1
|
Akaree N, Secco V, Levy-Adam F, Younis A, Carra S, Shalgi R. Regulation of physiological and pathological condensates by molecular chaperones. FEBS J 2025. [PMID: 39756021 DOI: 10.1111/febs.17390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/17/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
Biomolecular condensates are dynamic membraneless compartments that regulate a myriad of cellular functions. A particular type of physiological condensate called stress granules (SGs) has gained increasing interest due to its role in the cellular stress response and various diseases. SGs, composed of several hundred RNA-binding proteins, form transiently in response to stress to protect mRNAs from translation and disassemble when the stress subsides. Interestingly, SGs contain several aggregation-prone proteins, such as TDP-43, FUS, hnRNPA1, and others, which are typically found in pathological inclusions seen in autopsy tissues from amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) patients. Moreover, mutations in these genes lead to the familial form of ALS and FTD. This has led researchers to propose that pathological aggregation is seeded by aberrant SGs: SGs that fail to properly disassemble, lose their dynamic properties, and become pathological condensates which finally 'mature' into aggregates. Here, we discuss the evidence supporting this model for various ALS/FTD-associated proteins. We further continue to focus on molecular chaperone-mediated regulation of ALS/FTD-associated physiological condensates on one hand, and pathological condensates on the other. In addition to SGs, we review ALS/FTD-relevant nuclear condensates, namely paraspeckles, anisosomes, and nucleolar amyloid bodies, and discuss their emerging regulation by chaperones. As the majority of chaperoning mechanisms regulate physiological condensate disassembly, we highlight parallel themes of physiological and pathological condensation regulation across different chaperone families, underscoring the potential for early disease intervention.
Collapse
Affiliation(s)
- Nadeen Akaree
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Valentina Secco
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy
| | - Flonia Levy-Adam
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amal Younis
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Serena Carra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy
| | - Reut Shalgi
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
2
|
Huang L, Zhao B, Wan Y. Disruption of RNA-binding proteins in neurological disorders. Exp Neurol 2024; 385:115119. [PMID: 39709152 DOI: 10.1016/j.expneurol.2024.115119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/30/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
RNA-binding proteins (RBPs) are multifunctional proteins essential for the regulation of RNA processing and metabolism, contributing to the maintenance of cell homeostasis by modulating the expression of target genes. Many RBPs have been associated with neuron-specific processes vital for neuronal development and survival. RBP dysfunction may result in aberrations in RNA processing, which subsequently initiate a cascade of effects. Notably, RBPs are involved in the onset and progression of neurological disorders via diverse mechanisms. Disruption of RBPs not only affects RNA processing, but also promotes the abnormal aggregation of proteins into toxic inclusion bodies, and contributes to immune responses that drive the progression of neurological diseases. In this review, we summarize recent discoveries relating to the roles of RBPs in neurological diseases, discuss their contributions to such conditions, and highlight the unique functions of these RBPs within the nervous system.
Collapse
Affiliation(s)
- Luyang Huang
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Changchun 130062, Jilin, China
| | - Bo Zhao
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Changchun 130062, Jilin, China
| | - Youzhong Wan
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Changchun 130062, Jilin, China.
| |
Collapse
|
3
|
Tsumaki K, Bertens CJF, Nakayama M, Kato S, Jonao Y, Kuribayashi A, Sato K, Ishiyama S, Asakawa M, Aihara R, Yoshioka Y, Homma H, Tanaka H, Fujita K, Okazawa H, Sone M. Loss of function of VCP/TER94 causes neurodegeneration. Dis Model Mech 2024; 17:dmm050359. [PMID: 39629589 PMCID: PMC11698056 DOI: 10.1242/dmm.050359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
Variants in several genes are linked to human frontotemporal lobar degeneration (FTLD) associated with TDP43- and/or ubiquitin-positive inclusions. However, it is not yet clear whether the underlying mechanism is a gain-of-function or a loss-of-function one. To answer this question, we used Drosophila expressing double-stranded RNA against the FTLD-associated gene TER94 (an ortholog of VCP/p97) and found that the knockdown (KD) of this gene caused premature lethality, reduction in brain volume and alterations in the morphology of mushroom bodies. The changes caused by TER94 KD were rescued by wild-type TER94 but not by the human disease-linked A229E mutant, indicating that this mutant causes loss of function. Alterations were also observed in pupal brains and were partially rescued by co-expression of Mcm2, which is involved in control of the cell cycle, suggesting that dysregulation of neuronal proliferation caused the phenotypes. TER94 KD also caused the disappearance of TBPH (an ortholog of TDP43/TARDBP) from nuclei. These data from Drosophila genetics suggest that VCP-linked FTLD is caused by loss-of-function of VCP.
Collapse
Affiliation(s)
- Kohei Tsumaki
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Christian J. F. Bertens
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
- University Eye Clinic Maastricht, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6202AZ Maastricht, The Netherlands
- Department of Neuropathology, Institute of Science Tokyo, Tokyo 113-8510, Japan
| | - Minoru Nakayama
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Saya Kato
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Yuki Jonao
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Ayu Kuribayashi
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Konosuke Sato
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Shota Ishiyama
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Momoko Asakawa
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Riko Aihara
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Yuki Yoshioka
- Department of Neuropathology, Institute of Science Tokyo, Tokyo 113-8510, Japan
| | - Hidenori Homma
- Department of Neuropathology, Institute of Science Tokyo, Tokyo 113-8510, Japan
| | - Hikari Tanaka
- Department of Neuropathology, Institute of Science Tokyo, Tokyo 113-8510, Japan
| | - Kyota Fujita
- Department of Neuropathology, Institute of Science Tokyo, Tokyo 113-8510, Japan
| | - Hitoshi Okazawa
- Department of Neuropathology, Institute of Science Tokyo, Tokyo 113-8510, Japan
| | - Masaki Sone
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba 274-8510, Japan
- Department of Neuropathology, Institute of Science Tokyo, Tokyo 113-8510, Japan
| |
Collapse
|
4
|
Vanderhaeghe S, Prerad J, Tharkeshwar AK, Goethals E, Vints K, Beckers J, Scheveneels W, Debroux E, Princen K, Van Damme P, Fivaz M, Griffioen G, Van Den Bosch L. A pathogenic mutation in the ALS/FTD gene VCP induces mitochondrial hypermetabolism by modulating the permeability transition pore. Acta Neuropathol Commun 2024; 12:161. [PMID: 39390590 PMCID: PMC11465669 DOI: 10.1186/s40478-024-01866-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
Valosin-containing protein (VCP) is a ubiquitously expressed type II AAA+ ATPase protein, implicated in both amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). This study aimed to explore the impact of the disease-causing VCPR191Q/wt mutation on mitochondrial function using a CRISPR/Cas9-engineered neuroblastoma cell line. Mitochondria in these cells are enlarged, with a depolarized mitochondrial membrane potential associated with increased respiration and electron transport chain activity. Our results indicate that mitochondrial hypermetabolism could be caused, at least partially, by increased calcium-induced opening of the permeability transition pore (mPTP), leading to mild mitochondrial uncoupling. In conclusion, our findings reveal a central role of the ALS/FTD gene VCP in maintaining mitochondrial homeostasis and suggest a model of pathogenesis based on progressive alterations in mPTP physiology and mitochondrial energetics.
