1
|
Nehmeh B, Rebehmed J, Nehmeh R, Taleb R, Akoury E. Unlocking therapeutic frontiers: harnessing artificial intelligence in drug discovery for neurodegenerative diseases. Drug Discov Today 2024; 29:104216. [PMID: 39428082 DOI: 10.1016/j.drudis.2024.104216] [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: 07/04/2024] [Revised: 10/05/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Neurodegenerative diseases (NDs) pose serious healthcare challenges with limited therapeutic treatments and high social burdens. The integration of artificial intelligence (AI) into drug discovery has emerged as a promising approach to address these challenges. This review explores the application of AI techniques to unravel therapeutic frontiers for NDs. We examine the current landscape of AI-driven drug discovery and discuss the potentials of AI in accelerating the identification of novel therapeutic targets on ND research and drug development, optimization of drug candidates, and expediating personalized medicine approaches. Finally, we outline future directions and challenges in harnessing AI for the advancement of therapeutics in this critical area by emphasizing the importance of interdisciplinary collaboration and ethical considerations.
Collapse
Affiliation(s)
- Bilal Nehmeh
- Department of Physical Sciences, Lebanese American University, Beirut 1102-2801, Lebanon
| | - Joseph Rebehmed
- Department of Computer Science and Mathematics, Lebanese American University, Beirut 1102-2801, Lebanon
| | - Riham Nehmeh
- INSA Rennes, Institut d'électronique et de Télécommunications de Rennes IETR, UMR 6164, 35708 Rennes, France
| | - Robin Taleb
- Department of Physical Sciences, Lebanese American University, Byblos Campus, Blat, 4M8F+6QF, Lebanon
| | - Elias Akoury
- Department of Physical Sciences, Lebanese American University, Beirut 1102-2801, Lebanon.
| |
Collapse
|
2
|
Ho PC, Hsieh TC, Tsai KJ. TDP-43 proteinopathy in frontotemporal lobar degeneration and amyotrophic lateral sclerosis: From pathomechanisms to therapeutic strategies. Ageing Res Rev 2024; 100:102441. [PMID: 39069095 DOI: 10.1016/j.arr.2024.102441] [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: 05/31/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Proteostasis failure is a common pathological characteristic in neurodegenerative diseases. Revitalizing clearance systems could effectively mitigate these diseases. The transactivation response (TAR) DNA-binding protein 43 (TDP-43) plays a critical role as an RNA/DNA-binding protein in RNA metabolism and synaptic function. Accumulation of TDP-43 aggregates in the central nervous system is a hallmark of frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Autophagy, a major and highly conserved degradation pathway, holds the potential for degrading aggregated TDP-43 and alleviating FTLD/ALS. This review explores the causes of TDP-43 aggregation, FTLD/ALS-related genes, key autophagy factors, and autophagy-based therapeutic strategies targeting TDP-43 proteinopathy. Understanding the underlying pathological mechanisms of TDP-43 proteinopathy can facilitate therapeutic interventions.
Collapse
Affiliation(s)
- Pei-Chuan Ho
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Chi Hsieh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
3
|
Roychowdhury S, Joshi D, Singh VK, Faruq M, Das P. Genetic and in silico analysis of Indian sporadic young onset patient with amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:589-599. [PMID: 38450645 DOI: 10.1080/21678421.2024.2324896] [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: 12/16/2023] [Revised: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is an old onset devastating neurodegenerative disorder. Young-onset ALS cases especially sporadic ones who are between 25 and 45 years are rarely affected by the disease. Despite the identification of numerous candidate genes associated with ALS, the etiology of the disease remains elusive due to extreme genetic and phenotypic variability. The advent of affordable whole exome sequencing (WES) has opened new avenues for unraveling the disease's pathophysiology better. METHODS AND RESULTS We aimed to determine the genetic basis of an Indian-origin, young onset sporadic ALS patient with very rapid deterioration of the disease course without any cognitive decline who was screened for mutations in major ALS candidate genes by WES. Variants detected were reconfirmed by Sanger sequencing. The clinicopathological features were investigated and two heterozygous missense variants were identified: R452W, not previously associated with ALS, present in one of the four conserved C terminal domains in ANXA11 and R208W in SIGMAR1, respectively. Both of these variants were predicted to be damaging by pathogenicity prediction tools and various in silico methods. CONCLUSION Our study revealed two potentially pathogenic variants in two ALS candidate genes. The genetic makeup of ALS patients from India has been the subject of a few prior studies, but none of them examined ANXA11 and SIGMAR1 genes so far. These results establish the framework for additional research into the pathogenic processes behind these variations that result in sporadic ALS disease and further our understanding of the genetic makeup of Indian ALS patients.
Collapse
Affiliation(s)
- Saileyee Roychowdhury
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Deepika Joshi
- Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vinay Kumar Singh
- School of Biotechnology, Centre for Bioinformatics, Institute of Science, Banaras Hindu University, Varanasi, India, and
| | - Mohammed Faruq
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Parimal Das
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
4
|
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
|
5
|
Garcia-Montojo M, Fathi S, Rastegar C, Simula ER, Doucet-O'Hare T, Cheng YHH, Abrams RPM, Pasternack N, Malik N, Bachani M, Disanza B, Maric D, Lee MH, Wang H, Santamaria U, Li W, Sampson K, Lorenzo JR, Sanchez IE, Mezghrani A, Li Y, Sechi LA, Pineda S, Heiman M, Kellis M, Steiner J, Nath A. TDP-43 proteinopathy in ALS is triggered by loss of ASRGL1 and associated with HML-2 expression. Nat Commun 2024; 15:4163. [PMID: 38755145 PMCID: PMC11099023 DOI: 10.1038/s41467-024-48488-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
TAR DNA-binding protein 43 (TDP-43) proteinopathy in brain cells is the hallmark of amyotrophic lateral sclerosis (ALS) but its cause remains elusive. Asparaginase-like-1 protein (ASRGL1) cleaves isoaspartates, which alter protein folding and susceptibility to proteolysis. ASRGL1 gene harbors a copy of the human endogenous retrovirus HML-2, whose overexpression contributes to ALS pathogenesis. Here we show that ASRGL1 expression was diminished in ALS brain samples by RNA sequencing, immunohistochemistry, and western blotting. TDP-43 and ASRGL1 colocalized in neurons but, in the absence of ASRGL1, TDP-43 aggregated in the cytoplasm. TDP-43 was found to be prone to isoaspartate formation and a substrate for ASRGL1. ASRGL1 silencing triggered accumulation of misfolded, fragmented, phosphorylated and mislocalized TDP-43 in cultured neurons and motor cortex of female mice. Overexpression of ASRGL1 restored neuronal viability. Overexpression of HML-2 led to ASRGL1 silencing. Loss of ASRGL1 leading to TDP-43 aggregation may be a critical mechanism in ALS pathophysiology.
Collapse
Affiliation(s)
- Marta Garcia-Montojo
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Saeed Fathi
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Cyrus Rastegar
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Elena Rita Simula
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
- Struttura Complessa Microbiologia e Virologia, Azienda Ospedaliera Universitaria Sassari, Sassari, Italy
| | - Tara Doucet-O'Hare
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Y H Hank Cheng
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Rachel P M Abrams
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Nicholas Pasternack
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Nasir Malik
- Translational Neuroscience Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Muzna Bachani
- Translational Neuroscience Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Brianna Disanza
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Myoung-Hwa Lee
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Herui Wang
- Neuro-Oncology Branch, National Cancer Institute (NIH), Bethesda, MD, USA
| | - Ulisses Santamaria
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Wenxue Li
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kevon Sampson
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Juan Ramiro Lorenzo
- Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-CICPBA-UNCPBA, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro (FCV-UNCPBA), Tandil, Argentina
| | - Ignacio E Sanchez
- Protein Physiology Laboratory, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales and IQUIBICEN-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alexandre Mezghrani
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
- Centre de Biologie Structurale, Centre national de la recherche scientifique (CNRS), Montpellier, France
| | - Yan Li
- Protein/Peptide Sequencing Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Leonardo Antonio Sechi
- Struttura Complessa Microbiologia e Virologia, Azienda Ospedaliera Universitaria Sassari, Sassari, Italy
| | | | - Myriam Heiman
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Manolis Kellis
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joseph Steiner
- Translational Neuroscience Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
6
|
De Marchi F, Venkatesan S, Saraceno M, Mazzini L, Grossini E. Acetyl-L-carnitine and Amyotrophic Lateral Sclerosis: Current Evidence and Potential use. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:588-601. [PMID: 36998125 DOI: 10.2174/1871527322666230330083757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND The management of neurodegenerative diseases can be frustrating for clinicians, given the limited progress of conventional medicine in this context. AIM For this reason, a more comprehensive, integrative approach is urgently needed. Among various emerging focuses for intervention, the modulation of central nervous system energetics, oxidative stress, and inflammation is becoming more and more promising. METHODS In particular, electrons leakage involved in the mitochondrial energetics can generate reactive oxygen-free radical-related mitochondrial dysfunction that would contribute to the etiopathology of many disorders, such as Alzheimer's and other dementias, Parkinson's disease, multiple sclerosis, stroke, and amyotrophic lateral sclerosis (ALS). RESULTS In this context, using agents, like acetyl L-carnitine (ALCAR), provides mitochondrial support, reduces oxidative stress, and improves synaptic transmission. CONCLUSION This narrative review aims to update the existing literature on ALCAR molecular profile, tolerability, and translational clinical potential use in neurodegeneration, focusing on ALS.
Collapse
Affiliation(s)
- Fabiola De Marchi
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale 28100 Novara, Italy
| | - Sakthipriyan Venkatesan
- Laboratory of Physiology, Department of Translational Medicine, University of Piemonte Orientale 28100, Novara, Italy
| | - Massimo Saraceno
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale 28100 Novara, Italy
| | - Letizia Mazzini
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale 28100 Novara, Italy
| | - Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, University of Piemonte Orientale 28100, Novara, Italy
| |
Collapse
|
7
|
Zhou W, Xu R. Current insights in the molecular genetic pathogenesis of amyotrophic lateral sclerosis. Front Neurosci 2023; 17:1189470. [PMID: 37638324 PMCID: PMC10448825 DOI: 10.3389/fnins.2023.1189470] [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: 03/19/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and fatal neurodegenerative disease that leads to the massive loss of motor neurons in cerebrum, brain stem and spinal cord. It affects not only motor neurons but also other neurons and glial cells, resulting in the progressive muscle atrophy, the severe disability and the eventual death due to the respiratory failure. The pathogenesis of ALS is not fully understood. Currently, several factors are considered to be involved in the pathogenesis of ALS, such as genetic factors, imbalances in protein homeostasis, RNA metabolism disorders, mitochondrial dysfunctions, glutamate-mediated excitatory toxicities and intra-neuronal material transport disorders in neurons. The study of genetic mutations related to ALS pathogenesis will link the molecular and cellular mechanisms of the disease, thus enhancing the understanding of its occurrence and progression, thereby providing new insights for the pathogenesis of ALS. This review summarizes the current insights in the molecular genetic pathogenesis of ALS.
Collapse
Affiliation(s)
- Wan Zhou
- Medical College of Nanchang University, Nanchang, China
- Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, The Clinical College of Nanchang Medical College, Nanchang, China
| | - Renshi Xu
- Medical College of Nanchang University, Nanchang, China
- Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, The Clinical College of Nanchang Medical College, Nanchang, China
| |
Collapse
|
8
|
De Marchi F, Franjkic T, Schito P, Russo T, Nimac J, Chami AA, Mele A, Vidatic L, Kriz J, Julien JP, Apic G, Russell RB, Rogelj B, Cannon JR, Baralle M, Agosta F, Hecimovic S, Mazzini L, Buratti E, Munitic I. Emerging Trends in the Field of Inflammation and Proteinopathy in ALS/FTD Spectrum Disorder. Biomedicines 2023; 11:1599. [PMID: 37371694 PMCID: PMC10295684 DOI: 10.3390/biomedicines11061599] [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: 04/28/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Proteinopathy and neuroinflammation are two main hallmarks of neurodegenerative diseases. They also represent rare common events in an exceptionally broad landscape of genetic, environmental, neuropathologic, and clinical heterogeneity present in patients. Here, we aim to recount the emerging trends in amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD) spectrum disorder. Our review will predominantly focus on neuroinflammation and systemic immune imbalance in ALS and FTD, which have recently been highlighted as novel therapeutic targets. A common mechanism of most ALS and ~50% of FTD patients is dysregulation of TAR DNA-binding protein 43 (TDP-43), an RNA/DNA-binding protein, which becomes depleted from the nucleus and forms cytoplasmic aggregates in neurons and glia. This, in turn, via both gain and loss of function events, alters a variety of TDP-43-mediated cellular events. Experimental attempts to target TDP-43 aggregates or manipulate crosstalk in the context of inflammation will be discussed. Targeting inflammation, and the immune system in general, is of particular interest because of the high plasticity of immune cells compared to neurons.
Collapse
Affiliation(s)
- Fabiola De Marchi
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, 28100 Novara, Italy; (F.D.M.); (A.M.)
| | - Toni Franjkic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia;
- Metisox, Cambridge CB24 9NL, UK;
| | - Paride Schito
- Department of Neurology & Neuropathology Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (P.S.); (T.R.)
| | - Tommaso Russo
- Department of Neurology & Neuropathology Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (P.S.); (T.R.)
| | - Jerneja Nimac
- Department of Biotechnology, Jozef Stefan Institute, SI-1000 Ljubljana, Slovenia; (J.N.); (B.R.)
