
| |
Collapse
|
20
|
De Plano LM, Calabrese G, Conoci S, Guglielmino SPP, Oddo S, Caccamo A. Applications of CRISPR-Cas9 in Alzheimer's Disease and Related Disorders. Int J Mol Sci 2022; 23:ijms23158714. [PMID: 35955847 PMCID: PMC9368966 DOI: 10.3390/ijms23158714] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, and Huntington’s disease represent some of the most prevalent neurodegenerative disorders afflicting millions of people worldwide. Unfortunately, there is a lack of efficacious treatments to cure or stop the progression of these disorders. While the causes of such a lack of therapies can be attributed to various reasons, the disappointing results of recent clinical trials suggest the need for novel and innovative approaches. Since its discovery, there has been a growing excitement around the potential for CRISPR-Cas9 mediated gene editing to identify novel mechanistic insights into disease pathogenesis and to mediate accurate gene therapy. To this end, the literature is rich with experiments aimed at generating novel models of these disorders and offering proof-of-concept studies in preclinical animal models validating the great potential and versatility of this gene-editing system. In this review, we provide an overview of how the CRISPR-Cas9 systems have been used in these neurodegenerative disorders.
Collapse
Affiliation(s)
- Laura M. De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy
| | - Giovanna Calabrese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy
| | - Sabrina Conoci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy
| | - Salvatore P. P. Guglielmino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy
| | - Salvatore Oddo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy
| | - Antonella Caccamo
- Department of Drug and Health Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
- Correspondence:
| |
Collapse
|
21
|
Saul N, Dhondt I, Kuokkanen M, Perola M, Verschuuren C, Wouters B, von Chrzanowski H, De Vos WH, Temmerman L, Luyten W, Zečić A, Loier T, Schmitz-Linneweber C, Braeckman BP. Identification of healthspan-promoting genes in Caenorhabditis elegans based on a human GWAS study. Biogerontology 2022; 23:431-452. [PMID: 35748965 PMCID: PMC9388463 DOI: 10.1007/s10522-022-09969-8] [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: 02/14/2022] [Accepted: 05/16/2022] [Indexed: 12/03/2022]
Abstract
To find drivers of healthy ageing, a genome-wide association study (GWAS) was performed in healthy and unhealthy older individuals. Healthy individuals were defined as free from cardiovascular disease, stroke, heart failure, major adverse cardiovascular event, diabetes, dementia, cancer, chronic obstructive pulmonary disease (COPD), asthma, rheumatism, Crohn’s disease, malabsorption or kidney disease. Six single nucleotide polymorphisms (SNPs) with unknown function associated with ten human genes were identified as candidate healthspan markers. Thirteen homologous or closely related genes were selected in the model organism C. elegans for evaluating healthspan after targeted RNAi-mediated knockdown using pathogen resistance, muscle integrity, chemotaxis index and the activity of known longevity and stress response pathways as healthspan reporters. In addition, lifespan was monitored in the RNAi-treated nematodes. RNAi knockdown of yap-1, wwp-1, paxt-1 and several acdh genes resulted in heterogeneous phenotypes regarding muscle integrity, pathogen resistance, chemotactic behaviour, and lifespan. Based on these observations, we hypothesize that their human homologues WWC2, CDKN2AIP and ACADS may play a role in health maintenance in the elderly.
Collapse
Affiliation(s)
- Nadine Saul
- Molecular Genetics Group, Institute of Biology, Humboldt University of Berlin, Berlin, Germany.
| | - Ineke Dhondt
- Laboratory of Aging Physiology and Molecular Evolution, Biology Department, Ghent University, Ghent, Belgium
| | - Mikko Kuokkanen
- Genomics and Biomarkers Unit, Department of Health, National Institute for Health and Welfare, Helsinki, Finland.,Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Markus Perola
- Genomics and Biomarkers Unit, Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Clara Verschuuren
- Laboratory of Aging Physiology and Molecular Evolution, Biology Department, Ghent University, Ghent, Belgium
| | | | - Henrik von Chrzanowski
- Molecular Genetics Group, Institute of Biology, Humboldt University of Berlin, Berlin, Germany.,The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | | | | | - Aleksandra Zečić
- Laboratory of Aging Physiology and Molecular Evolution, Biology Department, Ghent University, Ghent, Belgium
| | - Tim Loier
- Laboratory of Aging Physiology and Molecular Evolution, Biology Department, Ghent University, Ghent, Belgium
| | | | - Bart P Braeckman
- Laboratory of Aging Physiology and Molecular Evolution, Biology Department, Ghent University, Ghent, Belgium
| |
Collapse
|
22
|
Tossing G, Livernoche R, Maios C, Bretonneau C, Labarre A, Parker JA. Genetic and pharmacological PARP inhibition reduces axonal degeneration in C. elegans models of ALS. Hum Mol Genet 2022; 31:3313-3324. [PMID: 35594544 DOI: 10.1093/hmg/ddac116] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/12/2022] Open
Abstract
Axonal degeneration is observed in early stages of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). This degeneration generally precedes apoptosis and therefore may be a promising therapeutic target. An increasing number of genes have been identified to actively regulate axonal degeneration and regeneration, however, only a few potential therapeutic targets have been identified in the context of neurodegenerative diseases. Here we investigate DLK-1, a major axonal regeneration pathway and its contribution to axonal degeneration phenotypes in several C. elegans ALS models. From this pathway, we identified the PAR polymerases (PARP) PARP-1 and PARP-2 as the most consistent modifiers of axonal degeneration in our models of ALS. Genetic and pharmacological inhibition of PARP-1 and PARP-2 reduces axonal degeneration and improves related motor phenotypes.