Collapse
Affiliation(s)
- Silke Vanderhaeghe
- Laboratory of Neurobiology, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium
- Laboratory of Neurobiology, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- reMYND, Leuven, Belgium
| | | | - Arun Kumar Tharkeshwar
- Department of Human Genetics, KU Leuven - University of Leuven, Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven - University of Leuven, Leuven, Belgium
| | - Elien Goethals
- Laboratory of Neurobiology, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium
- reMYND, Leuven, Belgium
| | - Katlijn Vints
- Electron Microscopy Platform and VIB-Bioimaging Core, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Jimmy Beckers
- Laboratory of Neurobiology, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium
- Laboratory of Neurobiology, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Wendy Scheveneels
- Laboratory of Neurobiology, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium
- Laboratory of Neurobiology, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | | | | | - Philip Van Damme
- Laboratory of Neurobiology, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | | | | | - Ludo Van Den Bosch
- Laboratory of Neurobiology, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium.
- Laboratory of Neurobiology, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.
| |
Collapse
|
5
|
Ramzan F, Kumar A, Abrar F, Gray RAV, Campbell ZE, Liao LMQ, Dang A, Akanni O, Guyn C, Martin DDO. Fatty links between multisystem proteinopathy and small VCP-interacting protein. Cell Death Discov 2024; 10:358. [PMID: 39117616 PMCID: PMC11310202 DOI: 10.1038/s41420-024-02118-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 07/09/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Multisystem proteinopathy (MSP) is a rare, dominantly inherited disorder that includes a cluster of diseases, including frontotemporal dementia, inclusion body myopathy, and Paget's disease of bone. MSP is caused by mutations in the gene encoding valosin-containing protein (VCP). Patients with the same mutation, even within the same family, can present with a different combination of any or all of the above diseases, along with amyotrophic lateral sclerosis (ALS). The pleiotropic effects may be linked to the greater than 50 VCP co-factors that direct VCP's many roles in the cell. Small VCP-interacting protein (SVIP) is a small protein that directs VCP to autophagosomes and lysosomes. We found that SVIP directs VCP localization to lysosomes in an acylation-dependent manner. We demonstrate that SVIP is myristoylated at Glycine 2 and palmitoylated at Cysteines 4 and 7. Acylation of SVIP is required to mediate cell death in the presence of the MSP-associated VCP variant (R155H-VCP), whereas blocking SVIP myristoylation prevents cytotoxicity. Therefore, SVIP acylation may present a novel target in MSP.
Collapse
Affiliation(s)
- Firyal Ramzan
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Ashish Kumar
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Fatima Abrar
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Rachel A V Gray
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Zurie E Campbell
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | | | - Anthony Dang
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | | | - Colm Guyn
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Dale D O Martin
- Department of Biology, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
6
|
Memudu AE, Olukade BA, Adebayo OS, Raza ML. Coffee and amyotrophic lateral sclerosis (ALS). PROGRESS IN BRAIN RESEARCH 2024; 289:81-105. [PMID: 39168583 DOI: 10.1016/bs.pbr.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder characterized by progressive loss of motor neurons. The effective treatments for ALS remain elusive, necessitating exploration into novel preventive strategies. ALS pathogenesis is triggered by oxidative stress which results in neuroinflammation, exicitotoxicity and neuronal cell death. Nutritional mechanism for halting progression of neurodegeneration is through dietary compounds with antioxidants, anti-inflammatory or neuromodulating activity. Coffee is a widely consumed beverage made up of polyphenols, caffeine and other compounds with possible antioxidants and neuro-protective roles. It is important to say that various epidemiological studies have documented association between coffee intake and ALS. This chapter is aimed to present a comprehensive review of existing literature on coffee consumption and ALS, involving epidemiological studies, preclinical research, and its mechanism of actions in animal model of ALS. It highlights key findings regarding the potential neuroprotective properties of coffee constituents such as caffeine, polyphenols, and other bioactive compounds. Furthermore, it discusses possible pathways through which coffee may modulate ALS pathogenesis, including suppressing oxidative stress and neuroinflammation while boosting adenosine function via the adenosine receptor two on the motor neuron cells membrane in the spinal cord to enhance motor function via the corticospinal tract. Overall, this chapter underscores the significance of further research to unravel the specific mechanisms by which coffee exerts its neuroprotective effects in ALS, with the ultimate goal of identifying dietary strategies for ALS prevention and management.
Collapse
Affiliation(s)
- Adejoke Elizabeth Memudu
- Anatomy Department, Neuroscience Unit, Faculty of Basic Medical Sciences Edo State University Uzairue, Edo State, Nigeria.
| | - Baliqis Adejoke Olukade
- Department of Molecular Medicine, Morsani College of Medicine, USF Health Byrd Alzheimer Institute, University of South Florida, Tampa, FL, United States
| | | | - Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Mathis S, Beauvais D, Duval F, Solé G, Le Masson G. The various forms of hereditary motor neuron disorders and their historical descriptions. J Neurol 2024; 271:3978-3990. [PMID: 38816479 DOI: 10.1007/s00415-024-12462-6] [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: 03/13/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
Motor neuron disorders comprise a clinically and pathologically heterogeneous group of neurologic diseases characterized by progressive degeneration of motor neurons (including both sporadic and hereditary diseases), affecting the upper motor neurons, lower motor neurons, or both. Hereditary motor neuron disorders themselves represent a vast and heterogeneous group, with numerous clinical and genetic overlaps that can be a source of error. This narrative review aims at providing an overview of the main types of inherited motor neuron disorders by recounting the stages in their historical descriptions. For practical purposes, this review of the literature sets out their various clinical characteristics and updates the list of all the genes involved in the various forms of inherited motor neuron disorders, including spinal muscular atrophy, familial amyotrophic lateral sclerosis, hereditary spastic paraplegia, distal hereditary motor neuropathies/neuronopathies, Kennedy's disease, riboflavin transporter deficiencies, VCPopathy and the neurogenic scapuloperoneal syndrome.