- Graduate School of Biomedicine, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Anna A. Chami
- CERVO Research Centre, Laval University, Quebec City, QC G1J 2G3, Canada; (A.A.C.); (J.K.); (J.-P.J.)
| | - Angelica Mele
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, 28100 Novara, Italy; (F.D.M.); (A.M.)
| | - Lea Vidatic
- Laboratory for Neurodegenerative Disease Research, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia; (L.V.); (S.H.)
| | - Jasna Kriz
- CERVO Research Centre, Laval University, Quebec City, QC G1J 2G3, Canada; (A.A.C.); (J.K.); (J.-P.J.)
| | - Jean-Pierre Julien
- CERVO Research Centre, Laval University, Quebec City, QC G1J 2G3, Canada; (A.A.C.); (J.K.); (J.-P.J.)
| | | | | | - Boris Rogelj
- Department of Biotechnology, Jozef Stefan Institute, SI-1000 Ljubljana, Slovenia; (J.N.); (B.R.)
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Jason R. Cannon
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA;
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | | | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Silva Hecimovic
- Laboratory for Neurodegenerative Disease Research, Division of Molecular Medicine, Ruder Boskovic Institute, 10000 Zagreb, Croatia; (L.V.); (S.H.)
| | - Letizia Mazzini
- Department of Neurology and ALS Centre, University of Piemonte Orientale, Maggiore Della Carità Hospital, Corso Mazzini 18, 28100 Novara, Italy; (F.D.M.); (A.M.)
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149 Trieste, Italy
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia;
| |
Collapse
|
9
|
Hosomi A, Okachi C, Fujiwara Y. Human SOD1 is secreted via a conventional secretion pathway in Saccharomyces cerevisiae. Biochem Biophys Res Commun 2023; 666:101-106. [PMID: 37182284 DOI: 10.1016/j.bbrc.2023.05.022] [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: 04/21/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 05/16/2023]
Abstract
Soluble proteins sorted through the secretory pathway contain an N-terminal signal peptide that induces their translocation into the endoplasmic reticulum (ER) from the cytosol. However, a few proteins that lack a signal peptide are still translocated into the ER, such as SOD1. SOD1 is a causative gene of amyotrophic lateral sclerosis (ALS). A relationship has been suggested between the secretion of SOD1 and the pathogenesis of ALS; however, the transport mechanism of SOD1 remains unclear. We herein report that SOD1 was translocated into the ER lumen through the translocon Sec61 and was then secreted extracellularly. The present results indicate the potential of suppressing the secretion of SOD1 as a therapeutic target for ALS.
Collapse
Affiliation(s)
- Akira Hosomi
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano, 399-4598, Japan; Faculty of Agriculture, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano, 399-4598, Japan.
| | - Chinatsu Okachi
- Faculty of Agriculture, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano, 399-4598, Japan
| | - Yudai Fujiwara
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano, 399-4598, Japan
| |
Collapse
|
10
|
Gao T, Huo J, Xin C, Yang J, Liu Q, Dong H, Li R, Liu Y. Protective effects of intrathecal injection of AAV9-RabGGTB-GFP+ in SOD1G93A mice. Front Aging Neurosci 2023; 15:1092607. [PMID: 36967828 PMCID: PMC10036913 DOI: 10.3389/fnagi.2023.1092607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/24/2023] [Indexed: 03/12/2023] Open
Abstract
IntroductionAmyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that widely affects motor neurons of the CNS. About 20% of patients with ALS have familial ALS (fALS). One of the classic models of ALS are SOD1G93A mice. Misfolded SOD1 protein can be overexpressed in motor neurons, which results in progressive paralysis of the limbs of mice. There is still no effective treatment for ALS. In recent years, the treatment of ALS by regulating autophagy has become a research hotspot. Autophagy obstacles have been confirmed to be one of the early pathological events of ALS. Rab7 is a member of the Ras superfamily and plays a key role in the late stage of autophagy. In our previous studies, we found that prenoylation of Rab7 was inhibited in the ALS model. Prenylation is a post-translational modification in which farnesyl or geranylgeranyl groups are covalently linked to target proteins. Based on these findings, we proposed the novel idea that the regulation of RabGGTB (the β-subunit of RabGGTase) mediated prenylation modification of Rab7, and that this can be used as a prevention and treatment of ALS associated with abnormal protein accumulation.MethodsIn the present study, RabGGTB was overexpressed in mouse spinal cord motoneurons by using adeno-associated virus as vector. Then immunofluorescence quantitative analysis was used for pathological study. The body weight, footprint analysis, the accelerating rotarod test, and neurological deficits score were used to evaluate animal behavior.ResultsOur results show that the protein level of RabGGTB was significantly increased in the lumbar and thoracic regions of spinal cord motoneurons of injected mice. Furthermore, the onset time and survival time of SOD1G93A mice injected with AAV9-RabGGTB-GFP+ were delayed compared with those of mice without overexpression. At the same time, we also observed a decrease in SOD1 misfolded and glial overactivation in the lumbar spinal cord of these SOD1G93A mice.ConclusionThe findings reported here show that RabGGTB plays a significant role in the pathogenesis of SOD1G93A mice and with great therapeutic potential for reducing abnormal aggregation of SOD1 in ALS.
Collapse
Affiliation(s)
- Tianchu Gao
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Jia Huo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Cheng Xin
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Jing Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Qi Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Hui Dong
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Rui Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
- *Correspondence: Rui Li, ; Yaling Liu,
| | - Yaling Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
- *Correspondence: Rui Li, ; Yaling Liu,
| |
Collapse
|
11
|
Haouari S, Andres CR, Lanznaster D, Marouillat S, Brulard C, Dangoumau A, Ung D, Veyrat-Durebex C, Laumonnier F, Blasco H, Couratier P, Corcia P, Vourc’h P. Study of Ubiquitin Pathway Genes in a French Population with Amyotrophic Lateral Sclerosis: Focus on HECW1 Encoding the E3 Ligase NEDL1. Int J Mol Sci 2023; 24:ijms24021268. [PMID: 36674783 PMCID: PMC9867363 DOI: 10.3390/ijms24021268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 01/02/2023] [Indexed: 01/10/2023] Open
Abstract
The ubiquitin pathway, one of the main actors regulating cell signaling processes and cellular protein homeostasis, is directly involved in the pathophysiology of amyotrophic lateral sclerosis (ALS). We first analyzed, by a next-generation sequencing (NGS) strategy, a series of genes of the ubiquitin pathway in two cohorts of familial and sporadic ALS patients comprising 176 ALS patients. We identified several pathogenic variants in different genes of this ubiquitin pathway already described in ALS, such as FUS, CCNF and UBQLN2. Other variants of interest were discovered in new genes studied in this disease, in particular in the HECW1 gene. We have shown that the HECT E3 ligase called NEDL1, encoded by the HECW1 gene, is expressed in neurons, mainly in their somas. Its overexpression is associated with increased cell death in vitro and, very interestingly, with the cytoplasmic mislocalization of TDP-43, a major protein involved in ALS. These results give new support for the role of the ubiquitin pathway in ALS, and suggest further studies of the HECW1 gene and its protein NEDL1 in the pathophysiology of ALS.
Collapse
Affiliation(s)
- Shanez Haouari
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Christian Robert Andres
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 2 Boulevard Tonnellé, 37044 Tours, France
| | - Debora Lanznaster
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Sylviane Marouillat
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Céline Brulard
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Audrey Dangoumau
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Devina Ung
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Charlotte Veyrat-Durebex
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 2 Boulevard Tonnellé, 37044 Tours, France
| | - Frédéric Laumonnier
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Hélène Blasco
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 2 Boulevard Tonnellé, 37044 Tours, France
| | - Philippe Couratier
- Centre SLA, CHU Limoges, 2 Avenue Martin Luther King, 87000 Limoges, France
| | - Philippe Corcia
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
- Centre SLA, CHU Tours, 2 Boulevard Tonnellé, 37044 Tours, France
| | - Patrick Vourc’h
- UMR 1253 iBrain, Université de Tours, Inserm, 10 Boulevard Tonnellé, 37032 Tours, France
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 2 Boulevard Tonnellé, 37044 Tours, France
- Correspondence: ; Tel.: +33-234378910
| |
Collapse
|
12
|
Quach TT, Stratton HJ, Khanna R, Mackey-Alfonso S, Deems N, Honnorat J, Meyer K, Duchemin AM. Neurodegenerative Diseases: From Dysproteostasis, Altered Calcium Signalosome to Selective Neuronal Vulnerability to AAV-Mediated Gene Therapy. Int J Mol Sci 2022; 23:ijms232214188. [PMID: 36430666 PMCID: PMC9694178 DOI: 10.3390/ijms232214188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Despite intense research into the multifaceted etiology of neurodegenerative diseases (ND), they remain incurable. Here we provide a brief overview of several major ND and explore novel therapeutic approaches. Although the cause (s) of ND are not fully understood, the accumulation of misfolded/aggregated proteins in the brain is a common pathological feature. This aggregation may initiate disruption of Ca++ signaling, which is an early pathological event leading to altered dendritic structure, neuronal dysfunction, and cell death. Presently, ND gene therapies remain unidimensional, elusive, and limited to modifying one pathological feature while ignoring others. Considering the complexity of signaling cascades in ND, we discuss emerging therapeutic concepts and suggest that deciphering the molecular mechanisms involved in dendritic pathology may broaden the phenotypic spectrum of ND treatment. An innovative multiplexed gene transfer strategy that employs silencing and/or over-expressing multiple effectors could preserve vulnerable neurons before they are lost. Such therapeutic approaches may extend brain health span and ameliorate burdensome chronic disease states.
Collapse
Affiliation(s)
- Tam T. Quach
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- INSERM U1217/CNRS UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, 69677 Lyon, France
| | | | - Rajesh Khanna
- Department of Molecular Pathobiology, New York University, New York, NY 10010, USA
| | - Sabrina Mackey-Alfonso
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Nicolas Deems
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jérome Honnorat
- INSERM U1217/CNRS UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, 69677 Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 69677 Lyon, France
- SynatAc Team, Institut NeuroMyoGène, 69677 Lyon, France
| | - Kathrin Meyer
- The Research Institute of Nationwide Children Hospital, Columbus, OH 43205, USA
- Department of Pediatric, The Ohio State University, Columbus, OH 43210, USA
| | - Anne-Marie Duchemin
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: ; Tel.: +1-614-293-5517; Fax: +1-614-293-7599
| |
Collapse
|
13
|
Imamura K, Izumi Y, Nagai M, Nishiyama K, Watanabe Y, Hanajima R, Egawa N, Ayaki T, Oki R, Fujita K, Uozumi R, Morinaga A, Hirohashi T, Fujii Y, Yamamoto T, Tatebe H, Tokuda T, Takahashi N, Morita S, Takahashi R, Inoue H. Safety and tolerability of bosutinib in patients with amyotrophic lateral sclerosis (iDReAM study): A multicentre, open-label, dose-escalation phase 1 trial. EClinicalMedicine 2022; 53:101707. [PMID: 36467452 PMCID: PMC9716331 DOI: 10.1016/j.eclinm.2022.101707] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease caused by the loss of motor neurons, and development of effective medicines is urgently required. Induced pluripotent stem cell (iPSC)-based drug repurposing identified the Src/c-Abl inhibitor bosutinib, which is approved for the treatment of chronic myelogenous leukemia (CML), as a candidate for the molecular targeted therapy of ALS. METHODS An open-label, multicentre, dose-escalation phase 1 study using a 3 + 3 design was conducted in 4 hospitals in Japan to evaluate the safety and tolerability of bosutinib in patients with ALS. Furthermore, the exploratory efficacy was evaluated using Revised ALS Functional Rating Scale (ALSFRS-R), predictive biomarkers including plasma neurofilament light chain (NFL) were explored, and single-cell RNA sequencing of iPSC-derived motor neurons was conducted. Patients, whose total ALSFRS-R scores decreased by 1-3 points during the 12-week, received escalating doses starting from 100 mg quaque die (QD) up to 400 mg QD based on dose-limiting toxicity (DLT) occurrence, and all participants who received one dose of the study drug were included in the primary analysis. This trial is registered with ClinicalTrials.gov, NCT04744532, as Induced pluripotent stem cell-based Drug Repurposing for Amyotrophic Lateral Sclerosis Medicine (iDReAM) study. FINDINGS Between March 29, 2019 and May 7, 2021, 20 patients were enrolled, 13 of whom received bosutinib treatment and 12 were included in the safety and efficacy analyses. No DLTs were observed up to 300 mg QD, but DLTs were observed in 3/3 patients of the 400 mg QD cohort. In all patients receiving 100 mg-400 mg, the prevalent adverse events (AEs) were gastrointestinal AEs in 12 patients (92.3%), liver function related AEs in 7 patients (53.8%), and rash in 3 patients (23.1%). The safety profile was consistent with that known for CML treatment, and ALS-specific AEs were not observed. A subset of patients (5/9 patients) was found to respond well to bosutinib treatment over the 12-week treatment period. It was found that the treatment-responsive patients could be distinguished by their lower levels of plasma NFL. Furthermore, single-cell RNA sequencing of iPSC-derived motor neurons revealed the pathogenesis related molecular signature in patients with ALS showing responsiveness to bosutinib. INTERPRETATION This is the first trial of a Src/c-Abl inhibitor, bosutinib, for patients with ALS. The safety and tolerability of bosutinib up to 300 mg, not 400 mg, in ALS were described, and responsiveness of patients on motor function was observed. Since this was an open-label trial within a short period with a limited number of patients, further clinical trials will be required. FUNDING AMED and iPS Cell Research Fund.