Collapse
Affiliation(s)
- Gilles Tossing
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Department of Neuroscience, Université de Montréal, Montreal, Canada
| | | | - Claudia Maios
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Constantin Bretonneau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Department of Neuroscience, Université de Montréal, Montreal, Canada
| | - Audrey Labarre
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Department of Neuroscience, Université de Montréal, Montreal, Canada
| | - J Alex Parker
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada.,Department of Neuroscience, Université de Montréal, Montreal, Canada
| |
Collapse
|
23
|
Invited review: Unearthing the mechanisms of age-related neurodegenerative disease using Caenorhabditis elegans. Comp Biochem Physiol A Mol Integr Physiol 2022; 267:111166. [PMID: 35176489 DOI: 10.1016/j.cbpa.2022.111166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
Abstract
As human life expectancy increases, neurodegenerative diseases present a growing public health threat, for which there are currently few effective treatments. There is an urgent need to understand the molecular and genetic underpinnings of these disorders so new therapeutic targets can be identified. Here we present the argument that the simple nematode worm Caenorhabditis elegans is a powerful tool to rapidly study neurodegenerative disorders due to their short lifespan and vast array of genetic tools, which can be combined with characterization of conserved neuronal processes and behavior orthologous to those disrupted in human disease. We review how pre-existing C. elegans models provide insight into human neurological disease as well as an overview of current tools available to study neurodegenerative diseases in the worm, with an emphasis on genetics and behavior. We also discuss open questions that C. elegans may be particularly well suited for in future studies and how worms will be a valuable preclinical model to better understand these devastating neurological disorders.
Collapse
|
24
|
Martin RM, Bereman MS, Marsden KC. The Cyanotoxin 2,4-DAB Reduces Viability and Causes Behavioral and Molecular Dysfunctions Associated with Neurodegeneration in Larval Zebrafish. Neurotox Res 2022; 40:347-364. [PMID: 35029765 PMCID: PMC9035002 DOI: 10.1007/s12640-021-00465-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/30/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022]
Abstract
Exposure to cyanotoxins has been linked to neurodegenerative diseases, including amyotrophic lateral sclerosis, Alzheimer's, and Parkinson's disease. While the cyanotoxin β-methylamino-L-alanine (BMAA) has received much attention, cyanobacteria produce many cyanotoxic compounds, several of which have been detected in nature alongside BMAA, including 2,4-diaminobutyric acid (2,4-DAB) and N-(2-aminoethyl)glycine (AEG). Thus, the question of whether 2,4-DAB and AEG also cause neurotoxic effects in vivo is of great interest, as is the question of whether they interact to enhance toxicity. Here, we evaluate the toxic and neurotoxic effects of these cyanotoxins alone or in combination by measuring zebrafish larval viability and behavior after exposure. 2,4-DAB was the most potent cyanotoxin as it decreased larval viability by approximately 50% at 6 days post fertilization, while BMAA and AEG decreased viability by just 16% and 8%, respectively. Although we only observed minor neurotoxic effects on spontaneous locomotion, BMAA and AEG enhanced acoustic startle sensitivity, and they interacted in an additive manner to exert their effects. 2,4-DAB; however, only modulated startle kinematics, an indication of motor dysfunction. To investigate the mechanisms of 2,4-DAB's effects, we analyzed the protein profile of larval zebrafish exposed to 500 µM 2,4-DAB at two time points and identified molecular signatures consistent with neurodegeneration, including disruption of metabolic pathways and downregulation of the ALS-associated genes SOD1 and UBQLN4. Together, our data demonstrate that BMAA and its isomers AEG and 2,4-DAB cause neurotoxic effects in vivo, with 2,4-DAB as the most potent of the three in the zebrafish model.
Collapse
Affiliation(s)
- Rubia M Martin
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Michael S Bereman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Kurt C Marsden
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
25
|
Barlow IL, Feriani L, Minga E, McDermott-Rouse A, O'Brien TJ, Liu Z, Hofbauer M, Stowers JR, Andersen EC, Ding SS, Brown AEX. Megapixel camera arrays enable high-resolution animal tracking in multiwell plates. Commun Biol 2022; 5:253. [PMID: 35322206 PMCID: PMC8943053 DOI: 10.1038/s42003-022-03206-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 03/01/2022] [Indexed: 01/13/2023] Open
Abstract
Tracking small laboratory animals such as flies, fish, and worms is used for phenotyping in neuroscience, genetics, disease modelling, and drug discovery. An imaging system with sufficient throughput and spatiotemporal resolution would be capable of imaging a large number of animals, estimating their pose, and quantifying detailed behavioural differences at a scale where hundreds of treatments could be tested simultaneously. Here we report an array of six 12-megapixel cameras that record all the wells of a 96-well plate with sufficient resolution to estimate the pose of C. elegans worms and to extract high-dimensional phenotypic fingerprints. We use the system to study behavioural variability across wild isolates, the sensitisation of worms to repeated blue light stimulation, the phenotypes of worm disease models, and worms' behavioural responses to drug treatment. Because the system is compatible with standard multiwell plates, it makes computational ethological approaches accessible in existing high-throughput pipelines.
Collapse
Affiliation(s)
- Ida L Barlow
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Luigi Feriani
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Eleni Minga
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Adam McDermott-Rouse
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Thomas James O'Brien
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - Ziwei Liu
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | | | | - Erik C Andersen
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Siyu Serena Ding
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, London, UK
- Max Planck Institute of Animal Behavior, Konstanz, Germany
| | - André E X Brown
- Institute of Clinical Sciences, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, London, UK.