Collapse
Affiliation(s)
- Stéphane Mathis
- Department of Neurology, Nerve-Muscle Unit, University Hospital (CHU) of Bordeaux (Pellegrin Hospital), Place Amélie Raba Léon, 3300, Bordeaux, France.
- ALS Reference Center, Nerve-Muscle Unit, University Hospital (CHU) of Bordeaux (Pellegrin Hospital), Place Amélie Raba Léon, 3300, Bordeaux, France.
- Reference Center for Neuromuscular Diseases 'AOC', University Hospitals of Bordeaux (Pellegrin Hospital), University of Bordeaux, FILNEMUS, Euro-NMD, Bordeaux, France.
| | - Diane Beauvais
- Department of Neurology, Nerve-Muscle Unit, University Hospital (CHU) of Bordeaux (Pellegrin Hospital), Place Amélie Raba Léon, 3300, Bordeaux, France
- ALS Reference Center, Nerve-Muscle Unit, University Hospital (CHU) of Bordeaux (Pellegrin Hospital), Place Amélie Raba Léon, 3300, Bordeaux, France
- Reference Center for Neuromuscular Diseases 'AOC', University Hospitals of Bordeaux (Pellegrin Hospital), University of Bordeaux, FILNEMUS, Euro-NMD, Bordeaux, France
| | - Fanny Duval
- Department of Neurology, Nerve-Muscle Unit, University Hospital (CHU) of Bordeaux (Pellegrin Hospital), Place Amélie Raba Léon, 3300, Bordeaux, France
- Reference Center for Neuromuscular Diseases 'AOC', University Hospitals of Bordeaux (Pellegrin Hospital), University of Bordeaux, FILNEMUS, Euro-NMD, Bordeaux, France
| | - Guilhem Solé
- Department of Neurology, Nerve-Muscle Unit, University Hospital (CHU) of Bordeaux (Pellegrin Hospital), Place Amélie Raba Léon, 3300, Bordeaux, France
- Reference Center for Neuromuscular Diseases 'AOC', University Hospitals of Bordeaux (Pellegrin Hospital), University of Bordeaux, FILNEMUS, Euro-NMD, Bordeaux, France
| | - Gwendal Le Masson
- Department of Neurology, Nerve-Muscle Unit, University Hospital (CHU) of Bordeaux (Pellegrin Hospital), Place Amélie Raba Léon, 3300, Bordeaux, France
- ALS Reference Center, Nerve-Muscle Unit, University Hospital (CHU) of Bordeaux (Pellegrin Hospital), Place Amélie Raba Léon, 3300, Bordeaux, France
- Reference Center for Neuromuscular Diseases 'AOC', University Hospitals of Bordeaux (Pellegrin Hospital), University of Bordeaux, FILNEMUS, Euro-NMD, Bordeaux, France
| |
Collapse
|
8
|
Magrinelli F, Tesson C, Angelova PR, Salazar-Villacorta A, Rodriguez JA, Scardamaglia A, Chung BHY, Jaconelli M, Vona B, Esteras N, Kwong AKY, Courtin T, Maroofian R, Alavi S, Nirujogi R, Severino M, Lewis PA, Efthymiou S, O’Callaghan B, Buchert R, Sofan L, Lis P, Pinon C, Breedveld GJ, Chui MMC, Murphy D, Pitz V, Makarious MB, Cassar M, Hassan BA, Iftikhar S, Rocca C, Bauer P, Tinazzi M, Svetel M, Samanci B, Hanağası HA, Bilgiç B, Obeso JA, Kurtis MM, Cogan G, Başak AN, Kiziltan G, Gül T, Yalçın G, Elibol B, Barišić N, Ng EWS, Fan SS, Hershkovitz T, Weiss K, Raza Alvi J, Sultan T, Azmi Alkhawaja I, Froukh T, E Alrukban HA, Fauth C, Schatz UA, Zöggeler T, Zech M, Stals K, Varghese V, Gandhi S, Blauwendraat C, Hardy JA, Lesage S, Bonifati V, Haack TB, Bertoli-Avella AM, Steinfeld R, Alessi DR, Steller H, Brice A, Abramov AY, Bhatia KP, Houlden H. PSMF1 variants cause a phenotypic spectrum from early-onset Parkinson's disease to perinatal lethality by disrupting mitochondrial pathways. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.19.24308302. [PMID: 39148840 PMCID: PMC11326324 DOI: 10.1101/2024.06.19.24308302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Dissecting biological pathways highlighted by Mendelian gene discovery has provided critical insights into the pathogenesis of Parkinson's disease (PD) and neurodegeneration. This approach ultimately catalyzes the identification of potential biomarkers and therapeutic targets. Here, we identify PSMF1 as a new gene implicated in PD and childhood neurodegeneration. We find that biallelic PSMF1 missense and loss-of-function variants co-segregate with phenotypes from early-onset PD and parkinsonism to perinatal lethality with neurological manifestations across 15 unrelated pedigrees with 22 affected subjects, showing clear genotype-phenotype correlation. PSMF1 encodes the proteasome regulator PSMF1/PI31, a highly conserved, ubiquitously expressed partner of the 20S proteasome and neurodegeneration-associated F-box-O 7 and valosin-containing proteins. We demonstrate that PSMF1 variants impair mitochondrial membrane potential, dynamics and mitophagy in patient-derived fibroblasts. Additionally, we develop models of psmf1 knockdown Drosophila and Psmf1 conditional knockout mouse exhibiting age-dependent motor impairment, with diffuse gliosis in mice. These findings unequivocally link defective PSMF1 to early-onset PD and neurodegeneration and suggest mitochondrial dysfunction as a mechanistic contributor.