Collapse
Affiliation(s)
- Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Makiko Nagai
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazutoshi Nishiyama
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yasuhiro Watanabe
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Naohiro Egawa
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryosuke Oki
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koji Fujita
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Ryuji Uozumi
- Department of Biomedical Statistics and Bioinformatics, Kyoto University, Kyoto, Japan
| | | | | | | | - Takuya Yamamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Harutsugu Tatebe
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Takahiko Tokuda
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Corresponding author. 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, Kyoto Pref., 606-8507, Japan.
| |
Collapse
|
14
|
Kanekura K, Kuroda M. How can we interpret the relationship between liquid-liquid phase separation and amyotrophic lateral sclerosis? J Transl Med 2022; 102:912-918. [PMID: 36775420 DOI: 10.1038/s41374-022-00791-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/12/2022] [Indexed: 01/08/2023] Open
Abstract
One of the critical definitions of neurodegenerative diseases is the formation of insoluble intracellular inclusion body. These inclusions are found in various neurodegenerative diseases such as Alzheimer's disease, amyotrophic lateral sclerosis (ALS), Huntington's disease, Parkinson's disease, and frontotemporal dementia (FTD). Each inclusion body contains disease-specific proteins and is also resistant to common detergent treatments. These aggregates are generally ubiquitinated and thus recognized as misfolded by the organism. They are observed in residual neurons at the affected sites in each disease, suggesting a contribution to disease pathogenesis. The molecular mechanisms for the formation of these inclusion bodies remain unclear. Some proteins, such as superoxide dismutase 1 (SOD1) mutant that causes familial ALS, are highly aggregative due to altered folding caused by point mutations. Still, the aggregates observed in neurodegenerative diseases contain wild-type proteins. In recent years, it has been reported that the proteins responsible for neurodegenerative diseases undergo liquid-liquid phase separation (LLPS). In particular, the ALS/FTD causative proteins such as TAR DNA-binding protein 43 kDa (TDP-43) and fused-in-sarcoma (FUS) undergo LLPS. LLPS increases the local concentration of these proteins, and these proteins eventually change their phase from liquid to solid (liquid-solid phase transition) due to abnormal folding during repetitive separation cycles into two phases and recovery to one phase. In addition to the inclusion body formation, sequestration of essential proteins into the LLPS droplets or changes in the LLPS status can directly impair neural functions and cause diseases. In this review, we will discuss the relationship between the LLPS observed in ALS causative proteins and the pathogenesis of the disease and outline potential therapeutic approaches.
Collapse
Affiliation(s)
- Kohsuke Kanekura
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan.
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| |
Collapse
|
15
|
Papendorf JJ, Krüger E, Ebstein F. Proteostasis Perturbations and Their Roles in Causing Sterile Inflammation and Autoinflammatory Diseases. Cells 2022; 11:cells11091422. [PMID: 35563729 PMCID: PMC9103147 DOI: 10.3390/cells11091422] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/04/2022] [Accepted: 04/14/2022] [Indexed: 12/17/2022] Open
Abstract
Proteostasis, a portmanteau of the words protein and homeostasis, refers to the ability of eukaryotic cells to maintain a stable proteome by acting on protein synthesis, quality control and/or degradation. Over the last two decades, an increasing number of disorders caused by proteostasis perturbations have been identified. Depending on their molecular etiology, such diseases may be classified into ribosomopathies, proteinopathies and proteasomopathies. Strikingly, most—if not all—of these syndromes exhibit an autoinflammatory component, implying a direct cause-and-effect relationship between proteostasis disruption and the initiation of innate immune responses. In this review, we provide a comprehensive overview of the molecular pathogenesis of these disorders and summarize current knowledge of the various mechanisms by which impaired proteostasis promotes autoinflammation. We particularly focus our discussion on the notion of how cells sense and integrate proteostasis perturbations as danger signals in the context of autoinflammatory diseases to provide insights into the complex and multiple facets of sterile inflammation.
Collapse
|
16
|
The pathogenesis of amyotrophic lateral sclerosis: Mitochondrial dysfunction, protein misfolding and epigenetics. Brain Res 2022; 1786:147904. [PMID: 35390335 DOI: 10.1016/j.brainres.2022.147904] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/24/2022] [Accepted: 04/01/2022] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease with multiple complex mechanisms involved. Among them, mitochondrial dysfunction plays an important role in ALS. Multiple studies have shown that mitochondria are closely associated with reactive oxygen species production and oxidative stress and exhibit different functional states in different genetic backgrounds. In this review we explored the roles of Ca2+, autophagy, mitochondrial quality control in the regulation of mitochondrial homeostasis and their relationship with ALS. In addition, we also summarized and analyzed the roles of protein misfolding and abnormal aggregation in the pathogenesis of ALS. Moreover, we also discussed how epigenetic mechanisms such as DNA methylation and protein post-translational modification affect initiation and progression of ALS. Nevertheless, existing events still cannot fully explain the pathogenesis of ALS at present, more studies are required to explore pathological mechanisms of ALS.
Collapse
|
17
|
Cai Q, Ganesan D. Regulation of neuronal autophagy and the implications in neurodegenerative diseases. Neurobiol Dis 2022; 162:105582. [PMID: 34890791 PMCID: PMC8764935 DOI: 10.1016/j.nbd.2021.105582] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/22/2021] [Accepted: 12/06/2021] [Indexed: 01/03/2023] Open
Abstract
Neurons are highly polarized and post-mitotic cells with the specific requirements of neurotransmission accompanied by high metabolic demands that create a unique challenge for the maintenance of cellular homeostasis. Thus, neurons rely heavily on autophagy that constitutes a key quality control system by which dysfunctional cytoplasmic components, protein aggregates, and damaged organelles are sequestered within autophagosomes and then delivered to the lysosome for degradation. While mature lysosomes are predominantly located in the soma of neurons, the robust, constitutive biogenesis of autophagosomes occurs in the synaptic terminal via a conserved pathway that is required to maintain synaptic integrity and function. Following formation, autophagosomes fuse with late endosomes and then are rapidly and efficiently transported by the microtubule-based cytoplasmic dynein motor along the axon toward the soma for lysosomal clearance. In this review, we highlight the recent knowledge of the roles of autophagy in neuronal health and disease. We summarize the available evidence about the normal functions of autophagy as a protective factor against neurodegeneration and discuss the mechanism underlying neuronal autophagy regulation. Finally, we describe how autophagy function is affected in major neurodegenerative diseases with a special focus on Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis.
Collapse
|
18
|
Chen C, Yamanaka Y, Ueda K, Li P, Miyagi T, Harada Y, Tezuka S, Narumi S, Sugimoto M, Kuroda M, Hayamizu Y, Kanekura K. Phase separation and toxicity of C9orf72 poly(PR) depends on alternate distribution of arginine. J Cell Biol 2021; 220:212626. [PMID: 34499080 PMCID: PMC8438627 DOI: 10.1083/jcb.202103160] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Arg (R)-rich dipeptide repeat proteins (DPRs; poly(PR): Pro-Arg and poly(GR): Gly-Arg), encoded by a hexanucleotide expansion in the C9ORF72 gene, induce neurodegeneration in amyotrophic lateral sclerosis (ALS). Although R-rich DPRs undergo liquid-liquid phase separation (LLPS), which affects multiple biological processes, mechanisms underlying LLPS of DPRs remain elusive. Here, using in silico, in vitro, and in cellulo methods, we determined that the distribution of charged Arg residues regulates the complex coacervation with anionic peptides and nucleic acids. Proteomic analyses revealed that alternate Arg distribution in poly(PR) facilitates entrapment of proteins with acidic motifs via LLPS. Transcription, translation, and diffusion of nucleolar nucleophosmin (NPM1) were impaired by poly(PR) with an alternate charge distribution but not by poly(PR) variants with a consecutive charge distribution. We propose that the pathogenicity of R-rich DPRs is mediated by disturbance of proteins through entrapment in the phase-separated droplets via sequence-controlled multivalent protein-protein interactions.
Collapse
Affiliation(s)
- Chen Chen
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Yoshiaki Yamanaka
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Koji Ueda
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Peiying Li
- Center for Biosystems Dynamics Research, RIKEN, Kanagawa, Japan
| | - Tamami Miyagi
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Yuichiro Harada
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Sayaka Tezuka
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Masahiro Sugimoto
- Research and Development Center for Minimally Invasive Therapies, Tokyo Medical University, Tokyo, Japan
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Yuhei Hayamizu
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Kohsuke Kanekura
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
19
|
Yamanaka Y, Miyagi T, Harada Y, Kuroda M, Kanekura K. Establishment of chemically oligomerizable TAR DNA-binding protein-43 which mimics amyotrophic lateral sclerosis pathology in mammalian cells. J Transl Med 2021; 101:1331-1340. [PMID: 34131277 DOI: 10.1038/s41374-021-00623-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 11/09/2022] Open
Abstract
One of the pathological hallmarks of amyotrophic lateral sclerosis (ALS) is mislocalized, cytosolic aggregation of TAR DNA-Binding Protein-43 (TDP-43). Not only TDP-43 per se is a causative gene of ALS but also mislocalization and aggregation of TDP-43 seems to be a common pathological change in both sporadic and familial ALS. The mechanism how nuclear TDP-43 transforms into cytosolic aggregates remains elusive, but recent studies using optogenetics have proposed that aberrant liquid-liquid phase separation (LLPS) of TDP-43 links to the aggregation process, leading to cytosolic distribution. Although LLPS plays an important role in the aggregate formation, there are still several technical problems in the optogenetic technique to be solved to progress further in vivo study. Here we report a chemically oligomerizable TDP-43 system. Oligomerization of TDP-43 was achieved by a small compound AP20187, and oligomerized TDP-43 underwent aggregate formation, followed by cytosolic mislocalization and induction of cell toxicity. The mislocalized TDP-43 co-aggregated with wt-TDP-43, Fused-in-sarcoma (FUS), TIA1 and sequestosome 1 (SQSTM1)/p62, mimicking ALS pathology. The chemically oligomerizable TDP-43 also revealed the roles of the N-terminal domain, RNA-recognition motif, nuclear export signal and low complexity domain in the aggregate formation and mislocalization of TDP-43. The aggregate-prone properties of TDP-43 were enhanced by a familial ALS-causative mutation. In conclusion, the chemically oligomerizable TDP-43 system could be useful to study the mechanisms underlying the droplet-aggregation phase transition and cytosolic mislocalization of TDP-43 in ALS and further study in vivo.
Collapse
Affiliation(s)
- Yoshiaki Yamanaka
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Tamami Miyagi
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Yuichiro Harada
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan.
| | - Kohsuke Kanekura
- Department of Molecular Pathology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan.
| |
Collapse
|
20
|
Ammal Kaidery N, Ahuja M, Sharma SM, Thomas B. An Emerging Role of miRNAs in Neurodegenerative Diseases: Mechanisms and Perspectives on miR146a. Antioxid Redox Signal 2021; 35:580-594. [PMID: 33403895 PMCID: PMC8388248 DOI: 10.1089/ars.2020.8256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: Advancements in and access to health care have led to unprecedented improvements in the quality of life and increased lifespan of human beings in the past century. However, aging is a significant risk factor for neurodegenerative diseases (NDs). Hence, improved life expectancy has led to an increased incidence of NDs. Despite intense research, effective treatments for NDs remain elusive. The future of neurotherapeutics development depends on effective disease modification strategies centered on carefully scrutinized targets. Recent Advances: As a promising new direction, recent evidence has demonstrated that epigenetic processes modify diverse biochemical pathways, including those related to NDs. Small non-coding RNAs, known as microRNAs (miRNAs), are components of the epigenetic system that alter the expression of target genes at the post-transcriptional level. Critical Issues: miRNAs are expressed abundantly in the central nervous system and are critical for the normal functioning and survival of neurons. Here, we review recent advances in elucidating miRNAs' roles in NDs and discuss their potential as therapeutic targets. In particular, neuroinflammation is a major pathological hallmark of NDs and miR146a is a crucial regulator of inflammation. Future Directions: Finally, we explore the possibilities of developing miR146a as a potential biomarker and therapeutic target where additional research may help facilitate the detection and amelioration of neuroinflammation in NDs. Antioxid. Redox Signal. 35, 580-594.