| |
Collapse
|
26
|
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
|
27
|
Yu CY, Chang HC. Glutamate signaling mediates C. elegans behavioral plasticity to pathogens. iScience 2022; 25:103919. [PMID: 35252815 PMCID: PMC8889136 DOI: 10.1016/j.isci.2022.103919] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Chun-Ying Yu
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, 62102, Taiwan
| | - Howard C. Chang
- Department of Cell Biology and Neuroscience, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
- Corresponding author
| |
Collapse
|
28
|
Xu H, Jia C, Cheng C, Wu H, Cai H, Le W. Activation of autophagy attenuates motor deficits and extends lifespan in a C. elegans model of ALS. Free Radic Biol Med 2022; 181:52-61. [PMID: 35114355 PMCID: PMC8996503 DOI: 10.1016/j.freeradbiomed.2022.01.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/18/2022] [Accepted: 01/30/2022] [Indexed: 10/19/2022]
Abstract
Mutations in Cu/Zn-superoxide dismutase 1 (SOD1) are linked to amyotrophic lateral sclerosis (ALS). Using a line of ALS-related mutant human SOD1 (hSOD1) transgenic Caenorhabditis elegans, we determined the effects of metformin on the progression of ALS-like pathological abnormalities. We found that metformin significantly extended the lifespan, improved motor performance, and enhanced antioxidant activity of mutant worms. We further showed that metformin enhanced expression of lgg-1, daf-16, skn-1 and other genes known to regulate autophagy, longevity and oxidative stress in hSOD1 transgenic worms. Accordingly, overexpression of lgg-1 or daf-16 attenuated the aging and pathological abnormalities of mutant human SOD1 worms, while genetic deletion of lgg-1 or daf-16 abolished the beneficial effects of metformin. Collectively, we demonstrate that metformin protects against mutant SOD1-induced cytotoxicity in part through enhancement of autophagy and extends lifespan through daf-16 pathway. Our findings suggest that metformin could be further explored as a potential therapeutic agent in treating ALS.
Collapse
Affiliation(s)
- Hui Xu
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Congcong Jia
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Cheng Cheng
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Haifeng Wu
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging (NIA), National Institutes of Health, Bethesda, MD, United States
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China; Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People's Hospital, Chengdu, 610072, China.
| |
Collapse
|
29
|
Kaur S, Sang Y, Aballay A. Myotubularin-related protein protects against neuronal degeneration mediated by oxidative stress or infection. J Biol Chem 2022; 298:101614. [PMID: 35101447 PMCID: PMC8889260 DOI: 10.1016/j.jbc.2022.101614] [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: 08/04/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 11/04/2022] Open
Abstract
Microbial infections have been linked to the onset and severity of neurodegenerative diseases such as amyotrophic lateral sclerosis, multiple sclerosis, Alzheimer's disease, but the underlying mechanisms remain largely unknown. Here, we used a genetic screen for genes involved in protection from infection-associated neurodegeneration and identified the gene mtm-10. We then validated the role of the encoded myotubularin-related protein, MTM-10, in protecting the dendrites of Caenorhabditis elegans from degeneration mediated by oxidative stress or Pseudomonas aeruginosa infection. Further experiments indicated that mtm-10 is expressed in the AWC neurons of C. elegans, where it functions in a cell-autonomous manner to protect the dendrite degeneration caused by pathogen infection. We also confirm that the changes observed in the dendrites of the animals were not because of premature death or overall sickness. Finally, our studies indicated that mtm-10 functions in AWC neurons to preserve chemosensation after pathogen infection. These results reveal an essential role for myotubularin-related protein 10 in the protection of dendrite morphology and function against the deleterious effects of oxidative stress or infection.
Collapse
Affiliation(s)
- Supender Kaur
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Yu Sang
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Alejandro Aballay
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
30
|
Baskoylu SN, Chapkis N, Unsal B, Lins J, Schuch K, Simon J, Hart AC. Disrupted autophagy and neuronal dysfunction in C. elegans knockin models of FUS amyotrophic lateral sclerosis. Cell Rep 2022; 38:110195. [PMID: 35081350 DOI: 10.1016/j.celrep.2021.110195] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/01/2021] [Accepted: 12/10/2021] [Indexed: 11/18/2022] Open
Abstract
How mutations in FUS lead to neuronal dysfunction in amyotrophic lateral sclerosis (ALS) patients remains unclear. To examine mechanisms underlying ALS FUS dysfunction, we generate C. elegans knockin models using CRISPR-Cas9-mediated genome editing, creating R524S and P525L ALS FUS models. Although FUS inclusions are not detected, ALS FUS animals show defective neuromuscular function and locomotion under stress. Unlike animals lacking the endogenous FUS ortholog, ALS FUS animals have impaired neuronal autophagy and increased SQST-1 accumulation in motor neurons. Loss of sqst-1, the C. elegans ortholog for ALS-linked, autophagy adaptor protein SQSTM1/p62, suppresses both neuromuscular and stress-induced locomotion defects in ALS FUS animals, but does not suppress neuronal autophagy defects. Therefore, autophagy dysfunction is upstream of, and not dependent on, SQSTM1 function in ALS FUS pathogenesis. Combined, our findings demonstrate that autophagy dysfunction likely contributes to protein homeostasis and neuromuscular defects in ALS FUS knockin animals.
Collapse
Affiliation(s)
- Saba N Baskoylu
- Department of Neuroscience and the Robert J. & Nancy D. Carney Institute for Brain Sciences, Brown University, Providence, RI 02906, USA
| | - Natalie Chapkis
- Department of Neuroscience and the Robert J. & Nancy D. Carney Institute for Brain Sciences, Brown University, Providence, RI 02906, USA
| | - Burak Unsal
- Department of Neuroscience and the Robert J. & Nancy D. Carney Institute for Brain Sciences, Brown University, Providence, RI 02906, USA; Department of Molecular Biology and Genetics, Bogazici University, Istanbul 34342, Turkey
| | - Jeremy Lins
- Department of Neuroscience and the Robert J. & Nancy D. Carney Institute for Brain Sciences, Brown University, Providence, RI 02906, USA
| | - Kelsey Schuch
- Department of Molecular Biology, Cellular Biology & Biochemistry, Brown University, Providence, RI 02906, USA
| | - Jonah Simon
- Department of Neuroscience and the Robert J. & Nancy D. Carney Institute for Brain Sciences, Brown University, Providence, RI 02906, USA
| | - Anne C Hart
- Department of Neuroscience and the Robert J. & Nancy D. Carney Institute for Brain Sciences, Brown University, Providence, RI 02906, USA.