Collapse
Affiliation(s)
- Francesca Magrinelli
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Christelle Tesson
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Plamena R. Angelova
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Ainara Salazar-Villacorta
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Jose A. Rodriguez
- Laboratory of Apoptosis and Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Annarita Scardamaglia
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Brian Hon-Yin Chung
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong Genome Institute, Hong Kong SAR, China
| | - Matthew Jaconelli
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Barbara Vona
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and Inner Ear Lab, University Medical Center Göttingen, Göttingen, Germany
| | - Noemi Esteras
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- Neurochemistry Research Institute, Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, Madrid, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| | - Anna Ka-Yee Kwong
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Thomas Courtin
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Reza Maroofian
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Shahryar Alavi
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Raja Nirujogi
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | | | - Patrick A. Lewis
- Royal Veterinary College, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Stephanie Efthymiou
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Benjamin O’Callaghan
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Rebecca Buchert
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Linda Sofan
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Pawel Lis
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Chloé Pinon
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Guido J. Breedveld
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Martin Man-Chun Chui
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David Murphy
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Vanessa Pitz
- Integrative Neurogenomics Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Mary B. Makarious
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Marlene Cassar
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Bassem A. Hassan
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Sana Iftikhar
- Department of Real-World evidence studies, CENTOGENE GmbH, Rostock, Germany
| | - Clarissa Rocca
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Peter Bauer
- Department of Medical Genetics, CENTOGENE GmbH, Rostock, Germany
| | - Michele Tinazzi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marina Svetel
- Movement Disorders Department, Neurology Clinic, University Clinical Center of Serbia, Belgrade, Serbia
| | - Bedia Samanci
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Haşmet A. Hanağası
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Basar Bilgiç
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - José A. Obeso
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
- HM CINAC, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- University CEU-San Pablo, Madrid, Spain
| | - Monica M. Kurtis
- Neurology Department, Hospital Ruber Internacional, Madrid, Spain
| | - Guillaume Cogan
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Ayşe Nazlı Başak
- Koç University, School of Medicine, Research Center for Translational Medicine KUTTAM-Neurodegeneration Research Laboratory NDAL, Istanbul, Turkey
| | - Güneş Kiziltan
- Department of Neurology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Tuğçe Gül
- Koç University, School of Medicine, Research Center for Translational Medicine KUTTAM-Neurodegeneration Research Laboratory NDAL, Istanbul, Turkey
| | - Gül Yalçın
- Department of Neurology, School of Medicine, Hacettepe University, Ankara, Turkey
| | - Bülent Elibol
- Department of Neurology, School of Medicine, Hacettepe University, Ankara, Turkey
| | - Nina Barišić
- Department of Pediatrics, University of Zagreb Medical School and University Hospital Center Zagreb, Zagreb, Croatia
| | - Earny Wei-Sen Ng
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sze-Shing Fan
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Tova Hershkovitz
- The Genetics Institute, Galilee Medical Center, Nahariya, Israel
| | - Karin Weiss
- Genetics Institute, Rambam Health Care Center, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Javeria Raza Alvi
- Department of Paediatric Neurology, The Children’s Hospital and the University of Child Health Sciences, Lahore, Punjab, Pakistan
| | - Tipu Sultan
- Department of Paediatric Neurology, The Children’s Hospital and the University of Child Health Sciences, Lahore, Punjab, Pakistan
| | - Issam Azmi Alkhawaja
- Pediatric Neurology Unit, Pediatric Department, Albashir Hospital, Amman, Jordan
| | - Tawfiq Froukh
- Department of Biotechnology and Genetics Engineering, Philadelphia University, Jordan
| | | | - Christine Fauth
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, Austria
| | - Ulrich A. Schatz
- Institute of Human Genetics, Medical University Innsbruck, Innsbruck, Austria
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Thomas Zöggeler
- Department of Pediatrics I, Medical University Innsbruck, Innsbruck, Austria
| | - Michael Zech
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum Munich, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | - Karen Stals
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | - Vinod Varghese
- All Wales Medical Genomics Service, Cardiff, United Kingdom
| | - Sonia Gandhi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| | - Cornelis Blauwendraat
- Integrative Neurogenomics Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - John A. Hardy
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Suzanne Lesage
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Vincenzo Bonifati
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tobias B. Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Institute of Neurogenomics, Helmholtz Zentrum Munich, Munich, Germany
| | | | - Robert Steinfeld
- Department of Pediatrics and Pediatric Neurology, University of Göttingen, Göttingen, Germany
- Department of Pediatric Neurology, Charité University Medicine, Berlin, Germany
| | - Dario R. Alessi
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Hermann Steller
- Laboratory of Apoptosis and Cancer Biology, The Rockefeller University, New York, NY, USA
| | - Alexis Brice
- Institut du Cerveau et de la Moelle épinière, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Andrey Y. Abramov
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Kailash P. Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
9
|
Brandão-Teles C, Zuccoli GS, de Moraes Vrechi TA, Ramos-da-Silva L, Santos AVS, Crunfli F, Martins-de-Souza D. Induced-pluripotent stem cells and neuroproteomics as tools for studying neurodegeneration. Biochem Soc Trans 2024; 52:163-176. [PMID: 38288874 DOI: 10.1042/bst20230341] [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: 10/16/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 02/29/2024]
Abstract
The investigation of neurodegenerative diseases advanced significantly with the advent of cell-reprogramming technology, leading to the creation of new models of human illness. These models, derived from induced pluripotent stem cells (iPSCs), facilitate the study of sporadic as well as hereditary diseases and provide a comprehensive understanding of the molecular mechanisms involved with neurodegeneration. Through proteomics, a quantitative tool capable of identifying thousands of proteins from small sample volumes, researchers have attempted to identify disease mechanisms by detecting differentially expressed proteins and proteoforms in disease models, biofluids, and postmortem brain tissue. The integration of these two technologies allows for the identification of novel pathological targets within the realm of neurodegenerative diseases. Here, we highlight studies from the past 5 years on the contributions of iPSCs within neuroproteomic investigations, which uncover the molecular mechanisms behind these illnesses.
Collapse
Affiliation(s)
- Caroline Brandão-Teles
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Giuliana S Zuccoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Talita Aparecida de Moraes Vrechi
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Lívia Ramos-da-Silva
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Aline Valéria Sousa Santos
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Fernanda Crunfli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas 13083-862, SP, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria, Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
- INCT in Modelling Human Complex Diseases with 3D Platforms (Model3D)
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil
| |
Collapse
|
10
|
Scarian E, Viola C, Dragoni F, Di Gerlando R, Rizzo B, Diamanti L, Gagliardi S, Bordoni M, Pansarasa O. New Insights into Oxidative Stress and Inflammatory Response in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:2698. [PMID: 38473944 DOI: 10.3390/ijms25052698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Oxidative stress (OS) and inflammation are two important and well-studied pathological hallmarks of neurodegenerative diseases (NDDs). Due to elevated oxygen consumption, the high presence of easily oxidizable polyunsaturated fatty acids and the weak antioxidant defenses, the brain is particularly vulnerable to oxidative injury. Uncertainty exists over whether these deficits contribute to the development of NDDs or are solely a consequence of neuronal degeneration. Furthermore, these two pathological hallmarks are linked, and it is known that OS can affect the inflammatory response. In this review, we will overview the last findings about these two pathways in the principal NDDs. Moreover, we will focus more in depth on amyotrophic lateral sclerosis (ALS) to understand how anti-inflammatory and antioxidants drugs have been used for the treatment of this still incurable motor neuron (MN) disease. Finally, we will analyze the principal past and actual clinical trials and the future perspectives in the study of these two pathological mechanisms.