Collapse
Affiliation(s)
- Navneet Ammal Kaidery
- Darby Children's Research Institute, Departments of Medical University of South Carolina, Charleston, South Carolina, USA.,Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Manuj Ahuja
- Darby Children's Research Institute, Departments of Medical University of South Carolina, Charleston, South Carolina, USA.,Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sudarshana M Sharma
- Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA.,Hollings Cancer Center, and Departments of Medical University of South Carolina, Charleston, South Carolina, USA
| | - Bobby Thomas
- Darby Children's Research Institute, Departments of Medical University of South Carolina, Charleston, South Carolina, USA.,Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA.,Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA.,Drug Discovery, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
21
|
Kok JR, Palminha NM, Dos Santos Souza C, El-Khamisy SF, Ferraiuolo L. DNA damage as a mechanism of neurodegeneration in ALS and a contributor to astrocyte toxicity. Cell Mol Life Sci 2021; 78:5707-5729. [PMID: 34173837 PMCID: PMC8316199 DOI: 10.1007/s00018-021-03872-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/27/2021] [Accepted: 06/05/2021] [Indexed: 12/11/2022]
Abstract
Increasing evidence supports the involvement of DNA damage in several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Elevated levels of DNA damage are consistently observed in both sporadic and familial forms of ALS and may also play a role in Western Pacific ALS, which is thought to have an environmental cause. The cause of DNA damage in ALS remains unclear but likely differs between genetic subgroups. Repeat expansion in the C9ORF72 gene is the most common genetic cause of familial ALS and responsible for about 10% of sporadic cases. These genetic mutations are known to cause R-loops, thus increasing genomic instability and DNA damage, and generate dipeptide repeat proteins, which have been shown to lead to DNA damage and impairment of the DNA damage response. Similarly, several genes associated with ALS including TARDBP, FUS, NEK1, SQSTM1 and SETX are known to play a role in DNA repair and the DNA damage response, and thus may contribute to neuronal death via these pathways. Another consistent feature present in both sporadic and familial ALS is the ability of astrocytes to induce motor neuron death, although the factors causing this toxicity remain largely unknown. In this review, we summarise the evidence for DNA damage playing a causative or secondary role in the pathogenesis of ALS as well as discuss the possible mechanisms involved in different genetic subtypes with particular focus on the role of astrocytes initiating or perpetuating DNA damage in neurons.
Collapse
Affiliation(s)
- Jannigje Rachel Kok
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Sheffield, UK
| | - Nelma M Palminha
- Department of Molecular Biology and Biotechnology, The Healthy Lifespan Institute, Sheffield, UK
- The Institute of Neuroscience, University of Sheffield, Sheffield, UK
| | - Cleide Dos Santos Souza
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Sheffield, UK
| | - Sherif F El-Khamisy
- Department of Molecular Biology and Biotechnology, The Healthy Lifespan Institute, Sheffield, UK.
- The Institute of Neuroscience, University of Sheffield, Sheffield, UK.
- The Institute of Cancer Therapeutics, West Yorkshire, UK.
| | - Laura Ferraiuolo
- University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), Sheffield, UK.
- The Institute of Neuroscience, University of Sheffield, Sheffield, UK.
| |
Collapse
|
22
|
Salucci S, Bartoletti Stella A, Battistelli M, Burattini S, Bavelloni A, Cocco LI, Gobbi P, Faenza I. How Inflammation Pathways Contribute to Cell Death in Neuro-Muscular Disorders. Biomolecules 2021; 11:1109. [PMID: 34439778 PMCID: PMC8391499 DOI: 10.3390/biom11081109] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
Neuro-muscular disorders include a variety of diseases induced by genetic mutations resulting in muscle weakness and waste, swallowing and breathing difficulties. However, muscle alterations and nerve depletions involve specific molecular and cellular mechanisms which lead to the loss of motor-nerve or skeletal-muscle function, often due to an excessive cell death. Morphological and molecular studies demonstrated that a high number of these disorders seem characterized by an upregulated apoptosis which significantly contributes to the pathology. Cell death involvement is the consequence of some cellular processes that occur during diseases, including mitochondrial dysfunction, protein aggregation, free radical generation, excitotoxicity and inflammation. The latter represents an important mediator of disease progression, which, in the central nervous system, is known as neuroinflammation, characterized by reactive microglia and astroglia, as well the infiltration of peripheral monocytes and lymphocytes. Some of the mechanisms underlying inflammation have been linked to reactive oxygen species accumulation, which trigger mitochondrial genomic and respiratory chain instability, autophagy impairment and finally neuron or muscle cell death. This review discusses the main inflammatory pathways contributing to cell death in neuro-muscular disorders by highlighting the main mechanisms, the knowledge of which appears essential in developing therapeutic strategies to prevent the consequent neuron loss and muscle wasting.
Collapse
Affiliation(s)
- Sara Salucci
- Department of Biomolecular Sciences (DiSB), Urbino University Carlo Bo, 61029 Urbino, Italy; (M.B.); (S.B.); (P.G.)
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (L.I.C.); (I.F.)
| | - Anna Bartoletti Stella
- Department of Diagnostic Experimental and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy;
| | - Michela Battistelli
- Department of Biomolecular Sciences (DiSB), Urbino University Carlo Bo, 61029 Urbino, Italy; (M.B.); (S.B.); (P.G.)
| | - Sabrina Burattini
- Department of Biomolecular Sciences (DiSB), Urbino University Carlo Bo, 61029 Urbino, Italy; (M.B.); (S.B.); (P.G.)
| | - Alberto Bavelloni
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Lucio Ildebrando Cocco
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (L.I.C.); (I.F.)
| | - Pietro Gobbi
- Department of Biomolecular Sciences (DiSB), Urbino University Carlo Bo, 61029 Urbino, Italy; (M.B.); (S.B.); (P.G.)
| | - Irene Faenza
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (L.I.C.); (I.F.)
| |
Collapse
|
23
|
Nahm M, Lim SM, Kim YE, Park J, Noh MY, Lee S, Roh JE, Hwang SM, Park CK, Kim YH, Lim G, Lee J, Oh KW, Ki CS, Kim SH. ANXA11 mutations in ALS cause dysregulation of calcium homeostasis and stress granule dynamics. Sci Transl Med 2021; 12:12/566/eaax3993. [PMID: 33087501 DOI: 10.1126/scitranslmed.aax3993] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 12/15/2019] [Accepted: 07/04/2020] [Indexed: 12/14/2022]
Abstract
Dysregulation of calcium ion homeostasis and abnormal protein aggregation have been proposed as major pathogenic hallmarks underpinning selective degeneration of motor neurons in amyotrophic lateral sclerosis (ALS). Recently, mutations in annexin A11 (ANXA11), a gene encoding a Ca2+-dependent phospholipid-binding protein, have been identified in familial and sporadic ALS. However, the physiological and pathophysiological roles of ANXA11 remain unknown. Here, we report functions of ANXA11 related to intracellular Ca2+ homeostasis and stress granule dynamics. We analyzed the exome sequences of 500 Korean patients with sALS and identified nine ANXA11 variants in 13 patients. The amino-terminal variants p.G38R and p.D40G within the low-complexity domain of ANXA11 enhanced aggregation propensity, whereas the carboxyl-terminal ANX domain variants p.H390P and p.R456H altered Ca2+ responses. Furthermore, all four variants in ANXA11 underwent abnormal phase separation to form droplets with aggregates and led to the alteration of the biophysical properties of ANXA11. These functional defects caused by ALS-linked variants induced alterations in both intracellular Ca2+ homeostasis and stress granule disassembly. We also revealed that p.G228Lfs*29 reduced ANXA11 expression and impaired Ca2+ homeostasis, as caused by missense variants. Ca2+-dependent interaction and coaggregation between ANXA11 and ALS-causative RNA-binding proteins, FUS and hnRNPA1, were observed in motor neuron cells and brain from a patient with ALS-FUS. The expression of ALS-linked ANXA11 variants in motor neuron cells caused cytoplasmic sequestration of endogenous FUS and triggered neuronal apoptosis. Together, our findings suggest that disease-associated ANXA11 mutations can contribute to ALS pathogenesis through toxic gain-of-function mechanisms involving abnormal protein aggregation.
Collapse
Affiliation(s)
- Minyeop Nahm
- Department of Neurology, College of Medicine, Hanyang University, Seoul 04763, Korea.,Biomedical Research Institute, Hanyang University, Seoul 04763, Korea
| | - Su Min Lim
- Department of Neurology, College of Medicine, Hanyang University, Seoul 04763, Korea.,Biomedical Research Institute, Hanyang University, Seoul 04763, Korea
| | - Young-Eun Kim
- Department of Laboratory Medicine, College of Medicine, Hanyang University, Seoul 04763, Korea
| | - Jinseok Park
- Department of Neurology, College of Medicine, Hanyang University, Seoul 04763, Korea
| | - Min-Young Noh
- Department of Neurology, College of Medicine, Hanyang University, Seoul 04763, Korea.,Biomedical Research Institute, Hanyang University, Seoul 04763, Korea
| | - Sanggon Lee
- Department of Neurology, College of Medicine, Hanyang University, Seoul 04763, Korea
| | - Ju Eun Roh
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea
| | - Sung-Min Hwang
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Korea
| | - GyuTae Lim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.,Department of Bioinformatics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Jinhyuk Lee
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.,Department of Bioinformatics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Ki-Wook Oh
- Department of Neurology, College of Medicine, Hanyang University, Seoul 04763, Korea
| | | | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul 04763, Korea. .,Biomedical Research Institute, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
24
|
Nishimura AL, Arias N. Synaptopathy Mechanisms in ALS Caused by C9orf72 Repeat Expansion. Front Cell Neurosci 2021; 15:660693. [PMID: 34140881 PMCID: PMC8203826 DOI: 10.3389/fncel.2021.660693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex neurodegenerative disease caused by degeneration of motor neurons (MNs). ALS pathogenic features include accumulation of misfolded proteins, glutamate excitotoxicity, mitochondrial dysfunction at distal axon terminals, and neuronal cytoskeleton changes. Synergies between loss of C9orf72 functions and gain of function by toxic effects of repeat expansions also contribute to C9orf72-mediated pathogenesis. However, the impact of haploinsufficiency of C9orf72 on neurons and in synaptic functions requires further examination. As the motor neurons degenerate, the disease symptoms will lead to neurotransmission deficiencies in the brain, spinal cord, and neuromuscular junction. Altered neuronal excitability, synaptic morphological changes, and C9orf72 protein and DPR localization at the synapses, suggest a potential involvement of C9orf72 at synapses. In this review article, we provide a conceptual framework for assessing the putative involvement of C9orf72 as a synaptopathy, and we explore the underlying and common disease mechanisms with other neurodegenerative diseases. Finally, we reflect on the major challenges of understanding C9orf72-ALS as a synaptopathy focusing on integrating mitochondrial and neuronal cytoskeleton degeneration as biomarkers and potential targets to treat ALS neurodegeneration.
Collapse
Affiliation(s)
- Agnes L Nishimura
- Department of Basic and Clinical Neuroscience, UK Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Natalia Arias
- Department of Basic and Clinical Neuroscience, UK Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
25
|
Corcia P, Camu W, Brulard C, Marouillat S, Couratier P, Camdessanché JP, Cintas P, Verschueren A, Soriani MH, Desnuelle C, Fleury MC, Guy N, Cassereau J, Viader F, Pittion-Vouyovitch S, Danel V, Kolev I, Le Masson G, Beltran S, Salachas F, Bernard E, Pradat PF, Blasco H, Lanznaster D, Hergesheimer R, Laumonnier F, Andres CR, Meininger V, Vourc'h P. Effect of familial clustering in the genetic screening of 235 French ALS families. J Neurol Neurosurg Psychiatry 2021; 92:479-484. [PMID: 33408239 DOI: 10.1136/jnnp-2020-325064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 01/15/2023]
Abstract
OBJECTIVES To determine whether the familial clustering of amyotrophic lateral sclerosis (ALS) cases and the phenotype of the disease may help identify the pathogenic genes involved. METHODS We conducted a targeted next-generation sequencing analysis on 235 French familial ALS (FALS), unrelated probands to identify mutations in 30 genes linked to the disease. The genealogy, that is, number of cases and generations with ALS, gender, age, site of onset and the duration of the disease were analysed. RESULTS Regarding the number of generations, 49 pedigrees had only one affected generation, 152 had two affected generations and 34 had at least three affected generations. Among the 149 pedigrees (63.4%) for which a deleterious variant was found, an abnormal G4C2 expansion in C9orf72 was found in 98 cases as well as SOD1, TARBP or FUS mutations in 30, 9 and 7 cases, respectively. Considering pedigrees from the number of generations, abnormal G4C2 expansion in C9orf72 was more frequent in pedigrees with pairs of affected ALS cases, which represented 65.2% of our cohort. SOD1 mutation involved all types of pedigrees. No TARDBP nor FUS mutation was present in monogenerational pedigrees. TARDBP mutation predominated in bigenerational pedigrees with at least three cases and FUS mutation in multigenerational pedigrees with more than seven cases, on average, and with an age of onset younger than 45 years. CONCLUSION Our results suggest that familial clustering, phenotypes and genotypes are interconnected in FALS, and thus it might be possible to target the genetic screening from the familial architecture and the phenotype of ALS cases.