| |
Collapse
|
31
|
Dong L, Liu L, Li Y, Li W, Zhou L, Xia Q. E3 ligase Smurf1 protects against misfolded SOD1 in neuronal cells by promoting its K63 ubiquitylation and aggresome formation. Hum Mol Genet 2022; 31:2035-2048. [PMID: 35022748 DOI: 10.1093/hmg/ddac008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/15/2021] [Accepted: 01/07/2022] [Indexed: 11/15/2022] Open
Abstract
K63-linked polyubiquitination of the neurodegenerative disease-associated misfolded protein copper-zinc superoxide dismutase (SOD1) is associated with the formation of inclusion bodies. Highly expressed E3 ligase Smurf1 promotes cellular homostasis through the enhanced capability of aggregate degradation. However, it is not well explored the role of Smurf1 in the dynamics of SOD1 aggresomes. In this study, we report that Smurf1 promotes the recruitment of SOD1 to form aggresomes. Mechanistically, Smurf1 interacts with mutant SOD1 to promote aggresome formation by modification of its K63-linked polyubiquitination. Moreover, overexpressed Smurf1 enhances mutant SOD1 aggresome formation and autophagic degradation to prevent cell death. Thus, our data suggest that Smurf1 plays an important role in attenuating protein misfolding-induced cell toxicity by both driving the sequestration of misfolded SOD1 into aggresomes and autophagic degradation.
Collapse
Affiliation(s)
- Lei Dong
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Liqun Liu
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yang Li
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Wenxuan Li
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Liying Zhou
- Beijing Tide Pharmaceutical CO., LTD, Beijing, 100000, China
| | - Qin Xia
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
32
|
Berdyński M, Miszta P, Safranow K, Andersen PM, Morita M, Filipek S, Żekanowski C, Kuźma-Kozakiewicz M. SOD1 mutations associated with amyotrophic lateral sclerosis analysis of variant severity. Sci Rep 2022; 12:103. [PMID: 34996976 PMCID: PMC8742055 DOI: 10.1038/s41598-021-03891-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023] Open
Abstract
Mutations in superoxide dismutase 1 gene (SOD1) are linked to amyotrophic lateral sclerosis (ALS), a neurodegenerative disorder predominantly affecting upper and lower motor neurons. The clinical phenotype of ALS shows inter- and intrafamilial heterogeneity. The aim of the study was to analyze the relations between individual SOD1 mutations and the clinical presentation using in silico methods to assess the SOD1 mutations severity. We identified SOD1 causative variants in a group of 915 prospectively tested consecutive Polish ALS patients from a neuromuscular clinical center, performed molecular modeling of mutated SOD1 proteins and in silico analysis of mutation impact on clinical phenotype and survival analysis of associations between mutations and hazard of clinical end-points. Fifteen SOD1 mutations were identified in 21.1% familial and 2.3% sporadic ALS cases. Their effects on SOD1 protein structure and functioning inferred from molecular modeling and in silico analyses correlate well with the clinical data. Molecular modeling results support the hypothesis that folding intermediates rather than mature SOD1 protein give rise to the source of cytotoxic conformations in ALS. Significant associations between type of mutation and clinical end-points were found.
Collapse
Affiliation(s)
- Mariusz Berdyński
- Laboratory of Neurogenetics, Department of Neurodegenerative Disorders, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland. .,Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden.
| | - Przemysław Miszta
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 72 Powstańców Wlkp. Str., 70-111, Szczecin, Poland
| | - Peter M Andersen
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| | - Mitsuya Morita
- Division of Neurology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Sławomir Filipek
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Cezary Żekanowski
- Laboratory of Neurogenetics, Department of Neurodegenerative Disorders, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Magdalena Kuźma-Kozakiewicz
- Department of Neurology, Medical University of Warsaw, Warsaw, Poland. .,Neurodegenerative Diseases Research Group, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
33
|
ZHANG X, GUO D, ZHANG X, ZHANG W, WANG T, ZHANG L. Three-N-butyphthalide alleviates early brain injury caused via subarachnoid hemorrhage via activating the LKB-1/ (AMP-activated protein kinase) pathway. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.86321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
34
|
Hughes KJ, Vidal-Gadea AG. Methods for Modulating and Measuring Neuromuscular Exertion in C. elegans. Methods Mol Biol 2022; 2468:339-356. [PMID: 35320575 DOI: 10.1007/978-1-0716-2181-3_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The nematode C. elegans has been used widely to study the genetic and cellular basis of behavior. Yet the laboratory conditions under which it is typically studied offer only a narrow glimpse into the richness of natural behaviors this remarkable animal evolved over 500 million years of evolution. For example, burrowing behavior naturally occurs in the wild, but it remains understudied. Our group studies burrowing in an attempt to expand our understanding of the natural behavioral repertoire of C. elegans. Aside from being an interesting and tractable behavior, burrowing is experimentally useful and permits the titration of the muscular output exerted by C. elegans. Here we describe several burrowing assays that allow the modulation of muscular exertion. We used these to study both adaptive and pathological muscular processes such as muscle hypertrophy and dystrophy, respectively. We believe these assays will be of use for researchers studying the production of locomotion under normal and disease-challenged conditions.
Collapse
Affiliation(s)
- Kiley J Hughes
- School of Biological Sciences, Illinois State University, Normal, IL, USA
| | | |
Collapse
|
35
|
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
|
36
|
Kounakis K, Tavernarakis N. Assessment of Neuronal Cell Death in Caenorhabditis elegans. Methods Mol Biol 2022; 2515:309-317. [PMID: 35776360 DOI: 10.1007/978-1-0716-2409-8_19] [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] [Indexed: 06/15/2023]
Abstract
The nematode Caenorhabditis elegans is a powerful experimental platform for cell biology studies. The molecular mechanisms that mediate cell death and neurodegeneration have been characterized extensively in the nematode. In addition, the availability of a wide arsenal of genetic and molecular tools and methodologies renders C. elegans an organism of choice for modeling human neurodegenerative diseases. Indeed, neuronal necrosis can readily be observed and examined in vivo, in the worm. In this chapter, we describe the two main approaches that are routinely used for monitoring and quantifying neuronal cell death in C. elegans. The first is based on direct visualization of dying cells via Nomarski differential interference contrast (DiC) microscopy, and the second on the assessment of neuronal survival by fluorescence microscopy.