Collapse
Affiliation(s)
- Eveljn Scarian
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Camilla Viola
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Via Agostino Bassi 21, 27100 Pavia, Italy
| | - Francesca Dragoni
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Adolfo Ferrata, 9, 27100 Pavia, Italy
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Rosalinda Di Gerlando
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Adolfo Ferrata, 9, 27100 Pavia, Italy
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Bartolo Rizzo
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Luca Diamanti
- Neuroncology Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Stella Gagliardi
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Matteo Bordoni
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| | - Orietta Pansarasa
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy
| |
Collapse
|
11
|
Noori L, Saqagandomabadi V, Di Felice V, David S, Caruso Bavisotto C, Bucchieri F, Cappello F, Conway de Macario E, Macario AJL, Scalia F. Putative Roles and Therapeutic Potential of the Chaperone System in Amyotrophic Lateral Sclerosis and Multiple Sclerosis. Cells 2024; 13:217. [PMID: 38334609 PMCID: PMC10854686 DOI: 10.3390/cells13030217] [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: 12/15/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
The putative pathogenic roles and therapeutic potential of the chaperone system (CS) in amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS) are reviewed to provide a bibliographic and conceptual platform for launching research on the diagnostic and therapeutic applications of CS components. Various studies suggest that dysfunction of the CS contributes to the pathogenesis of ALS and MS, and here, we identify some of the implicated CS members. The physiology and pathophysiology of the CS members can be properly understood if they are studied or experimentally or clinically manipulated for diagnostic or therapeutic purposes, bearing in mind that they belong to a physiological system with multiple interacting and dynamic components, widespread throughout the body, intra- and extracellularly. Molecular chaperones, some called heat shock protein (Hsp), are the chief components of the CS, whose canonical functions are cytoprotective. However, abnormal chaperones can be etiopathogenic factors in a wide range of disorders, chaperonopathies, including ALS and MS, according to the data reviewed. Chaperones typically form teams, and these build functional networks to maintain protein homeostasis, the canonical role of the CS. However, members of the CS also display non-canonical functions unrelated to protein homeostasis. Therefore, chaperones and other members of the CS, if abnormal, may disturb not only protein synthesis, maturation, and migration but also other physiological processes. Thus, in elucidating the role of CS components in ALS and MS, one must look at protein homeostasis abnormalities and beyond, following the clues emerging from the works discussed here.
Collapse
Affiliation(s)
- Leila Noori
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (L.N.); (V.S.); (V.D.F.); (S.D.); (C.C.B.); (F.B.); (F.C.)
- Department of Anatomy, School of Medicine, Medical University of Babol, Babol 47176-47745, Iran
| | - Vahid Saqagandomabadi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (L.N.); (V.S.); (V.D.F.); (S.D.); (C.C.B.); (F.B.); (F.C.)
| | - Valentina Di Felice
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (L.N.); (V.S.); (V.D.F.); (S.D.); (C.C.B.); (F.B.); (F.C.)
| | - Sabrina David
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (L.N.); (V.S.); (V.D.F.); (S.D.); (C.C.B.); (F.B.); (F.C.)
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (L.N.); (V.S.); (V.D.F.); (S.D.); (C.C.B.); (F.B.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
| | - Fabio Bucchieri
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (L.N.); (V.S.); (V.D.F.); (S.D.); (C.C.B.); (F.B.); (F.C.)
| | - Francesco Cappello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (L.N.); (V.S.); (V.D.F.); (S.D.); (C.C.B.); (F.B.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
| | - Everly Conway de Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore—Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore—Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
| | - Federica Scalia
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (L.N.); (V.S.); (V.D.F.); (S.D.); (C.C.B.); (F.B.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
| |
Collapse
|
12
|
Marks JD, Ayuso VE, Carlomagno Y, Yue M, Todd TW, Hao Y, Li Z, McEachin ZT, Shantaraman A, Duong DM, Daughrity LM, Jansen-West K, Shao W, Calliari A, Bejarano JG, DeTure M, Rawlinson B, Casey MC, Lilley MT, Donahue MH, Jawahar VM, Boeve BF, Petersen RC, Knopman DS, Oskarsson B, Graff-Radford NR, Wszolek ZK, Dickson DW, Josephs KA, Qi YA, Seyfried NT, Ward ME, Zhang YJ, Prudencio M, Petrucelli L, Cook CN. TMEM106B core deposition associates with TDP-43 pathology and is increased in risk SNP carriers for frontotemporal dementia. Sci Transl Med 2024; 16:eadf9735. [PMID: 38232138 PMCID: PMC10841341 DOI: 10.1126/scitranslmed.adf9735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/18/2023] [Indexed: 01/19/2024]
Abstract
Genetic variation at the transmembrane protein 106B gene (TMEM106B) has been linked to risk of frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP) through an unknown mechanism. We found that presence of the TMEM106B rs3173615 protective genotype was associated with longer survival after symptom onset in a postmortem FTLD-TDP cohort, suggesting a slower disease course. The seminal discovery that filaments derived from TMEM106B is a common feature in aging and, across a range of neurodegenerative disorders, suggests that genetic variants in TMEM106B could modulate disease risk and progression through modulating TMEM106B aggregation. To explore this possibility and assess the pathological relevance of TMEM106B accumulation, we generated a new antibody targeting the TMEM106B filament core sequence. Analysis of postmortem samples revealed that the TMEM106B rs3173615 risk allele was associated with higher TMEM106B core accumulation in patients with FTLD-TDP. In contrast, minimal TMEM106B core deposition was detected in carriers of the protective allele. Although the abundance of monomeric full-length TMEM106B was unchanged, carriers of the protective genotype exhibited an increase in dimeric full-length TMEM106B. Increased TMEM106B core deposition was also associated with enhanced TDP-43 dysfunction, and interactome data suggested a role for TMEM106B core filaments in impaired RNA transport, local translation, and endolysosomal function in FTLD-TDP. Overall, these findings suggest that prevention of TMEM106B core accumulation is central to the mechanism by which the TMEM106B protective haplotype reduces disease risk and slows progression.