Collapse
Affiliation(s)
- Philippe Corcia
- ALS Centre, Department of Neurology, CHU Tours, Tours, Centre, France .,UMR 1253 Imaging and Brain, Tours, Centre-Val de Loire, France
| | - William Camu
- Montpellier 2 University, Montpellier, Languedoc-Roussillon, France
| | - Celine Brulard
- UMR 1253 Imaging and Brain, Tours, Centre-Val de Loire, France
| | | | - Philippe Couratier
- ALS Center, Departement of neurology, CHU Limoges, Limoges, Limousin, France.,UMR-S 1094 NET, Limoges, France
| | | | - Pascal Cintas
- Neurology, CHU Toulouse, Toulouse, Midi-Pyrénées, France
| | - Annie Verschueren
- Centre de référence des maladies neuromusculaires et de la SLA, Marseille Public University Hospital System, Marseille, Provence-Alpes-Côte d'Azu, France
| | | | - Claude Desnuelle
- Centre de reference des maladies neuromusculaires SLA, Neurosciences department, CHU Nice, Nice, Provence-Alpes-Côte d'Azu, France
| | | | | | | | | | | | - Veronique Danel
- Medical Pharmacology, Lille University Hospital Center, Lille, Hauts-de-France, France
| | - Ivan Kolev
- Hospital Centre Saint Brieuc, Saint Brieuc, Bretagne, France
| | - Gwendal Le Masson
- Neurology, Centre Hospitalier Universitaire de Bordeaux Groupe Hospitalier Pellegrin, Bordeaux, Aquitaine, France
| | - Stephane Beltran
- ALS Center, Francois-Rabelais University, Tours, Centre-Val de Loire, France
| | - Francois Salachas
- ALS Center, Neurology, Hopital Universitaire Pitie Salpetriere, Paris, Île-de-France, France
| | - Emilien Bernard
- University Hospital Centre Lyon, Lyon, Auvergne-Rhône-Alpes, France
| | - Pierre-François Pradat
- Fédération de Neurologie, Centre Référent SLA, Hopital Universitaire Pitie Salpetriere, Paris, Île-de-France, France.,CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Université Pierre et Marie Curie Faculté de Médecine, Paris, Île-de-France, France
| | - Hélène Blasco
- Biochemistry and Molecular Biology Department, Université Francois-Rabelais de Tours, Tours, Centre-Val de Loire, France.,Neurogenetics and Neurometabolomics, Imagerie et cerveau, Tours, France
| | | | | | | | - Christian R Andres
- Biochemistry and Molecular Biology Department, Université Francois-Rabelais de Tours, Tours, Centre-Val de Loire, France
| | - Vincent Meininger
- Neurogenetics and Neurometabolomics, Imagerie et cerveau, Tours, France
| | - Patrick Vourc'h
- Biochemistry and Molecular Biology Department, Université Francois-Rabelais de Tours, Tours, Centre-Val de Loire, France
| |
Collapse
|
26
|
Don EK, Maschirow A, Radford RAW, Scherer NM, Vidal-Itriago A, Hogan A, Maurel C, Formella I, Stoddart JJ, Hall TE, Lee A, Shi B, Cole NJ, Laird AS, Badrock AP, Chung RS, Morsch M. In vivo Validation of Bimolecular Fluorescence Complementation (BiFC) to Investigate Aggregate Formation in Amyotrophic Lateral Sclerosis (ALS). Mol Neurobiol 2021; 58:2061-2074. [PMID: 33415684 PMCID: PMC8018926 DOI: 10.1007/s12035-020-02238-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/25/2020] [Indexed: 10/28/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a form of motor neuron disease (MND) that is characterized by the progressive loss of motor neurons within the spinal cord, brainstem, and motor cortex. Although ALS clinically manifests as a heterogeneous disease, with varying disease onset and survival, a unifying feature is the presence of ubiquitinated cytoplasmic protein inclusion aggregates containing TDP-43. However, the precise mechanisms linking protein inclusions and aggregation to neuronal loss are currently poorly understood. Bimolecular fluorescence complementation (BiFC) takes advantage of the association of fluorophore fragments (non-fluorescent on their own) that are attached to an aggregation-prone protein of interest. Interaction of the proteins of interest allows for the fluorescent reporter protein to fold into its native state and emit a fluorescent signal. Here, we combined the power of BiFC with the advantages of the zebrafish system to validate, optimize, and visualize the formation of ALS-linked aggregates in real time in a vertebrate model. We further provide in vivo validation of the selectivity of this technique and demonstrate reduced spontaneous self-assembly of the non-fluorescent fragments in vivo by introducing a fluorophore mutation. Additionally, we report preliminary findings on the dynamic aggregation of the ALS-linked hallmark proteins Fus and TDP-43 in their corresponding nuclear and cytoplasmic compartments using BiFC. Overall, our data demonstrates the suitability of this BiFC approach to study and characterize ALS-linked aggregate formation in vivo. Importantly, the same principle can be applied in the context of other neurodegenerative diseases and has therefore critical implications to advance our understanding of pathologies that underlie aberrant protein aggregation.
Collapse
Affiliation(s)
- Emily K Don
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Alina Maschirow
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Rowan A W Radford
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Natalie M Scherer
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Andrés Vidal-Itriago
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Alison Hogan
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Cindy Maurel
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Isabel Formella
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Jack J Stoddart
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Thomas E Hall
- Institute for Molecular Bioscience, The University of Queensland, QLD, St Lucia, 4072, Australia
| | - Albert Lee
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Bingyang Shi
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Nicholas J Cole
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Angela S Laird
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Andrew P Badrock
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| | - Roger S Chung
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Marco Morsch
- Centre for Motor Neuron Disease Research, Faculty of Medicine, Health and Human Sciences, Department of Biomedical Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
27
|
Amyotrophic Lateral Sclerosis and Frontotemporal Lobar Degenerations: Similarities in Genetic Background. Diagnostics (Basel) 2021; 11:diagnostics11030509. [PMID: 33805659 PMCID: PMC7998502 DOI: 10.3390/diagnostics11030509] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/06/2021] [Accepted: 03/11/2021] [Indexed: 12/27/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating, uniformly lethal progressive degenerative disorder of motor neurons that overlaps with frontotemporal lobar degeneration (FTLD) clinically, morphologically, and genetically. Although many distinct mutations in various genes are known to cause amyotrophic lateral sclerosis, it remains poorly understood how they selectively impact motor neuron biology and whether they converge on common pathways to cause neuronal degeneration. Many of the gene mutations are in proteins that share similar functions. They can be grouped into those associated with cell axon dynamics and those associated with cellular phagocytic machinery, namely protein aggregation and metabolism, apoptosis, and intracellular nucleic acid transport. Analysis of pathways implicated by mutant ALS genes has provided new insights into the pathogenesis of both familial forms of ALS (fALS) and sporadic forms (sALS), although, regrettably, this has not yet yielded definitive treatments. Many genes play an important role, with TARDBP, SQSTM1, VCP, FUS, TBK1, CHCHD10, and most importantly, C9orf72 being critical genetic players in these neurological disorders. In this mini-review, we will focus on the molecular mechanisms of these two diseases.
Collapse
|
28
|
Imamura K, Yada Y, Izumi Y, Morita M, Kawata A, Arisato T, Nagahashi A, Enami T, Tsukita K, Kawakami H, Nakagawa M, Takahashi R, Inoue H. Prediction Model of Amyotrophic Lateral Sclerosis by Deep Learning with Patient Induced Pluripotent Stem Cells. Ann Neurol 2021; 89:1226-1233. [PMID: 33565152 PMCID: PMC8247989 DOI: 10.1002/ana.26047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/21/2022]
Abstract
In amyotrophic lateral sclerosis (ALS), early diagnosis is essential for both current and potential treatments. To find a supportive approach for the diagnosis, we constructed an artificial intelligence‐based prediction model of ALS using induced pluripotent stem cells (iPSCs). Images of spinal motor neurons derived from healthy control subject and ALS patient iPSCs were analyzed by a convolutional neural network, and the algorithm achieved an area under the curve of 0.97 for classifying healthy control and ALS. This prediction model by deep learning algorithm with iPSC technology could support the diagnosis and may provide proactive treatment of ALS through future prospective research. ANN NEUROL 2021;89:1226–1233
Collapse
Affiliation(s)
- Keiko Imamura
- Medical-Risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project, Kyoto, Japan.,Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
| | - Yuichiro Yada
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
| | - Yuishin Izumi
- Department of Clinical Neuroscience, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Mitsuya Morita
- Division of Neurology, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Akihiro Kawata
- Department of Neurology, Tokyo Metropolitan Neurological Hospital, Tokyo, Japan
| | - Takayo Arisato
- Department of Neurology, National Hospital Organization Minamikyusyu Hospital, Kagoshima, Japan
| | - Ayako Nagahashi
- Medical-Risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project, Kyoto, Japan.,Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Takako Enami
- Medical-Risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project, Kyoto, Japan.,Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Kayoko Tsukita
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan
| | - Hideshi Kawakami
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Masanori Nakagawa
- Department of Neurology, North Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhisa Inoue
- Medical-Risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project, Kyoto, Japan.,Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center, Kyoto, Japan.,Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
29
|
Dangoumau A, Marouillat S, Coelho R, Wurmser F, Brulard C, Haouari S, Laumonnier F, Corcia P, Andres CR, Blasco H, Vourc’h P. Dysregulations of Expression of Genes of the Ubiquitin/SUMO Pathways in an In Vitro Model of Amyotrophic Lateral Sclerosis Combining Oxidative Stress and SOD1 Gene Mutation. Int J Mol Sci 2021; 22:ijms22041796. [PMID: 33670299 PMCID: PMC7918082 DOI: 10.3390/ijms22041796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/24/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Protein aggregates in affected motor neurons are a hallmark of amyotrophic lateral sclerosis (ALS), but the molecular pathways leading to their formation remain incompletely understood. Oxidative stress associated with age, the major risk factor in ALS, contributes to this neurodegeneration in ALS. We show that several genes coding for enzymes of the ubiquitin and small ubiquitin-related modifier (SUMO) pathways exhibit altered expression in motor neuronal cells exposed to oxidative stress, such as the CCNF gene mutated in ALS patients. Eleven of these genes were further studied in conditions combining oxidative stress and the expression of an ALS related mutant of the superoxide dismutase 1 (SOD1) gene. We observed a combined effect of these two environmental and genetic factors on the expression of genes, such as Uhrf2, Rbx1, Kdm2b, Ube2d2, Xaf1, and Senp1. Overall, we identified dysregulations in the expression of enzymes of the ubiquitin and SUMO pathways that may be of interest to better understand the pathophysiology of ALS and to protect motor neurons from oxidative stress and genetic alterations.
Collapse
Affiliation(s)
- Audrey Dangoumau
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | - Sylviane Marouillat
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | - Roxane Coelho
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | - François Wurmser
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | | | - Shanez Haouari
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | - Frédéric Laumonnier
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
| | - Philippe Corcia
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
- Service de Neurologie, Centre de Référence sur la SLA, CHRU de Tours, 37000 Tours, France
| | - Christian R. Andres
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37000 Tours, France
| | - Hélène Blasco
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37000 Tours, France
| | - Patrick Vourc’h
- UMR iBrain, Université de Tours, Inserm, 37000 Tours, France; (A.D.); (S.M.); (R.C.); (F.W.); (S.H.); (F.L.); (P.C.); (C.R.A.); (H.B.)
- UTTIL, CHRU de Tours, 37000 Tours, France;
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37000 Tours, France
- Correspondence: ; Tel.: +33-(0)-234-378-910
| |
Collapse
|
30
|
Manzano R, Toivonen JM, Moreno-Martínez L, de la Torre M, Moreno-García L, López-Royo T, Molina N, Zaragoza P, Calvo AC, Osta R. What skeletal muscle has to say in amyotrophic lateral sclerosis: Implications for therapy. Br J Pharmacol 2020; 178:1279-1297. [PMID: 32986860 DOI: 10.1111/bph.15276] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/03/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult onset disorder characterized by progressive neuromuscular junction (NMJ) dismantling and degeneration of motor neurons leading to atrophy and paralysis of voluntary muscles responsible for motion and breathing. Except for a minority of patients harbouring genetic mutations, the origin of most ALS cases remains elusive. Peripheral tissues, and particularly skeletal muscle, have lately demonstrated an active contribution to disease pathology attracting a growing interest for these tissues as therapeutic targets in ALS. In this sense, molecular mechanisms essential for cell and tissue homeostasis have been shown to be deregulated in the disease. These include muscle metabolism and mitochondrial activity, RNA processing, tissue-resident stem cell function responsible for muscle regeneration, and proteostasis that regulates muscle mass in adulthood. This review aims to compile scientific evidence that demonstrates the role of skeletal muscle in ALS pathology and serves as reference for development of novel therapeutic strategies targeting this tissue to delay disease onset and progression. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.