Collapse
Affiliation(s)
- Konstantinos Kounakis
- Department of Basic Sciences, Faculty of Medicine, University of Crete, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas , Crete, Greece
| | - Nektarios Tavernarakis
- Department of Basic Sciences, Faculty of Medicine, University of Crete, Crete, Greece.
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas , Crete, Greece.
| |
Collapse
|
37
|
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
|
38
|
Narayanan RK, Brewer MH, Perez-Siles G, Ellis M, Ly C, Burgess A, Neumann B, Nicholson GA, Vucic S, Kennerson ML. Charcot-Marie-tooth disease causing mutation (p.R158H) in pyruvate dehydrogenase kinase 3 (PDK3) affects synaptic transmission, ATP production and causes neurodegeneration in a CMTX6 C. elegans model. Hum Mol Genet 2021; 31:133-145. [PMID: 34387338 PMCID: PMC8682796 DOI: 10.1093/hmg/ddab228] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/05/2022] Open
Abstract
Charcot–Marie-Tooth (CMT) is a commonly inherited, non-fatal neurodegenerative disorder that affects sensory and motor neurons in patients. More than 90 genes are known to cause axonal and demyelinating forms of CMT. The p.R158H mutation in the pyruvate dehydrogenase kinase 3 (PDK3) gene is the genetic cause for an X linked form of axonal CMT (CMTX6). In vitro studies using patient fibroblasts and iPSC-derived motor neurons have shown that this mutation causes deficits in energy metabolism and mitochondrial function. Animal models that recapitulate pathogenic in vivo events in patients are crucial for investigating mechanisms of axonal degeneration and developing therapies for CMT. We have developed a C. elegans model of CMTX6 by knocking-in the p.R158H mutation in pdhk-2, the ortholog of PDK3. In addition, we have developed animal models overexpressing the wild type and mutant form of human PDK3 specifically in the GABAergic motor neurons of C. elegans. CMTX6 mutants generated in this study exhibit synaptic transmission deficits, locomotion defects and show signs of progressive neurodegeneration. Furthermore, the CMTX6 in vivo models display energy deficits that recapitulate the phenotype observed in patient fibroblasts and iPSC-derived motor neurons. Our CMTX6 animals represent the first in vivo model for this form of CMT and have provided novel insights into the cellular function and metabolic pathways perturbed by the p.R158H mutation, all the while closely replicating the clinical presentation observed in CMTX6 patients.
Collapse
Affiliation(s)
- Ramesh K Narayanan
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Megan H Brewer
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia
| | - Gonzalo Perez-Siles
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Melina Ellis
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia
| | - Carolyn Ly
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia
| | - Andrew Burgess
- Cell Division Laboratory, ANZAC Research Institute, Sydney, NSW, Australia
| | - Brent Neumann
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Garth A Nicholson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Molecular Medicine Laboratory, Concord General Repatriation Hospital, Sydney, NSW, Australia
| | - Steve Vucic
- Concord Clinical School, University of Sydney, Sydney, NSW, Australia
| | - Marina L Kennerson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Molecular Medicine Laboratory, Concord General Repatriation Hospital, Sydney, NSW, Australia
| |
Collapse
|
39
|
Santur K, Reinartz E, Lien Y, Tusche M, Altendorf T, Sevenich M, Tamgüney G, Mohrlüder J, Willbold D. Ligand-Induced Stabilization of the Native Human Superoxide Dismutase 1. ACS Chem Neurosci 2021; 12:2520-2528. [PMID: 34138531 DOI: 10.1021/acschemneuro.1c00253] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A common characteristic of familial (fALS) and sporadic amyotrophic lateral sclerosis (sALS) is the accumulation of aberrant proteinaceous species in the motor neurons and spinal cord of ALS patients-including aggregates of the human superoxide dismutase 1 (hSOD1). hSOD1 is an enzyme that occurs as a stable dimeric protein with several post-translational modifications such as the formation of an intramolecular disulfide bond and the acquisition of metal cofactors that are essential for enzyme activity and further contribute to protein stability. Some mutations and/or destabilizing factors promote hSOD1 misfolding, causing neuronal death. Aggregates containing misfolded wild-type hSOD1 have been found in the spinal cords of sALS as well as in non-hSOD1 fALS patients, leading to the hypothesis that hSOD1 misfolding is a common part of the ALS pathomechanism. Therefore, stabilizing the native conformation of SOD1 may be a promising approach to prevent the formation of toxic hSOD1 species and thus ALS pathogenesis. Here, we present the 16-mer peptide S1VL-21 that interferes with hSOD1 aggregation. S1VL-21 was identified by phage display selection with the native conformation of hSOD1 as a target. Several methods such as microscale thermophoresis (MST) measurements, aggregation assays, and cell viability assays revealed that S1VL-21 has a micromolar binding affinity to native hSOD1 and considerably reduces the formation of hSOD1 aggregates. This present work therefore provides the first important data on a potential lead compound for hSOD1-related drug development for ALS therapy.
Collapse
Affiliation(s)
- Karoline Santur
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Elke Reinartz
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Yi Lien
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Markus Tusche
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Tim Altendorf
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Marc Sevenich
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Gültekin Tamgüney
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Jeannine Mohrlüder
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Dieter Willbold
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
40
|
Braems E, Tziortzouda P, Van Den Bosch L. Exploring the alternative: Fish, flies and worms as preclinical models for ALS. Neurosci Lett 2021; 759:136041. [PMID: 34118308 DOI: 10.1016/j.neulet.2021.136041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 04/15/2021] [Accepted: 06/01/2021] [Indexed: 12/22/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disorder characterized by the loss of upper and lower motor neurons. In general, patients succumb to respiratory insufficiency due to respiratory muscle weakness. Despite many promising therapeutic strategies primarily identified in rodent models, patient trials remain rather unsuccessful. There is a clear need for alternative approaches, which could provide directions towards the justified use of rodents and which increase the likelihood to identify new promising clinical candidates. In the last decades, the use of fast genetic approaches and the development of high-throughput screening platforms in the nematode Caenorhabditis elegans, in the fruit fly (Drosophila melanogaster) and in zebrafish (Danio rerio) have contributed to new insights into ALS pathomechanisms, disease modifiers and therapeutic targets. In this mini-review, we provide an overview of these alternative small animal studies, modeling the most common ALS genes and discuss the most recent preclinical discoveries. We conclude that small animal models will not replace rodent models, yet they clearly represent an important asset for preclinical studies.