Collapse
Affiliation(s)
- Jordan D. Marks
- Medical Scientist Training Program, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | - Virginia Estades Ayuso
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yari Carlomagno
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Mei Yue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tiffany W. Todd
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ying Hao
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ziyi Li
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zachary T. McEachin
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30307, USA
- Department for Human Genetics, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Anantharaman Shantaraman
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Duc M. Duong
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30307, USA
| | | | - Karen Jansen-West
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Wei Shao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Anna Calliari
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Bailey Rawlinson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Meredith T. Lilley
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | - Megan H. Donahue
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | | | | | - Björn Oskarsson
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | - Dennis W. Dickson
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Yue A. Qi
- Center for Alzheimer’s and Related Dementias (CARD), National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicholas T. Seyfried
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Michael E. Ward
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong-Jie Zhang
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Mercedes Prudencio
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Leonard Petrucelli
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Casey N. Cook
- Neuroscience Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
13
|
Geraci J, Bhargava R, Qorri B, Leonchyk P, Cook D, Cook M, Sie F, Pani L. Machine learning hypothesis-generation for patient stratification and target discovery in rare disease: our experience with Open Science in ALS. Front Comput Neurosci 2024; 17:1199736. [PMID: 38260713 PMCID: PMC10801647 DOI: 10.3389/fncom.2023.1199736] [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: 04/03/2023] [Accepted: 11/20/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Advances in machine learning (ML) methodologies, combined with multidisciplinary collaborations across biological and physical sciences, has the potential to propel drug discovery and development. Open Science fosters this collaboration by releasing datasets and methods into the public space; however, further education and widespread acceptance and adoption of Open Science approaches are necessary to tackle the plethora of known disease states. Motivation In addition to providing much needed insights into potential therapeutic protein targets, we also aim to demonstrate that small patient datasets have the potential to provide insights that usually require many samples (>5,000). There are many such datasets available and novel advancements in ML can provide valuable insights from these patient datasets. Problem statement Using a public dataset made available by patient advocacy group AnswerALS and a multidisciplinary Open Science approach with a systems biology augmented ML technology, we aim to validate previously reported drug targets in ALS and provide novel insights about ALS subpopulations and potential drug targets using a unique combination of ML methods and graph theory. Methodology We use NetraAI to generate hypotheses about specific patient subpopulations, which were then refined and validated through a combination of ML techniques, systems biology methods, and expert input. Results We extracted 8 target classes, each comprising of several genes that shed light into ALS pathophysiology and represent new avenues for treatment. These target classes are broadly categorized as inflammation, epigenetic, heat shock, neuromuscular junction, autophagy, apoptosis, axonal transport, and excitotoxicity. These findings are not mutually exclusive, and instead represent a systematic view of ALS pathophysiology. Based on these findings, we suggest that simultaneous targeting of ALS has the potential to mitigate ALS progression, with the plausibility of maintaining and sustaining an improved quality of life (QoL) for ALS patients. Even further, we identified subpopulations based on disease onset. Conclusion In the spirit of Open Science, this work aims to bridge the knowledge gap in ALS pathophysiology to aid in diagnostic, prognostic, and therapeutic strategies and pave the way for the development of personalized treatments tailored to the individual's needs.
Collapse
Affiliation(s)
- Joseph Geraci
- NetraMark Corp, Toronto, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
- Centre for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Arthur C. Clarke Center for Human Imagination, School of Physical Sciences, University of California San Diego, San Diego, CA, United States
| | - Ravi Bhargava
- Department of Biomedical and Molecular Science, Queens University, Kingston, ON, Canada
- Science and Research, Roche Integrated Informatics, F. Hoffmann La-Roche, Toronto, ON, Canada
| | | | | | - Douglas Cook
- NetraMark Corp, Toronto, ON, Canada
- Department of Surgery, Queen's University, Kingston, ON, Canada
| | - Moses Cook
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Fanny Sie
- Science and Research, Roche Integrated Informatics, F. Hoffmann La-Roche, Toronto, ON, Canada
| | - Luca Pani
- NetraMark Corp, Toronto, ON, Canada
- Department of Psychiatry and Behavioral Sciences, Leonard M. Miller School of Medicine, University of Miami, Coral Gables, FL, United States
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
14
|
Jensen BK. Astrocyte-Neuron Interactions Contributing to Amyotrophic Lateral Sclerosis Progression. ADVANCES IN NEUROBIOLOGY 2024; 39:285-318. [PMID: 39190080 DOI: 10.1007/978-3-031-64839-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex disease impacting motor neurons of the brain, brainstem, and spinal cord. Disease etiology is quite heterogeneous with over 40 genes causing the disease and a vast ~90% of patients having no prior family history. Astrocytes are major contributors to ALS, particularly through involvement in accelerating disease progression. Through study of genetic forms of disease including SOD1, TDP43, FUS, C9orf72, VCP, TBK1, and more recently patient-derived cells from sporadic individuals, many biological mechanisms have been identified to cause intrinsic or glial-mediated neurotoxicity to motor neurons. Overall, many of the normally supportive and beneficial roles that astrocytes contribute to neuronal health and survival instead switch to become deleterious and neurotoxic. While the exact pathways may differ based on disease-origin, altered astrocyte-neuron communication is a common feature of ALS. Within this chapter, distinct genetic forms are examined in detail, along with what is known from sporadic patient-derived cells. Overall, this chapter highlights the interplay between astrocytes and neurons in this complex disease and describes the key features underlying: astrocyte-mediated motor neuron toxicity, excitotoxicity, oxidative/nitrosative stress, protein dyshomeostasis, metabolic imbalance, inflammation, trophic factor withdrawal, blood-brain/blood-spinal cord barrier involvement, disease spreading, and the extracellular matrix/cell adhesion/TGF-β signaling pathways.
Collapse
Affiliation(s)
- Brigid K Jensen
- Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Todd TW, Shao W, Zhang YJ, Petrucelli L. The endolysosomal pathway and ALS/FTD. Trends Neurosci 2023; 46:1025-1041. [PMID: 37827960 PMCID: PMC10841821 DOI: 10.1016/j.tins.2023.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 08/23/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are considered to be part of a disease spectrum that is associated with causative mutations and risk variants in a wide range of genes. Mounting evidence indicates that several of these genes are linked to the endolysosomal system, highlighting the importance of this pathway in ALS/FTD. Although many studies have focused on how disruption of this pathway impacts on autophagy, recent findings reveal that this may not be the whole picture: specifically, disrupting autophagy may not be sufficient to induce disease, whereas disrupting the endolysosomal system could represent a crucial pathogenic driver. In this review we discuss the connections between ALS/FTD and the endolysosomal system, including a breakdown of how disease-associated genes are implicated in this pathway. We also explore the potential downstream consequences of disrupting endolysosomal activity in the brain, outside of an effect on autophagy.