Collapse
Affiliation(s)
- Raquel Manzano
- Department of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Agroalimentary Institute of Aragon (IA2), Institute of Health Research of Aragon (IIS), Zaragoza, Spain
| | - Janne Markus Toivonen
- Department of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Agroalimentary Institute of Aragon (IA2), Institute of Health Research of Aragon (IIS), Zaragoza, Spain
| | - Laura Moreno-Martínez
- Department of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Agroalimentary Institute of Aragon (IA2), Institute of Health Research of Aragon (IIS), Zaragoza, Spain
| | - Miriam de la Torre
- Department of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Agroalimentary Institute of Aragon (IA2), Institute of Health Research of Aragon (IIS), Zaragoza, Spain
| | - Leticia Moreno-García
- Department of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Agroalimentary Institute of Aragon (IA2), Institute of Health Research of Aragon (IIS), Zaragoza, Spain
| | - Tresa López-Royo
- Department of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Agroalimentary Institute of Aragon (IA2), Institute of Health Research of Aragon (IIS), Zaragoza, Spain
| | - Nora Molina
- Department of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Agroalimentary Institute of Aragon (IA2), Institute of Health Research of Aragon (IIS), Zaragoza, Spain.,Geriatrics Service, Hospital Nuestra Señora de Gracia, Zaragoza, Spain
| | - Pilar Zaragoza
- Department of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Agroalimentary Institute of Aragon (IA2), Institute of Health Research of Aragon (IIS), Zaragoza, Spain
| | - Ana Cristina Calvo
- Department of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Agroalimentary Institute of Aragon (IA2), Institute of Health Research of Aragon (IIS), Zaragoza, Spain
| | - Rosario Osta
- Department of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Agroalimentary Institute of Aragon (IA2), Institute of Health Research of Aragon (IIS), Zaragoza, Spain
| |
Collapse
|
31
|
Morello G, Salomone S, D’Agata V, Conforti FL, Cavallaro S. From Multi-Omics Approaches to Precision Medicine in Amyotrophic Lateral Sclerosis. Front Neurosci 2020; 14:577755. [PMID: 33192262 PMCID: PMC7661549 DOI: 10.3389/fnins.2020.577755] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating and fatal neurodegenerative disorder, caused by the degeneration of upper and lower motor neurons for which there is no truly effective cure. The lack of successful treatments can be well explained by the complex and heterogeneous nature of ALS, with patients displaying widely distinct clinical features and progression patterns, and distinct molecular mechanisms underlying the phenotypic heterogeneity. Thus, stratifying ALS patients into consistent and clinically relevant subgroups can be of great value for the development of new precision diagnostics and targeted therapeutics for ALS patients. In the last years, the use and integration of high-throughput "omics" approaches have dramatically changed our thinking about ALS, improving our understanding of the complex molecular architecture of ALS, distinguishing distinct patient subtypes and providing a rational foundation for the discovery of biomarkers and new individualized treatments. In this review, we discuss the most significant contributions of omics technologies in unraveling the biological heterogeneity of ALS, highlighting how these approaches are revealing diagnostic, prognostic and therapeutic targets for future personalized interventions.
Collapse
Affiliation(s)
- Giovanna Morello
- Institute for Research and Biomedical Innovation (IRIB), Italian National Research Council (CNR), Catania, Italy
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Salvatore Salomone
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Velia D’Agata
- Human Anatomy and Histology, University of Catania, Catania, Italy
| | | | - Sebastiano Cavallaro
- Institute for Research and Biomedical Innovation (IRIB), Italian National Research Council (CNR), Catania, Italy
| |
Collapse
|
32
|
Limanaqi F, Biagioni F, Mastroiacovo F, Polzella M, Lazzeri G, Fornai F. Merging the Multi-Target Effects of Phytochemicals in Neurodegeneration: From Oxidative Stress to Protein Aggregation and Inflammation. Antioxidants (Basel) 2020; 9:antiox9101022. [PMID: 33092300 PMCID: PMC7589770 DOI: 10.3390/antiox9101022] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023] Open
Abstract
Wide experimental evidence has been provided in the last decade concerning the neuroprotective effects of phytochemicals in a variety of neurodegenerative disorders. Generally, the neuroprotective effects of bioactive compounds belonging to different phytochemical classes are attributed to antioxidant, anti-aggregation, and anti-inflammatory activity along with the restoration of mitochondrial homeostasis and targeting alterations of cell-clearing systems. Far from being independent, these multi-target effects represent interconnected events that are commonly implicated in the pathogenesis of most neurodegenerative diseases, independently of etiology, nosography, and the specific misfolded proteins being involved. Nonetheless, the increasing amount of data applying to a variety of neurodegenerative disorders joined with the multiple effects exerted by the wide variety of plant-derived neuroprotective agents may rather confound the reader. The present review is an attempt to provide a general guideline about the most relevant mechanisms through which naturally occurring agents may counteract neurodegeneration. With such an aim, we focus on some popular phytochemical classes and bioactive compounds as representative examples to design a sort of main highway aimed at deciphering the most relevant protective mechanisms which make phytochemicals potentially useful in counteracting neurodegeneration. In this frame, we emphasize the potential role of the cell-clearing machinery as a kernel in the antioxidant, anti-aggregation, anti-inflammatory, and mitochondrial protecting effects of phytochemicals.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
| | - Francesca Biagioni
- Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (F.M.)
| | - Federica Mastroiacovo
- Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (F.M.)
| | - Maico Polzella
- Aliveda Laboratories, Viale Karol Wojtyla 19, 56042 Crespina Lorenzana, Italy;
| | - Gloria Lazzeri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
- Correspondence: (G.L.); (F.F.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
- Istituto di Ricovero e Cura a Carattere Scientifico Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (F.M.)
- Correspondence: (G.L.); (F.F.)
| |
Collapse
|
33
|
Proteasome Subunits Involved in Neurodegenerative Diseases. Arch Med Res 2020; 52:1-14. [PMID: 32962866 DOI: 10.1016/j.arcmed.2020.09.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/25/2020] [Accepted: 09/04/2020] [Indexed: 12/29/2022]
Abstract
The ubiquitin-proteasome system is the major pathway for the maintenance of protein homeostasis. Its inhibition causes accumulation of ubiquitinated proteins; this accumulation has been associated with several of the most common neurodegenerative diseases. Several genetic factors have been identified for most neurodegenerative diseases, however, most cases are considered idiopathic, thus making the study of the mechanisms of protein accumulation a relevant field of research. It is often mentioned that the biggest risk factor for neurodegenerative diseases is aging, and several groups have reported an age-related alteration of the expression of some of the 26S proteasome subunits and a reduction of its activity. Proteasome subunits interact with proteins that are known to accumulate in neurodegenerative diseases such as α-synuclein in Parkinson's, tau in Alzheimer's, and huntingtin in Huntington's diseases. These interactions have been explored for several years, but only until recently, we are beginning to understand them. In this review, we discuss the known interactions, the underlying patterns, and the phenotypes associated with the 26S proteasome subunits in the etiology and progression of neurodegenerative diseases where there is evidence of proteasome involvement. Special emphasis is made in reviewing proteasome subunits that interact with proteins known to have an age-related altered expression or to be involved in neurodegenerative diseases to explore key effectors that may trigger or augment their progression. Interestingly, while the causes of age-related reduction of some of the proteasome subunits are not known, there are specific relationships between the observed neurodegenerative disease and the affected proteasome subunits.
Collapse
|
34
|
Kitajima Y, Yoshioka K, Suzuki N. The ubiquitin-proteasome system in regulation of the skeletal muscle homeostasis and atrophy: from basic science to disorders. J Physiol Sci 2020; 70:40. [PMID: 32938372 PMCID: PMC10717345 DOI: 10.1186/s12576-020-00768-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/05/2020] [Indexed: 02/07/2023]
Abstract
Skeletal muscle is one of the most abundant and highly plastic tissues. The ubiquitin-proteasome system (UPS) is recognised as a major intracellular protein degradation system, and its function is important for muscle homeostasis and health. Although UPS plays an essential role in protein degradation during muscle atrophy, leading to the loss of muscle mass and strength, its deficit negatively impacts muscle homeostasis and leads to the occurrence of several pathological phenotypes. A growing number of studies have linked UPS impairment not only to matured muscle fibre degeneration and weakness, but also to muscle stem cells and deficiency in regeneration. Emerging evidence suggests possible links between abnormal UPS regulation and several types of muscle diseases. Therefore, understanding of the role of UPS in skeletal muscle may provide novel therapeutic insights to counteract muscle wasting, and various muscle diseases. In this review, we focussed on the role of proteasomes in skeletal muscle and its regeneration, including a brief explanation of the structure of proteasomes. In addition, we summarised the recent findings on several diseases and elaborated on how the UPS is related to their pathological states.
Collapse
Affiliation(s)
- Yasuo Kitajima
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto, 860-0811, Japan.
| | - Kiyoshi Yoshioka
- Institute for Research On Productive Aging (IRPA), #201 Kobe hybrid business center, Minami-cho 6-7-6, Minatojima, Kobe, 650-0047, Japan
| | - Naoki Suzuki
- Department of Neurology, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
- Department of Neurology, Shodo-Kai Southern Tohoku General Hospital, 1-2-5, Satonomori, Iwanuma, Miyagi, 989-2483, Japan.
| |
Collapse
|
35
|
Pro-Oxidant Activity of an ALS-Linked SOD1 Mutant in Zn-Deficient Form. Molecules 2020; 25:molecules25163600. [PMID: 32784718 PMCID: PMC7464938 DOI: 10.3390/molecules25163600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 07/30/2020] [Accepted: 08/05/2020] [Indexed: 11/17/2022] Open
Abstract
Cu, Zn superoxide dismutase (SOD1) is a representative antioxidant enzyme that catalyzes dismutation of reactive oxygen species in cells. However, (E,E)-SOD1 mutants in which both copper and zinc ions were deleted exhibit pro-oxidant activity, contrary to their antioxidant nature, at physiological temperatures, following denaturation and subsequent recombination of Cu2+. This oxidative property is likely related to the pathogenesis of amyotrophic lateral sclerosis (ALS); however, the mechanism by which Cu2+ re-binds to the denatured (E,E)-SOD1 has not been elucidated, since the concentration of free copper ions in cells is almost zero. In this study, we prepared the (Cu,E) form in which only a zinc ion was deleted using ALS-linked mutant H43R (His43→Arg) and found that (Cu,E)-H43R showed an increase in the pro-oxidant activity even at physiological temperature. The increase in the pro-oxidant activity of (Cu,E)-H43R was also observed in solution mimicking intracellular environment and at high temperature. These results suggest that the zinc-deficient (Cu,E) form can contribute to oxidative stress in cells, and that the formation of (E,E)-SOD1 together with the subsequent Cu2+ rebinding is not necessary for the acquisition of the pro-oxidant activity.
Collapse
|
36
|
Clark CM, Clark RM, Hoyle JA, Dickson TC. Pathogenic or protective? Neuropeptide Y in amyotrophic lateral sclerosis. J Neurochem 2020; 156:273-289. [PMID: 32654149 DOI: 10.1111/jnc.15125] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/16/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022]
Abstract
Neuropeptide Y (NPY) is an endogenous peptide of the central and enteric nervous systems which has gained significant interest as a potential neuroprotective agent for treatment of neurodegenerative disease. Amyotrophic lateral sclerosis (ALS) is an aggressive and fatal neurodegenerative disease characterized by motor deficits and motor neuron loss. In ALS, recent evidence from ALS patients and animal models has indicated that NPY may have a role in the disease pathogenesis. Increased NPY levels were found to correlate with disease progression in ALS patients. Similarly, NPY expression is increased in the motor cortex of ALS mice by end stages of the disease. Although the functional consequence of increased NPY levels in ALS is currently unknown, NPY has been shown to exert a diverse range of neuroprotective roles in other neurodegenerative diseases; through modulation of potassium channel activity, increased production of neurotrophins, inhibition of endoplasmic reticulum stress and autophagy, reduction of excitotoxicity, oxidative stress, neuroinflammation and hyperexcitability. Several of these mechanisms and signalling pathways are heavily implicated in the pathogenesis of ALS. Therefore, in this review, we discuss possible effects of NPY and NPY-receptor signalling in the ALS disease context, as determining NPY's contribution to, or impact on, ALS disease mechanisms will be essential for future studies investigating the NPY system as a therapeutic strategy in this devastating disease.
Collapse
Affiliation(s)
- Courtney M Clark
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Rosemary M Clark
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Joshua A Hoyle
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Tracey C Dickson
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
37
|
Zhang C, Ötjengerdes RM, Roewe J, Mejias R, Marschall ALJ. Applying Antibodies Inside Cells: Principles and Recent Advances in Neurobiology, Virology and Oncology. BioDrugs 2020; 34:435-462. [PMID: 32301049 PMCID: PMC7391400 DOI: 10.1007/s40259-020-00419-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To interfere with cell function, many scientists rely on methods that target DNA or RNA due to the ease with which they can be applied. Proteins are usually the final executors of function but are targeted only indirectly by these methods. Recent advances in targeted degradation of proteins based on proteolysis-targeting chimaeras (PROTACs), ubiquibodies, deGradFP (degrade Green Fluorescent Protein) and other approaches have demonstrated the potential of interfering directly at the protein level for research and therapy. Proteins can be targeted directly and very specifically by antibodies, but using antibodies inside cells has so far been considered to be challenging. However, it is possible to deliver antibodies or other proteins into the cytosol using standard laboratory equipment. Physical methods such as electroporation have been demonstrated to be efficient and validated thoroughly over time. The expression of intracellular antibodies (intrabodies) inside cells is another way to interfere with intracellular targets at the protein level. Methodological strategies to target the inside of cells with antibodies, including delivered antibodies and expressed antibodies, as well as applications in the research areas of neurobiology, viral infections and oncology, are reviewed here. Antibodies have already been used to interfere with a wide range of intracellular targets. Disease-related targets included proteins associated with neurodegenerative diseases such as Parkinson's disease (α-synuclein), Alzheimer's disease (amyloid-β) or Huntington's disease (mutant huntingtin [mHtt]). The applications of intrabodies in the context of viral infections include targeting proteins associated with HIV (e.g. HIV1-TAT, Rev, Vif, gp41, gp120, gp160) and different oncoviruses such as human papillomavirus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV) and Epstein-Barr virus, and they have been used to interfere with various targets related to different processes in cancer, including oncogenic pathways, proliferation, cell cycle, apoptosis, metastasis, angiogenesis or neo-antigens (e.g. p53, human epidermal growth factor receptor-2 [HER2], signal transducer and activator of transcription 3 [STAT3], RAS-related RHO-GTPase B (RHOB), cortactin, vascular endothelial growth factor receptor 2 [VEGFR2], Ras, Bcr-Abl). Interfering at the protein level allows questions to be addressed that may remain unanswered using alternative methods. This review addresses why direct targeting of proteins allows unique insights, what is currently feasible in vitro, and how this relates to potential therapeutic applications.