Collapse
Affiliation(s)
- Elke Braems
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Paraskevi Tziortzouda
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.
| |
Collapse
|
41
|
Liguori F, Amadio S, Volonté C. Where and Why Modeling Amyotrophic Lateral Sclerosis. Int J Mol Sci 2021; 22:ijms22083977. [PMID: 33921446 PMCID: PMC8070525 DOI: 10.3390/ijms22083977] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023] Open
Abstract
Over the years, researchers have leveraged a host of different in vivo models in order to dissect amyotrophic lateral sclerosis (ALS), a neurodegenerative/neuroinflammatory disease that is heterogeneous in its clinical presentation and is multigenic, multifactorial and non-cell autonomous. These models include both vertebrates and invertebrates such as yeast, worms, flies, zebrafish, mice, rats, guinea pigs, dogs and, more recently, non-human primates. Despite their obvious differences and peculiarities, only the concurrent and comparative analysis of these various systems will allow the untangling of the causes and mechanisms of ALS for finally obtaining new efficacious therapeutics. However, harnessing these powerful organisms poses numerous challenges. In this context, we present here an updated and comprehensive review of how eukaryotic unicellular and multicellular organisms that reproduce a few of the main clinical features of the disease have helped in ALS research to dissect the pathological pathways of the disease insurgence and progression. We describe common features as well as discrepancies among these models, highlighting new insights and emerging roles for experimental organisms in ALS.
Collapse
Affiliation(s)
- Francesco Liguori
- Preclinical Neuroscience, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (F.L.); (S.A.)
| | - Susanna Amadio
- Preclinical Neuroscience, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (F.L.); (S.A.)
| | - Cinzia Volonté
- Preclinical Neuroscience, IRCCS Santa Lucia Foundation, 00143 Rome, Italy; (F.L.); (S.A.)
- Institute for Systems Analysis and Computer Science “A. Ruberti”, National Research Council (IASI—CNR), 00185 Rome, Italy
- Correspondence: ; Tel.: +39-06-50170-3084
| |
Collapse
|
42
|
Giunti S, Andersen N, Rayes D, De Rosa MJ. Drug discovery: Insights from the invertebrate Caenorhabditis elegans. Pharmacol Res Perspect 2021; 9:e00721. [PMID: 33641258 PMCID: PMC7916527 DOI: 10.1002/prp2.721] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/06/2021] [Indexed: 12/18/2022] Open
Abstract
Therapeutic drug development is a long, expensive, and complex process that usually takes 12-15 years. In the early phases of drug discovery, in particular, there is a growing need for animal models that ensure the reduction in both cost and time. Caenorhabditis elegans has been traditionally used to address fundamental aspects of key biological processes, such as apoptosis, aging, and gene expression regulation. During the last decade, with the advent of large-scale platforms for screenings, this invertebrate has also emerged as an essential tool in the pharmaceutical research industry to identify novel drugs and drug targets. In this review, we discuss the reasons why C. elegans has been positioned as an outstanding cost-effective option for drug discovery, highlighting both the advantages and drawbacks of this model. Particular attention is paid to the suitability of this nematode in large-scale genetic and pharmacological screenings. High-throughput screenings in C. elegans have indeed contributed to the breakthrough of a wide variety of candidate compounds involved in extensive fields including neurodegeneration, pathogen infections and metabolic disorders. The versatility of this nematode, which enables its instrumentation as a model of human diseases, is another attribute also herein underscored. As illustrative examples, we discuss the utility of C. elegans models of both human neurodegenerative diseases and parasitic nematodes in the drug discovery industry. Summing up, this review aims to demonstrate the impact of C. elegans models on the drug discovery pipeline.
Collapse
Affiliation(s)
- Sebastián Giunti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - Natalia Andersen
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - Diego Rayes
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - María José De Rosa
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| |
Collapse
|
43
|
Quelle-Regaldie A, Sobrido-Cameán D, Barreiro-Iglesias A, Sobrido MJ, Sánchez L. Zebrafish Models of Autosomal Dominant Ataxias. Cells 2021; 10:421. [PMID: 33671313 PMCID: PMC7922657 DOI: 10.3390/cells10020421] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Hereditary dominant ataxias are a heterogeneous group of neurodegenerative conditions causing cerebellar dysfunction and characterized by progressive motor incoordination. Despite many efforts put into the study of these diseases, there are no effective treatments yet. Zebrafish models are widely used to characterize neuronal disorders due to its conserved vertebrate genetics that easily support genetic edition and their optic transparency that allows observing the intact CNS and its connections. In addition, its small size and external fertilization help to develop high throughput assays of candidate drugs. Here, we discuss the contributions of zebrafish models to the study of dominant ataxias defining phenotypes, genetic function, behavior and possible treatments. In addition, we review the zebrafish models created for X-linked repeat expansion diseases X-fragile/fragile-X tremor ataxia. Most of the models reviewed here presented neuronal damage and locomotor deficits. However, there is a generalized lack of zebrafish adult heterozygous models and there are no knock-in zebrafish models available for these diseases. The models created for dominant ataxias helped to elucidate gene function and mechanisms that cause neuronal damage. In the future, the application of new genetic edition techniques would help to develop more accurate zebrafish models of dominant ataxias.
Collapse
Affiliation(s)
- Ana Quelle-Regaldie
- Department of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary Science, Universidade of Santiago de Compostela, 27002 Lugo, Spain; (A.Q.-R.); (L.S.)
| | - Daniel Sobrido-Cameán
- Department of Functional Biology, CIBUS, Faculty of Biology, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain;
| | - Antón Barreiro-Iglesias
- Department of Functional Biology, CIBUS, Faculty of Biology, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain;
| | - María Jesús Sobrido
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servicio Galego de Saúde, 15006 Coruña, Spain;
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary Science, Universidade of Santiago de Compostela, 27002 Lugo, Spain; (A.Q.-R.); (L.S.)