Collapse
Affiliation(s)
- Tiffany W Todd
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Wei Shao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|
16
|
Wentink A, Rosenzweig R. Protein disaggregation machineries in the human cytosol. Curr Opin Struct Biol 2023; 83:102735. [PMID: 38000128 DOI: 10.1016/j.sbi.2023.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023]
Abstract
Proteins carry out the vast majority of functions in cells, but can only do so when properly folded. Following stress or mutation, proteins can lose their proper fold, resulting in misfolding, inactivity, and aggregation-posing a threat to cellular health. In order to counteract protein aggregation, cells have evolved a remarkable subset of molecular chaperones, called protein disaggregases, which collaboratively possess the ability to forcibly untangle protein aggregates. Here, we review the different chaperone disaggregation machineries present in the human cytosol and their mechanisms of action. Understanding, how these disaggregases function, is both universally and clinically important, as protein aggregation has been linked to multiple, debilitating neurodegenerative diseases.
Collapse
Affiliation(s)
- Anne Wentink
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, Netherlands.
| | - Rina Rosenzweig
- Chemical and Structural Biology Department, Weizmann Institute of Science, Rehovot, 761000, Israel.
| |
Collapse
|
17
|
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: 3] [Impact Index Per Article: 1.5] [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
|
18
|
Li C, Wen Y, Zhao M, Wang Y, Li P, Wang L, Wang S. A novel splice-site mutation in CHMP2B associated with frontotemporal dementia: The first report from China and literature review. Mol Genet Genomic Med 2023; 11:e2222. [PMID: 37272767 PMCID: PMC10422061 DOI: 10.1002/mgg3.2222] [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: 11/15/2022] [Revised: 05/05/2023] [Accepted: 05/23/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND Frontotemporal dementia (FTD) has genetic heterogeneity, and the endosomal ESCRTIII-complex subunit CHMP2B variant is a rare cause of FTD. The mutations in CHMP2B were first identified in a large Danish pedigree with autosomal dominant FTD, and have also been found in several individuals from Belgium, France, the United States, and Türkiye. In the Chinese population, cases of CHMP2B variant-associated FTD have never been reported. METHODS The spectrum of clinical symptoms and the genetic analysis of the presented patient were identified and investigated. Besides this case, we assessed previously reported cases with CHMP2B gene mutations. RESULTS This study presents a Chinese patient harboring a novel heterozygous A-to-T variant (NM_014043:c.532-2A>T) in CHMP2B with a phenotype compatible with FTD. Although previous reports suggested cases of CHMP2B variant-associated FTD initially presented with personality changes and stereotypical movements at the age of 50, this case was characterized by psychosis involving delusion of persecution, auditory hallucination, and suspiciousness at the earlier onset age of 44. Minigene splicing assay revealed that the splice-site variant could result in the retention of intron 5. CONCLUSION This is the first case of CHMP2B variant-associated FTD reported in the Chinese population. The novel c.532-2A>T variant in the acceptor splice site of exon 6 retaining intron 5 was predicted to cause truncated protein and protein conformation changes. This discovery may expand the genetic and phenotypic spectrum of CHMP2B variant-associated FTD.
Collapse
Affiliation(s)
- Chang Li
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Ya Wen
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Mengqiu Zhao
- Neurological Laboratory of Hebei ProvinceShijiazhuangChina
| | - Yaye Wang
- Neurological Laboratory of Hebei ProvinceShijiazhuangChina
| | - Ping Li
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
- Neurological Laboratory of Hebei ProvinceShijiazhuangChina
| | - Liang Wang
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
- Neurological Laboratory of Hebei ProvinceShijiazhuangChina
| | - Shan Wang
- Department of NeurologySecond Hospital of Hebei Medical UniversityShijiazhuangChina
- Neurological Laboratory of Hebei ProvinceShijiazhuangChina
| |
Collapse
|
19
|
Martinelli I, Zucchi E, Simonini C, Gianferrari G, Zamboni G, Pinti M, Mandrioli J. The landscape of cognitive impairment in superoxide dismutase 1-amyotrophic lateral sclerosis. Neural Regen Res 2023; 18:1427-1433. [PMID: 36571338 PMCID: PMC10075107 DOI: 10.4103/1673-5374.361535] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although mutations in the superoxide dismutase 1 gene account for only a minority of total amyotrophic lateral sclerosis cases, the discovery of this gene has been crucial for amyotrophic lateral sclerosis research. Since the identification of superoxide dismutase 1 in 1993, the field of amyotrophic lateral sclerosis genetics has considerably widened, improving our understanding of the diverse pathogenic basis of amyotrophic lateral sclerosis. In this review, we focus on cognitive impairment in superoxide dismutase 1-amyotrophic lateral sclerosis patients. Literature has mostly reported that cognition remains intact in superoxide dismutase 1-amyotrophic lateral sclerosis patients, but recent reports highlight frontal lobe function frailty in patients carrying different superoxide dismutase 1-amyotrophic lateral sclerosis mutations. We thoroughly reviewed all the various mutations reported in the literature to contribute to a comprehensive database of superoxide dismutase 1-amyotrophic lateral sclerosis genotype-phenotype correlation. Such a resource could ultimately improve our mechanistic understanding of amyotrophic lateral sclerosis, enabling a more robust assessment of how the amyotrophic lateral sclerosis phenotype responds to different variants across genes, which is important for the therapeutic strategy targeting genetic mutations. Cognition in superoxide dismutase 1-amyotrophic lateral sclerosis deserves further longitudinal research since this peculiar frailty in patients with similar mutations can be conditioned by external factors, including environment and other unidentified agents including modifier genes.
Collapse
Affiliation(s)
- Ilaria Martinelli
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia; Department of Neurosciences, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Elisabetta Zucchi
- Department of Neurosciences, Azienda Ospedaliero-Universitaria di Modena; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cecilia Simonini
- Department of Neurosciences, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Giulia Gianferrari
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanna Zamboni
- Department of Neurosciences, Azienda Ospedaliero-Universitaria di Modena; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jessica Mandrioli
- Department of Neurosciences, Azienda Ospedaliero-Universitaria di Modena; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
20
|
Gao Y, Zhu Y, Sun Q, Chen D. Argonaute-dependent ribosome-associated protein quality control. Trends Cell Biol 2023; 33:260-272. [PMID: 35981909 DOI: 10.1016/j.tcb.2022.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022]
Abstract
Ribosome-associated protein quality control (RQC) is a protein surveillance mechanism that eliminates defective nascent polypeptides. The E3 ubiquitin ligase, Ltn1, is a key regulator of RQC that targets substrates for ubiquitination. Argonaute proteins (AGOs) are central players in miRNA-mediated gene silencing and have recently been shown to also regulate RQC by facilitating Ltn1. Therefore, AGOs directly coordinate post-transcriptional gene silencing and RQC, ensuring efficient gene silencing. We summarize the principles of RQC and the functions of AGOs in miRNA-mediated gene silencing, and discuss how AGOs associate with the endoplasmic reticulum (ER) to assist Ltn1 in controlling RQC. We highlight that RQC not only eliminates defective nascent polypeptides but also removes unwanted protein products when AGOs participate.