Collapse
Affiliation(s)
- Congcong Zhang
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rina M Ötjengerdes
- Hannover Medical School (MHH), Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Julian Roewe
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain TumorImmunology (D170), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rebeca Mejias
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Andrea L J Marschall
- Technische Universität Braunschweig, Institute of Biochemistry, Biotechnology and Bioinformatics, Brunswick, Germany.
| |
Collapse
|
38
|
Gomes C, Sequeira C, Barbosa M, Cunha C, Vaz AR, Brites D. Astrocyte regional diversity in ALS includes distinct aberrant phenotypes with common and causal pathological processes. Exp Cell Res 2020; 395:112209. [PMID: 32739211 DOI: 10.1016/j.yexcr.2020.112209] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022]
Abstract
Astrocytes are major contributors of motor neuron (MN) degeneration in amyotrophic lateral sclerosis (ALS). We investigated whether regional and cell maturation differences influence ALS astrocyte malfunction. Spinal and cortical astrocytes from SOD1G93A (mSOD1) 7-day-old mice were cultured for 5 and 13 days in vitro (DIV). Astrocyte aberrancies predominated in 13DIV cells with region specificity. 13DIV cortical mSOD1 astrocytes showed early morphological changes and a predominant reactive and inflammatory phenotype, while repressed proteins and genes were found in spinal cells. Inflammatory-associated miRNAs, e.g. miR-155/miR-21/miR-146a, were downregulated in the first and upregulated in the later ones. Interestingly, depleted miR-155/miR-21/miR-146a in small extracellular vesicles (sEVs/exosomes) was a common pathological feature. Cortical mSOD1 astrocytes induced late apoptosis and kinesin-1 downregulation in mSOD1 NSC-34 MNs, whereas spinal cells upregulated dynein, while decreased nNOS and synaptic-related genes. Both regional-distinct mSOD1 astrocytes enhanced iNOS gene expression in mSOD1 MNs. We provide information on the potential contribution of astrocytes to ALS bulbar-vs. spinal-onset pathology, local influence on neuronal dysfunction and their shared miRNA-depleted exosome trafficking. These causal and common features may have potential therapeutic implications in ALS. Future studies should clarify if astrocyte-derived sEVs are active players in ALS-related neuroinflammation and glial activation.
Collapse
Affiliation(s)
- Cátia Gomes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Catarina Sequeira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Marta Barbosa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Carolina Cunha
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Ana Rita Vaz
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003, Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
39
|
Studying ALS: Current Approaches, Effect on Potential Treatment Strategy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1241:195-217. [PMID: 32383122 DOI: 10.1007/978-3-030-41283-8_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is one of the most common neurodegenerative diseases, characterized by inevitable progressive paralysis. To date, only two disease modifying therapeutic options are available for the patients with ALS, although they show very modest effect on disease course. The main reason of failure in the field of pharmacological correction of ALS is inability to untangle complex relationships taking place during ALS initiation and progression. Traditional methods of research, based on morphology or transgenic animal models studying provided lots of information about ALS throughout the years. However, translation of these results to humans was unsuccessful due to incomplete recapitulation of molecular pathology and overall inadequacy of the models used in the research.In this review we summarize current knowledge regarding ALS molecular pathology with depiction of novel methods applied recently for the studies. Furthermore we describe present and potential treatment strategies that are based on the recent findings in ALS disease mechanisms.
Collapse
|
40
|
Limanaqi F, Busceti CL, Biagioni F, Cantini F, Lenzi P, Fornai F. Cell-Clearing Systems Bridging Repeat Expansion Proteotoxicity and Neuromuscular Junction Alterations in ALS and SBMA. Int J Mol Sci 2020; 21:ijms21114021. [PMID: 32512809 PMCID: PMC7312203 DOI: 10.3390/ijms21114021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
The coordinated activities of autophagy and the ubiquitin proteasome system (UPS) are key to preventing the aggregation and toxicity of misfold-prone proteins which manifest in a number of neurodegenerative disorders. These include proteins which are encoded by genes containing nucleotide repeat expansions. In the present review we focus on the overlapping role of autophagy and the UPS in repeat expansion proteotoxicity associated with chromosome 9 open reading frame 72 (C9ORF72) and androgen receptor (AR) genes, which are implicated in two motor neuron disorders, amyotrophic lateral sclerosis (ALS) and spinal-bulbar muscular atrophy (SBMA), respectively. At baseline, both C9ORF72 and AR regulate autophagy, while their aberrantly-expanded isoforms may lead to a failure in both autophagy and the UPS, further promoting protein aggregation and toxicity within motor neurons and skeletal muscles. Besides proteotoxicity, autophagy and UPS alterations are also implicated in neuromuscular junction (NMJ) alterations, which occur early in both ALS and SBMA. In fact, autophagy and the UPS intermingle with endocytic/secretory pathways to regulate axonal homeostasis and neurotransmission by interacting with key proteins which operate at the NMJ, such as agrin, acetylcholine receptors (AChRs), and adrenergic beta2 receptors (B2-ARs). Thus, alterations of autophagy and the UPS configure as a common hallmark in both ALS and SBMA disease progression. The findings here discussed may contribute to disclosing overlapping molecular mechanisms which are associated with a failure in cell-clearing systems in ALS and SBMA.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (F.L.); (F.C.); (P.L.)
| | | | - Francesca Biagioni
- I.R.C.C.S. Neuromed, Via Atinense, 18, 86077 Pozzilli, Italy; (C.L.B.); (F.B.)
| | - Federica Cantini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (F.L.); (F.C.); (P.L.)
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (F.L.); (F.C.); (P.L.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (F.L.); (F.C.); (P.L.)
- I.R.C.C.S. Neuromed, Via Atinense, 18, 86077 Pozzilli, Italy; (C.L.B.); (F.B.)
- Correspondence:
| |
Collapse
|
41
|
Lanznaster D, Veyrat-Durebex C, Vourc’h P, Andres CR, Blasco H, Corcia P. Metabolomics: A Tool to Understand the Impact of Genetic Mutations in Amyotrophic Lateral Sclerosis. Genes (Basel) 2020; 11:genes11050537. [PMID: 32403313 PMCID: PMC7288444 DOI: 10.3390/genes11050537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolomics studies performed in patients with amyotrophic lateral sclerosis (ALS) reveal a set of distinct metabolites that can shed light on the pathological alterations taking place in each individual. Metabolites levels are influenced by disease status, and genetics play an important role both in familial and sporadic ALS cases. Metabolomics analysis helps to unravel the differential impact of the most common ALS-linked genetic mutations (as C9ORF72, SOD1, TARDBP, and FUS) in specific signaling pathways. Further, studies performed in genetic models of ALS reinforce the role of TDP-43 pathology in the vast majority of ALS cases. Studies performed in differentiated cells from ALS-iPSC (induced Pluripotent Stem Cells) reveal alterations in the cell metabolism that are also found in ALS models and ultimately in ALS patients. The development of metabolomics approaches in iPSC derived from ALS patients allow addressing and ultimately understanding the pathological mechanisms taking place in any patient. Lately, the creation of a "patient in a dish" will help to identify patients that may benefit from specific treatments and allow the implementation of personalized medicine.
Collapse
Affiliation(s)
- Débora Lanznaster
- UMR 1253, iBrain, University of Tours, Inserm, 37000 Tours, France; (C.V.-D.); (P.V.); (C.R.A.); (H.B.); (P.C.)
- Correspondence:
| | - Charlotte Veyrat-Durebex
- UMR 1253, iBrain, University of Tours, Inserm, 37000 Tours, France; (C.V.-D.); (P.V.); (C.R.A.); (H.B.); (P.C.)
- CHU de Tours, Service de Biochimie et Biologie Moléculaire, 37000 Tours, France
| | - Patrick Vourc’h
- UMR 1253, iBrain, University of Tours, Inserm, 37000 Tours, France; (C.V.-D.); (P.V.); (C.R.A.); (H.B.); (P.C.)
- CHU de Tours, Service de Biochimie et Biologie Moléculaire, 37000 Tours, France
| | - Christian R. Andres
- UMR 1253, iBrain, University of Tours, Inserm, 37000 Tours, France; (C.V.-D.); (P.V.); (C.R.A.); (H.B.); (P.C.)
- CHU de Tours, Service de Biochimie et Biologie Moléculaire, 37000 Tours, France
| | - Hélène Blasco
- UMR 1253, iBrain, University of Tours, Inserm, 37000 Tours, France; (C.V.-D.); (P.V.); (C.R.A.); (H.B.); (P.C.)
- CHU de Tours, Service de Biochimie et Biologie Moléculaire, 37000 Tours, France
| | - Philippe Corcia
- UMR 1253, iBrain, University of Tours, Inserm, 37000 Tours, France; (C.V.-D.); (P.V.); (C.R.A.); (H.B.); (P.C.)
- CHU de Tours, Service de Neurologie, 37000 Tours, France
| |
Collapse
|
42
|
Limanaqi F, Biagioni F, Gambardella S, Familiari P, Frati A, Fornai F. Promiscuous Roles of Autophagy and Proteasome in Neurodegenerative Proteinopathies. Int J Mol Sci 2020; 21:E3028. [PMID: 32344772 PMCID: PMC7215558 DOI: 10.3390/ijms21083028] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Alterations in autophagy and the ubiquitin proteasome system (UPS) are commonly implicated in protein aggregation and toxicity which manifest in a number of neurological disorders. In fact, both UPS and autophagy alterations are bound to the aggregation, spreading and toxicity of the so-called prionoid proteins, including alpha synuclein (α-syn), amyloid-beta (Aβ), tau, huntingtin, superoxide dismutase-1 (SOD-1), TAR-DNA-binding protein of 43 kDa (TDP-43) and fused in sarcoma (FUS). Recent biochemical and morphological studies add to this scenario, focusing on the coordinated, either synergistic or compensatory, interplay that occurs between autophagy and the UPS. In fact, a number of biochemical pathways such as mammalian target of rapamycin (mTOR), transcription factor EB (TFEB), Bcl2-associated athanogene 1/3 (BAG3/1) and glycogen synthase kinase beta (GSk3β), which are widely explored as potential targets in neurodegenerative proteinopathies, operate at the crossroad between autophagy and UPS. These biochemical steps are key in orchestrating the specificity and magnitude of the two degradation systems for effective protein homeostasis, while intermingling with intracellular secretory/trafficking and inflammatory pathways. The findings discussed in the present manuscript are supposed to add novel viewpoints which may further enrich our insight on the complex interactions occurring between cell-clearing systems, protein misfolding and propagation. Discovering novel mechanisms enabling a cross-talk between the UPS and autophagy is expected to provide novel potential molecular targets in proteinopathies.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
| | - Francesca Biagioni
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (S.G.); (A.F.)
| | - Stefano Gambardella
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (S.G.); (A.F.)
| | - Pietro Familiari
- Department of Human Neurosciences, Division of Neurosurgery, Sapienza University of Rome, 00185 Roma, Italy;
| | - Alessandro Frati
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (S.G.); (A.F.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (S.G.); (A.F.)
| |
Collapse
|
43
|
Hergesheimer RC, Chami AA, de Assis DR, Vourc'h P, Andres CR, Corcia P, Lanznaster D, Blasco H. The debated toxic role of aggregated TDP-43 in amyotrophic lateral sclerosis: a resolution in sight? Brain 2020; 142:1176-1194. [PMID: 30938443 PMCID: PMC6487324 DOI: 10.1093/brain/awz078] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/11/2019] [Accepted: 02/16/2019] [Indexed: 12/11/2022] Open
Abstract
Transactive response DNA-binding protein-43 (TDP-43) is an RNA/DNA binding protein that forms phosphorylated and ubiquitinated aggregates in the cytoplasm of motor neurons in amyotrophic lateral sclerosis, which is a hallmark of this disease. Amyotrophic lateral sclerosis is a neurodegenerative condition affecting the upper and lower motor neurons. Even though the aggregative property of TDP-43 is considered a cornerstone of amyotrophic lateral sclerosis, there has been major controversy regarding the functional link between TDP-43 aggregates and cell death. In this review, we attempt to reconcile the current literature surrounding this debate by discussing the results and limitations of the published data relating TDP-43 aggregates to cytotoxicity, as well as therapeutic perspectives of TDP-43 aggregate clearance. We point out key data suggesting that the formation of TDP-43 aggregates and the capacity to self-template and propagate among cells as a 'prion-like' protein, another pathological property of TDP-43 aggregates, are a significant cause of motor neuronal death. We discuss the disparities among the various studies, particularly with respect to the type of models and the different forms of TDP-43 used to evaluate cellular toxicity. We also examine how these disparities can interfere with the interpretation of the results pertaining to a direct toxic effect of TDP-43 aggregates. Furthermore, we present perspectives for improving models in order to better uncover the toxic role of aggregated TDP-43. Finally, we review the recent studies on the enhancement of the cellular clearance mechanisms of autophagy, the ubiquitin proteasome system, and endocytosis in an attempt to counteract TDP-43 aggregation-induced toxicity. Altogether, the data available so far encourage us to suggest that the cytoplasmic aggregation of TDP-43 is key for the neurodegeneration observed in motor neurons in patients with amyotrophic lateral sclerosis. The corresponding findings provide novel avenues toward early therapeutic interventions and clinical outcomes for amyotrophic lateral sclerosis management.