- Preclinical Animal Models Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| |
Collapse
|
44
|
Ryan VH, Perdikari TM, Naik MT, Saueressig CF, Lins J, Dignon GL, Mittal J, Hart AC, Fawzi NL. Tyrosine phosphorylation regulates hnRNPA2 granule protein partitioning and reduces neurodegeneration. EMBO J 2021; 40:e105001. [PMID: 33349959 PMCID: PMC7849316 DOI: 10.15252/embj.2020105001] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 10/14/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
Abstract
mRNA transport in neurons requires formation of transport granules containing many protein components, and subsequent alterations in phosphorylation status can release transcripts for translation. Further, mutations in a structurally disordered domain of the transport granule protein hnRNPA2 increase its aggregation and cause hereditary proteinopathy of neurons, myocytes, and bone. We examine in vitro hnRNPA2 granule component phase separation, partitioning specificity, assembly/disassembly, and the link to neurodegeneration. Transport granule components hnRNPF and ch-TOG interact weakly with hnRNPA2 yet partition specifically into liquid phase droplets with the low complexity domain (LC) of hnRNPA2, but not FUS LC. In vitro hnRNPA2 tyrosine phosphorylation reduces hnRNPA2 phase separation, prevents partitioning of hnRNPF and ch-TOG into hnRNPA2 LC droplets, and decreases aggregation of hnRNPA2 disease variants. The expression of chimeric hnRNPA2 D290V in Caenorhabditis elegans results in stress-induced glutamatergic neurodegeneration; this neurodegeneration is rescued by loss of tdp-1, suggesting gain-of-function toxicity. The expression of Fyn, a tyrosine kinase that phosphorylates hnRNPA2, reduces neurodegeneration associated with chimeric hnRNPA2 D290V. These data suggest a model where phosphorylation alters LC interaction specificity, aggregation, and toxicity.
Collapse
Affiliation(s)
- Veronica H Ryan
- Neuroscience Graduate ProgramBrown UniversityProvidenceRIUSA
| | | | - Mandar T Naik
- Department of Molecular Pharmacology, Physiology, and BiotechnologyBrown UniversityProvidenceRIUSA
| | | | - Jeremy Lins
- Department of NeuroscienceBrown UniversityProvidenceRIUSA
| | - Gregory L Dignon
- Department of Chemical and Biomolecular EngineeringLehigh UniversityBethlehemPAUSA
| | - Jeetain Mittal
- Department of Chemical and Biomolecular EngineeringLehigh UniversityBethlehemPAUSA
| | - Anne C Hart
- Department of NeuroscienceBrown UniversityProvidenceRIUSA
| | - Nicolas L Fawzi
- Department of Molecular Pharmacology, Physiology, and BiotechnologyBrown UniversityProvidenceRIUSA
| |
Collapse
|
45
|
Osborne JF, Yanagi KS, Hart AC. Genetic interactions in a C. elegans sod-1 ALS model: glutamatergic neuron degeneration. MICROPUBLICATION BIOLOGY 2021; 2021. [PMID: 33474528 PMCID: PMC7812382 DOI: 10.17912/micropub.biology.000338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jennifer F Osborne
- Department of Neuroscience, Brown University, Providence, RI 02912.,Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912
| | - Katherine S Yanagi
- Department of Neuroscience, Brown University, Providence, RI 02912.,Neuroscience Graduate Program, Brown University, Providence, RI 02912.,Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912
| | - Anne C Hart
- Department of Neuroscience, Brown University, Providence, RI 02912.,Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912
| |
Collapse
|
46
|
McAlary L, Chew YL, Lum JS, Geraghty NJ, Yerbury JJ, Cashman NR. Amyotrophic Lateral Sclerosis: Proteins, Proteostasis, Prions, and Promises. Front Cell Neurosci 2020; 14:581907. [PMID: 33328890 PMCID: PMC7671971 DOI: 10.3389/fncel.2020.581907] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the progressive degeneration of the motor neurons that innervate muscle, resulting in gradual paralysis and culminating in the inability to breathe or swallow. This neuronal degeneration occurs in a spatiotemporal manner from a point of onset in the central nervous system (CNS), suggesting that there is a molecule that spreads from cell-to-cell. There is strong evidence that the onset and progression of ALS pathology is a consequence of protein misfolding and aggregation. In line with this, a hallmark pathology of ALS is protein deposition and inclusion formation within motor neurons and surrounding glia of the proteins TAR DNA-binding protein 43, superoxide dismutase-1, or fused in sarcoma. Collectively, the observed protein aggregation, in conjunction with the spatiotemporal spread of symptoms, strongly suggests a prion-like propagation of protein aggregation occurs in ALS. In this review, we discuss the role of protein aggregation in ALS concerning protein homeostasis (proteostasis) mechanisms and prion-like propagation. Furthermore, we examine the experimental models used to investigate these processes, including in vitro assays, cultured cells, invertebrate models, and murine models. Finally, we evaluate the therapeutics that may best prevent the onset or spread of pathology in ALS and discuss what lies on the horizon for treating this currently incurable disease.