Collapse
Affiliation(s)
- Yajie Gao
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China
| | - Yuanxiang Zhu
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China
| | - Qinmiao Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem Cells and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| | - Dahua Chen
- Institute of Biomedical Research, Yunnan University, Kunming 650500, China.
| |
Collapse
|
21
|
McCluskey G, Morrison KE, Donaghy C, Rene F, Duddy W, Duguez S. Extracellular Vesicles in Amyotrophic Lateral Sclerosis. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010121. [PMID: 36676070 PMCID: PMC9867379 DOI: 10.3390/life13010121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023]
Abstract
Amyotrophic Lateral Sclerosis is a progressive neurodegenerative disease and is the most common adult motor neuron disease. The disease pathogenesis is complex with the perturbation of multiple pathways proposed, including mitochondrial dysfunction, RNA processing, glutamate excitotoxicity, endoplasmic reticulum stress, protein homeostasis and endosomal transport/extracellular vesicle (EV) secretion. EVs are nanoscopic membrane-bound particles that are released from cells, involved in the intercellular communication of proteins, lipids and genetic material, and there is increasing evidence of their role in ALS. After discussing the biogenesis of EVs, we review their roles in the propagation of pathological proteins in ALS, such as TDP-43, SOD1 and FUS, and their contribution to disease pathology. We also discuss the ALS related genes which are involved in EV formation and vesicular trafficking, before considering the EV protein and RNA dysregulation found in ALS and how these have been investigated as potential biomarkers. Finally, we highlight the potential use of EVs as therapeutic agents in ALS, in particular EVs derived from mesenchymal stem cells and EVs as drug delivery vectors for potential treatment strategies.
Collapse
Affiliation(s)
- Gavin McCluskey
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry BT47 6SB, UK
- Department of Neurology, Altnagelvin Hospital, Derry BT47 6SB, UK
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
- Correspondence: (G.M.); (S.D.)
| | - Karen E. Morrison
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
- Faculty of Medicine, Health & Life Sciences, Queen’s University, Belfast BT9 6AG, UK
| | - Colette Donaghy
- Department of Neurology, Altnagelvin Hospital, Derry BT47 6SB, UK
| | - Frederique Rene
- INSERM U1118, Centre de Recherche en Biomédecine de Strasbourg, Université de Strasbourg, 67000 Strasbourg, France
| | - William Duddy
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry BT47 6SB, UK
| | - Stephanie Duguez
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry BT47 6SB, UK
- Correspondence: (G.M.); (S.D.)
| |
Collapse
|
22
|
Ahlstedt BA, Ganji R, Raman M. The functional importance of VCP to maintaining cellular protein homeostasis. Biochem Soc Trans 2022; 50:1457-1469. [PMID: 36196920 PMCID: PMC9704522 DOI: 10.1042/bst20220648] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022]
Abstract
The AAA-ATPase (ATPases associated with diverse cellular activities) valosin-containing protein (VCP), is essential for many cellular pathways including but not limited to endoplasmic reticulum-associated degradation (ERAD), DNA damage responses, and cell cycle regulation. VCP primarily identifies ubiquitylated proteins in these pathways and mediates their unfolding and degradation by the 26S proteasome. This review summarizes recent research on VCP that has uncovered surprising new ways that this ATPase is regulated, new aspects of recognition of substrates and novel pathways and substrates that utilize its activity.
Collapse
Affiliation(s)
- Brittany A. Ahlstedt
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, U.S.A
| | - Rakesh Ganji
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, U.S.A
| | - Malavika Raman
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, U.S.A
| |
Collapse
|
23
|
Bordoni M, Pansarasa O, Scarian E, Cristofani R, Leone R, Fantini V, Garofalo M, Diamanti L, Bernuzzi S, Gagliardi S, Carelli S, Poletti A, Cereda C. Lysosomes Dysfunction Causes Mitophagy Impairment in PBMCs of Sporadic ALS Patients. Cells 2022; 11:cells11081272. [PMID: 35455952 PMCID: PMC9030813 DOI: 10.3390/cells11081272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 02/06/2023] Open
Abstract
Mitochondria alterations are present in tissues derived from patients and animal models, but no data are available for peripheral blood mononuclear cells (PBMCs) of ALS patients. This work aims to investigate mitophagy in PBMCs of sporadic (sALS) patients and how this pathway can be tuned by using small molecules. We found the presence of morphologically atypical mitochondria by TEM and morphological abnormalities by MitoTracker™. We found a decreased number of healthy mitochondria in sALS PBMCs and an impairment of mitophagy with western blot and immunofluorescence. After rapamycin treatment, we found a higher increase in the LC3 marker in sALS PBMCs, while after NH4Cl treatment, we found a lower increase in the LC3 marker. Finally, mTOR-independent autophagy induction with trehalose resulted in a significant decrease in the lysosomes level sALS PBMCs. Our data suggest that the presence of morphologically altered mitochondria and an inefficient turnover of damaged mitochondria in PBMCs of sALS patients rely on the impairment of the mitophagy pathway. We also found that the induction of the mTOR-independent autophagy pathway leads to a decrease in lysosomes level, suggesting a more sensitivity of sALS PBMCs to trehalose. Such evidence suggests that trehalose could represent an effective treatment for ALS patients.
Collapse
Affiliation(s)
- Matteo Bordoni
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Orietta Pansarasa
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
- Correspondence: ; Tel.: +0382-380-248
| | - Eveljn Scarian
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Università Degli Studi di Milano, 20133 Milano, Italy; (R.C.); (A.P.)
| | | | - Valentina Fantini
- Laboratory of Neurobiology and Neurogenetic, Golgi-Cenci Foundation, 20081 Abbiategrasso, Italy;
| | - Maria Garofalo
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Luca Diamanti
- Neuroncology Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy;
| | - Stefano Bernuzzi
- Immunohematological and Transfusional Service and Centre of Transplantation Immunology, IRCCS “San Matteo Foundation”, 27100 Pavia, Italy;
| | - Stella Gagliardi
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Centre Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, 20157 Milan, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Università Degli Studi di Milano, 20133 Milano, Italy; (R.C.); (A.P.)
| | - Cristina Cereda
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| |
Collapse
|