Collapse
Affiliation(s)
| | - Anna A Chami
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France
| | | | - Patrick Vourc'h
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France.,CHU de Tours, Service de Biochimie et Biologie Moléculaire, Tours, France
| | - Christian R Andres
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France.,CHU de Tours, Service de Biochimie et Biologie Moléculaire, Tours, France
| | - Philippe Corcia
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France.,CHU de Tours, Service de Neurologie, Tours, France
| | | | - Hélène Blasco
- UMR 1253, iBRAIN, Université de Tours, INSERM, Tours, France.,CHU de Tours, Service de Biochimie et Biologie Moléculaire, Tours, France
| |
Collapse
|
44
|
Xiang C, Zhang Y, Guo W, Liang XJ. Biomimetic carbon nanotubes for neurological disease therapeutics as inherent medication. Acta Pharm Sin B 2020; 10:239-248. [PMID: 32082970 PMCID: PMC7016289 DOI: 10.1016/j.apsb.2019.11.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 01/01/2023] Open
Abstract
Nowadays, nanotechnology is revolutionizing the approaches to different fields from manufacture to health. Carbon nanotubes (CNTs) as promising candidates in nanomedicine have great potentials in developing novel entities for central nervous system pathologies, due to their excellent physicochemical properties and ability to interface with neurons and neuronal circuits. However, most of the studies mainly focused on the drug delivery and bioimaging applications of CNTs, while neglect their application prospects as therapeutic drugs themselves. At present, the relevant reviews are not available yet. Herein we summarized the latest advances on the biomedical and therapeutic applications of CNTs in vitro and in vivo for neurological diseases treatments as inherent therapeutic drugs. The biological mechanisms of CNTs-mediated bio-medical effects and potential toxicity of CNTs were also intensely discussed. It is expected that CNTs will exploit further neurological applications on disease therapy in the near future.
Collapse
Key Words
- AD, Alzheimer's disease
- ALS, amyotrophic lateral sclerosis
- BBB, blood–brain barrier
- CNS, central nervous system
- CNT-N, nitrogen-doped carbon nanotubes
- CNTs, carbon nanotubes
- Carbon nanotubes
- CpG, oligodeoxynucleotides
- DTPA, diethylentriaminepentaacetic
- Drug delivery
- EBs, embryoid bodies
- EDC·HCl, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
- GO, graphene oxide
- HD, Huntington's disease
- Inherent medication
- MCAO, middle cerebral artery occlusion
- METH, methamphetamine
- MPO, myeloperoxidase
- MWCNTTs, multi-walled nanotube towers
- MWCNTs, multi-walled carbon nanotubes
- ND, nanodiamond
- NHS, N-hydroxysuccinimide
- NR, nanorod
- NSCs, neural stem cells
- Nervous system diseases
- PBEC, porcine brain endothelial cells
- PCL, polycaprolactone
- PD, Parkinson's disease
- PEG, polyethylene-glycol
- PET, position emission tomography
- PMo11V, tetrabutylammonium salt of phosphovanadomolybdate
- POCs, polycyclic organic compounds
- PPy/SWCNT, polypyrrole/single-walled carbon nanotube
- RES, reticuloendothelial system
- SWCNTP, single-walled nanotube paper
- SWCNTs, single-walled carbon nanotubes
- TLR9, the toll-like receptor-9
- TMZ, temozolomide
- Therapeutic drug
- Toxicity
- aSWCNTs, aggregated SWCNTs
- f-CNTs, functionalized carbon nanotubes
- hNSCs, human neural stem cells
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Chenyang Xiang
- Translational Medicine Center, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Yuxuan Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Weisheng Guo
- Translational Medicine Center, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Xing-Jie Liang
- Translational Medicine Center, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| |
Collapse
|
45
|
Imamura K, Izumi Y, Banno H, Uozumi R, Morita S, Egawa N, Ayaki T, Nagai M, Nishiyama K, Watanabe Y, Hanajima R, Oki R, Fujita K, Takahashi N, Ikeda T, Shimizu A, Morinaga A, Hirohashi T, Fujii Y, Takahashi R, Inoue H. Induced pluripotent stem cell-based Drug Repurposing for Amyotrophic lateral sclerosis Medicine (iDReAM) study: protocol for a phase I dose escalation study of bosutinib for amyotrophic lateral sclerosis patients. BMJ Open 2019; 9:e033131. [PMID: 31796494 PMCID: PMC7003406 DOI: 10.1136/bmjopen-2019-033131] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a progressive and severe neurodegenerative disease caused by motor neuron death. There have as yet been no fundamental curative medicines, and the development of a medicine for ALS is urgently required. Induced pluripotent stem cell (iPSC)-based drug repurposing identified an Src/c-Abl inhibitor, bosutinib, as a candidate molecular targeted therapy for ALS. The objectives of this study are to evaluate the safety and tolerability of bosutinib for the treatment of patients with ALS and to explore the efficacy of bosutinib on ALS. This study is the first clinical trial of administered bosutinib for patients with ALS. METHODS AND ANALYSIS An open-label, multicentre phase I dose escalation study has been designed. The study consists of a 12-week observation period, a 1-week transitional period, a 12-week study treatment period and a 4-week follow-up period. After completion of the transitional period, subjects whose total ALS Functional Rating Scale-Revised (ALSFRS-R) score decreased by 1-3 points during the 12-week observation period receive bosutinib for 12 weeks. Three to six patients with ALS are enrolled in each of the four bosutinib dose levels (100, 200, 300 or 400 mg/day) to evaluate the safety and tolerability under a 3+3 dose escalation study design. Dose escalation and maximum tolerated dose are determined by the safety assessment committee comprising oncologists/haematologists and neurologists based on the incidence of dose-limiting toxicity in the first 4 weeks of the treatment at each dose level. A recommended phase II dose is determined by the safety assessment committee on completion of the 12-week study treatment in all subjects at all dose levels. The efficacy of bosutinib is also evaluated exploratorily using ALS clinical scores and biomarkers. ETHICS AND DISSEMINATION This study received full ethical approval from the institutional review board of each participating site. The findings of the study will be disseminated in peer-reviewed journals and at scientific conferences. TRIAL REGISTRATION NUMBER UMIN000036295; Pre-results, JMA-IIA00419; Pre-results.
Collapse
Affiliation(s)
- Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Haruhiko Banno
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Ryuji Uozumi
- Department of Biomedical Statistics and Bioinformatics, Kyoto University, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University, Kyoto, Japan
| | - Naohiro Egawa
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makiko Nagai
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazutoshi Nishiyama
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yasuhiro Watanabe
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ryosuke Oki
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koji Fujita
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Takafumi Ikeda
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Akira Shimizu
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | | | | | | | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
46
|
Garcia C, Vidal-Taboada JM, Syriani E, Salvado M, Morales M, Gamez J. Haplotype Analysis of the First A4V- SOD1 Spanish Family: Two Separate Founders or a Single Common Founder? Front Genet 2019; 10:1109. [PMID: 31781168 PMCID: PMC6857184 DOI: 10.3389/fgene.2019.01109] [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: 03/04/2019] [Accepted: 10/16/2019] [Indexed: 12/02/2022] Open
Abstract
Despite the genetic heterogeneity reported in familial amyotrophic lateral sclerosis (ALS) (fALS), Cu/Zn superoxide-dismutase (SOD1) gene mutations are the second most common cause of the disease, accounting for around 20% of all families (ALS1) and isolated sporadic cases (sALS). At least 186 different mutations in the SOD1 gene have been reported to date. The possibility of a single founder and separate founders have been investigated for D90A (p.D91A) and A4V (p.A5V), the most common mutations worldwide. High-throughput single nucleotide polymorphism genotyping studies have suggested two founders for A4V (one for the Amerindian population and another for the European population) although the possibility that the two populations are descended from a single ancient founder cannot be ruled out. We used 15 genetic variants spanning the human chromosome 21 from the SOD1 gene to the SCAF4 gene, comparing them with the population reference panels, to demonstrate that the first A4V Spanish pedigree shared the genetic background reported in the European population.
Collapse
Affiliation(s)
- Cecilia Garcia
- ALS Unit, Neurology Department, Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain
| | - Jose Manuel Vidal-Taboada
- ALS Unit, Neurology Department, Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain
| | - Enrique Syriani
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain
| | - Maria Salvado
- ALS Unit, Neurology Department, Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Miguel Morales
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain
| | - Josep Gamez
- ALS Unit, Neurology Department, Vall d'Hebron University Hospital, Barcelona, Spain.,Vall d'Hebron Research Institute (VHIR), Barcelona, Spain.,European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
47
|
Maurel C, Chami AA, Thépault RA, Marouillat S, Blasco H, Corcia P, Andres CR, Vourc'h P. A role for SUMOylation in the Formation and Cellular Localization of TDP-43 Aggregates in Amyotrophic Lateral Sclerosis. Mol Neurobiol 2019; 57:1361-1373. [PMID: 31728929 DOI: 10.1007/s12035-019-01810-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022]
Abstract
In amyotrophic lateral sclerosis, motor neurons undergoing degeneration are characterized by the presence of cytoplasmic aggregates containing TDP-43 protein. SUMOylation, a posttranslational modification of proteins, has been previously implicated in the formation of aggregates positives for SOD1, another protein enriched in a subset of ALS patients. We show in this study that TDP-43 is also a target of SUMOylation. The inhibition of the first step of the SUMOylation process by anacardic acid significantly reduces the presence of TDP-43 aggregates and improves neuritogenesis and cell viability in vitro. Interestingly, the mutation of the unique SUMOylation site on TDP-43, using site-directed mutagenesis, modifies the intracellular localization of TDP-43 aggregates. Instead of being cytoplasmic where they are associated with toxic effects, they are located inside the nucleus. This change of localization results in improvement in cell viability and in global cellular functions. Our results implicate the SUMOylation site of TDP-43 in the formation of cytoplasmic TDP-43 aggregates, a hallmark of ALS, and thus identifies this region as a new target for novel therapeutic strategies.
Collapse
Affiliation(s)
- Cindy Maurel
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| | - Anna A Chami
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | | | - Hélène Blasco
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37044, Tours, France
| | - Philippe Corcia
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Service de Neurologie, CHRU de Tours, 37044, Tours, France
| | - Christian R Andres
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37044, Tours, France
| | - Patrick Vourc'h
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37044, Tours, France
| |
Collapse
|
48
|
Halpern M, Brennand KJ, Gregory J. Examining the relationship between astrocyte dysfunction and neurodegeneration in ALS using hiPSCs. Neurobiol Dis 2019; 132:104562. [PMID: 31381978 DOI: 10.1016/j.nbd.2019.104562] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/28/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a complex and fatal neurodegenerative disease for which the causes of disease onset and progression remain unclear. Recent advances in human induced pluripotent stem cell (hiPSC)-based models permit the study of the genetic factors associated with ALS in patient-derived neural cell types, including motor neurons and glia. While astrocyte dysfunction has traditionally been thought to exacerbate disease progression, astrocytic dysfunction may play a more direct role in disease initiation and progression. Such non-cell autonomous mechanisms expand the potential targets of therapeutic intervention, but only a handful of ALS risk-associated genes have been examined for their impact on astrocyte dysfunction and neurodegeneration. This review summarizes what is currently known about astrocyte function in ALS and suggests ways in which hiPSC-based models can be used to more effectively study the role of astrocytes in neurodegenerative disease.
Collapse
Affiliation(s)
- Madeline Halpern
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Kristen J Brennand
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America.
| | - James Gregory
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, United States of America.
| |
Collapse
|
49
|
RNA Granules and Their Role in Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1203:195-245. [DOI: 10.1007/978-3-030-31434-7_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
Ma L, Shi Y, Chen Z, Li S, Zhang J. A novel SETX gene mutation associated with Juvenile amyotrophic lateral sclerosis. Brain Behav 2018; 8:e01066. [PMID: 30052327 PMCID: PMC6160657 DOI: 10.1002/brb3.1066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/24/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023] Open
Abstract
OBJECTS This study aimed to report a novel point mutation associated with juvenile amyotrophic lateral sclerosis (JALS) in a Chinese Han family. METHODS Detailed clinical assessment was applied to two patients, including proband (II-2) and his mother (I-2). Next-generation sequencing (NGS), also known as high-throughput sequencing in whole exon sequence, was performed in the proband to reach the target region. Sanger sequencing was also used to detect DNA sequence variants of the proband and other three members of his family. RESULTS The proband (II-2) and his mother (I-2) were successfully diagnosed according to the clinical manifestations and physical examination. A novel point mutation c.1157T > C in the exon 10 of the SETX gene was identified in II-2 and I-2, resulting in a substitution of methionine (ATG) to threonine (ACG). However, we ultimately did not find the same variant in the other two normal members of his family in addition to 100 unrelated normal subjects. CONCLUSION We presented a novel probably pathogenic missense mutation in exon 10 of SETX gene in a Chinese Han family with JALS.
Collapse
Affiliation(s)
- Limin Ma
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingying Shi
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhongcan Chen
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Shujian Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Jiewen Zhang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|