Collapse
Affiliation(s)
- Luke McAlary
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Yee Lian Chew
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Jeremy Stephen Lum
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Nicholas John Geraghty
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Justin John Yerbury
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Neil R. Cashman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
47
|
Franklin JP, Azzouz M, Shaw PJ. SOD1-targeting therapies for neurodegenerative diseases: a review of current findings and future potential. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1835638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- John P. Franklin
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| |
Collapse
|
48
|
Caldwell KA, Willicott CW, Caldwell GA. Modeling neurodegeneration in Caenorhabditis elegans. Dis Model Mech 2020; 13:13/10/dmm046110. [PMID: 33106318 PMCID: PMC7648605 DOI: 10.1242/dmm.046110] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The global burden of neurodegenerative diseases underscores the urgent need for innovative strategies to define new drug targets and disease-modifying factors. The nematode Caenorhabditis elegans has served as the experimental subject for multiple transformative discoveries that have redefined our understanding of biology for ∼60 years. More recently, the considerable attributes of C. elegans have been applied to neurodegenerative diseases, including amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease and Huntington's disease. Transgenic nematodes with genes encoding normal and disease variants of proteins at the single- or multi-copy level under neuronal-specific promoters limits expression to select neuronal subtypes. The anatomical transparency of C. elegans affords the use of co-expressed fluorescent proteins to follow the progression of neurodegeneration as the animals age. Significantly, a completely defined connectome facilitates detailed understanding of the impact of neurodegeneration on organismal health and offers a unique capacity to accurately link cell death with behavioral dysfunction or phenotypic variation in vivo. Moreover, chemical treatments, as well as forward and reverse genetic screening, hasten the identification of modifiers that alter neurodegeneration. When combined, these chemical-genetic analyses establish critical threshold states to enhance or reduce cellular stress for dissecting associated pathways. Furthermore, C. elegans can rapidly reveal whether lifespan or healthspan factor into neurodegenerative processes. Here, we outline the methodologies employed to investigate neurodegeneration in C. elegans and highlight numerous studies that exemplify its utility as a pre-clinical intermediary to expedite and inform mammalian translational research. Summary: While unsurpassed as an experimental system for fundamental biology, Caenorhabditis elegans remains undervalued for its translational potential. Here, we highlight significant outcomes from, and resources available for, C. elegans-based research into neurodegenerative disorders.
Collapse
Affiliation(s)
- Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA .,Departments of Neurobiology, Neurology, Center for Neurodegeneration and Experimental Therapeutics, and Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corey W Willicott
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA.,Departments of Neurobiology, Neurology, Center for Neurodegeneration and Experimental Therapeutics, and Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
49
|
Agudelo A, St Amand V, Grissom L, Lafond D, Achilli T, Sahin A, Reenan R, Stilwell G. Age-dependent degeneration of an identified adult leg motor neuron in a Drosophila SOD1 model of ALS. Biol Open 2020; 9:bio049692. [PMID: 32994185 PMCID: PMC7595701 DOI: 10.1242/bio.049692] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 09/21/2020] [Indexed: 12/31/2022] Open
Abstract
Mutations in superoxide dismutase 1 (SOD1) cause familial amyotrophic lateral sclerosis (ALS) in humans. ALS is a neurodegenerative disease characterized by progressive motor neuron loss leading to paralysis and inevitable death in affected individuals. Using a gene replacement strategy to introduce disease mutations into the orthologous Drosophila sod1 (dsod1) gene, here, we characterize changes at the neuromuscular junction using longer-lived dsod1 mutant adults. Homozygous dsod1H71Y/H71Y or dsod1null/null flies display progressive walking defects with paralysis of the third metathoracic leg. In dissected legs, we assessed age-dependent changes in a single identified motor neuron (MN-I2) innervating the tibia levitator muscle. At adult eclosion, MN-I2 of dsod1H71Y/H71Y or sod1null/null flies is patterned similar to wild-type flies indicating no readily apparent developmental defects. Over the course of 10 days post-eclosion, MN-I2 shows an overall reduction in arborization with bouton swelling and loss of the post-synaptic marker discs-large (dlg) in mutant dsod1 adults. In addition, increases in polyubiquitinated proteins correlate with the timing and extent of MN-I2 changes. Because similar phenotypes are observed between flies homozygous for either dsod1H71Y or dsod1null alleles, we conclude these NMJ changes are mainly associated with sod loss-of-function. Together these studies characterize age-related morphological and molecular changes associated with axonal retraction in a Drosophila model of ALS that recapitulate an important aspect of the human disease.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Anthony Agudelo
- Department of Biology, Rhode Island College, 600 Mt. Pleasant Ave., Providence, RI, 02908 USA
| | - Victoria St Amand
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI, 02912 USA
| | - Lindsey Grissom
- Department of Biology, Rhode Island College, 600 Mt. Pleasant Ave., Providence, RI, 02908 USA
| | - Danielle Lafond
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI, 02912 USA
| | - Toni Achilli
- Department of Biology, Rhode Island College, 600 Mt. Pleasant Ave., Providence, RI, 02908 USA
| | - Asli Sahin
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI, 02912 USA
| | - Robert Reenan
- Department of Molecular Biology, Cell Biology & Biochemistry, Brown University, Providence, RI, 02912 USA
| | - Geoff Stilwell
- Department of Biology, Rhode Island College, 600 Mt. Pleasant Ave., Providence, RI, 02908 USA
| |
Collapse
|
50
|
Hahnel SR, Dilks CM, Heisler I, Andersen EC, Kulke D. Caenorhabditis elegans in anthelmintic research - Old model, new perspectives. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2020; 14:237-248. [PMID: 33249235 PMCID: PMC7704361 DOI: 10.1016/j.ijpddr.2020.09.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/13/2022]
Abstract
For more than four decades, the free-living nematode Caenorhabditis elegans has been extensively used in anthelmintic research. Classic genetic screens and heterologous expression in the C. elegans model enormously contributed to the identification and characterization of molecular targets of all major anthelmintic drug classes. Although these findings provided substantial insights into common anthelmintic mechanisms, a breakthrough in the treatment and control of parasitic nematodes is still not in sight. Instead, we are facing increasing evidence that the enormous diversity within the phylum Nematoda cannot be recapitulated by any single free-living or parasitic species and the development of novel broad-spectrum anthelmintics is not be a simple goal. In the present review, we summarize certain milestones and challenges of the C. elegans model with focus on drug target identification, anthelmintic drug discovery and identification of resistance mechanisms. Furthermore, we present new perspectives and strategies on how current progress in C. elegans research will support future anthelmintic research.
Collapse
Affiliation(s)
| | - Clayton M Dilks
- Northwestern University, Department of Molecular Biosciences, Evanston, IL, USA.
| | | | - Erik C Andersen
- Northwestern University, Department of Molecular Biosciences, Evanston, IL, USA.
| | | |
Collapse
|