51
|
Anakor E, Le Gall L, Dumonceaux J, Duddy WJ, Duguez S. Exosomes in Ageing and Motor Neurone Disease: Biogenesis, Uptake Mechanisms, Modifications in Disease and Uses in the Development of Biomarkers and Therapeutics. Cells 2021; 10:2930. [PMID: 34831153 PMCID: PMC8616058 DOI: 10.3390/cells10112930] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/07/2023] Open
Abstract
Intercellular communication between neurons and their surrounding cells occurs through the secretion of soluble molecules or release of vesicles such as exosomes into the extracellular space, participating in brain homeostasis. Under neuro-degenerative conditions associated with ageing, such as amyotrophic lateral sclerosis (ALS), Alzheimer's or Parkinson's disease, exosomes are suspected to propagate toxic proteins. The topic of this review is the role of exosomes in ageing conditions and more specifically in ALS. Our current understanding of exosomes and exosome-related mechanisms is first summarized in a general sense, including their biogenesis and secretion, heterogeneity, cellular interaction and intracellular fate. Their role in the Central Nervous System (CNS) and ageing of the neuromotor system is then considered in the context of exosome-induced signaling. The review then focuses on exosomes in age-associated neurodegenerative disease. The role of exosomes in ALS is highlighted, and their use as potential biomarkers to diagnose and prognose ALS is presented. The therapeutic implications of exosomes for ALS are considered, whether as delivery vehicles, neurotoxic targets or as corrective drugs in and of themselves. A diverse set of mechanisms underpin the functional roles, both confirmed and potential, of exosomes, generally in ageing and specifically in motor neurone disease. Aspects of their contents, biogenesis, uptake and modifications offer many plausible routes towards the development of novel biomarkers and therapeutics.
Collapse
Affiliation(s)
- Ekene Anakor
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry-Londonderry BT47 6SB, UK; (E.A.); (L.L.G.); (J.D.); (W.J.D.)
| | - Laura Le Gall
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry-Londonderry BT47 6SB, UK; (E.A.); (L.L.G.); (J.D.); (W.J.D.)
- NIHR Biomedical Research Centre, Great Ormond Street Institute of Child Health, Great Ormond Street Hospital NHS Trust, University College London, London WC1N 1EH, UK
| | - Julie Dumonceaux
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry-Londonderry BT47 6SB, UK; (E.A.); (L.L.G.); (J.D.); (W.J.D.)
- NIHR Biomedical Research Centre, Great Ormond Street Institute of Child Health, Great Ormond Street Hospital NHS Trust, University College London, London WC1N 1EH, UK
| | - William John Duddy
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry-Londonderry BT47 6SB, UK; (E.A.); (L.L.G.); (J.D.); (W.J.D.)
| | - Stephanie Duguez
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry-Londonderry BT47 6SB, UK; (E.A.); (L.L.G.); (J.D.); (W.J.D.)
| |
Collapse
|
52
|
Lin F, Lin W, Zhu C, Lin J, Zhu J, Li XY, Wang Z, Wang C, Huang H. Sequencing of neurofilament genes identified NEFH Ser787Arg as a novel risk variant of sporadic amyotrophic lateral sclerosis in Chinese subjects. BMC Med Genomics 2021; 14:222. [PMID: 34511133 PMCID: PMC8436554 DOI: 10.1186/s12920-021-01073-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/01/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease with neuronal cell inclusions composed of neurofilaments and other abnormal aggregative proteins as pathological hallmarks. Approximately 90% of patients have sporadic cases (sALS), and at least 4 genes, i.e. C9orf72, SOD1, FUS and TARDBP, have been identified as the main causative genes, while many others have been proposed as potential risk genes. However, these mutations could explain only ~ 10% of sALS cases. The neurofilament polypeptides encoded by NEFH, NEFM, and NEFL are promising protein biomarkers for ALS and other degenerative diseases. However, whether the genetic variants of these genes were associated with ALS remain ambiguous. METHODS Here, we used PCR-Sanger to sequence the exons of these three genes in a cohort of 371 sALS patients and 711 healthy controls (Phase I) and validated the risk variant in another 300 sALS patients and 1076 controls (Phase II). RESULTS A total of 92 variants were identified, including 36 rare heterozygous variants in NEFH, 27 in NEFM, and 16 in NEFL, and only rs568759161 (p.Ser787Arg) in NEFH reached nominal statistical power (P = 0.02 at Phase I, P = 0.009 at Phase II) in the case-control comparison. Together, the Phase I and II studies showed the significantly higher frequency of the variant in cases (9/1342, 0.67%) than in controls (2/3574, 0.07%) (OR 12.06; 95% CI 2.60-55.88; P = 0.0003). No variants passed multiple testing in the discovery cohort, but rs568759161 was associated with ALS in a replication cohort. CONCLUSIONS Our results confirmed that NEFH Ser787Arg is a novel sALS risk variant in Chinese subjects, but NEFM and NEFL were not associated with sALS. These data may have implications for genetic counselling and for understanding the pathogenesis of sALS.
Collapse
Affiliation(s)
- Feng Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fujian, 350001, China
| | - Wanhui Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fujian, 350001, China
| | - Chaofeng Zhu
- Department of Neurology, Fujian Medical University Union Hospital, Fujian, 350001, China
| | - Jilan Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fujian, 350001, China
| | - Junge Zhu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Xu-Ying Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Zhanjun Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Huapin Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fujian, 350001, China.
| |
Collapse
|
53
|
The Way forward for the Origin of Life: Prions and Prion-Like Molecules First Hypothesis. Life (Basel) 2021; 11:life11090872. [PMID: 34575021 PMCID: PMC8467930 DOI: 10.3390/life11090872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/11/2021] [Accepted: 08/22/2021] [Indexed: 11/18/2022] Open
Abstract
In this paper the hypothesis that prions and prion-like molecules could have initiated the chemical evolutionary process which led to the eventual emergence of life is reappraised. The prions first hypothesis is a specific application of the protein-first hypothesis which asserts that protein-based chemical evolution preceded the evolution of genetic encoding processes. This genetics-first hypothesis asserts that an “RNA-world era” came before protein-based chemical evolution and rests on a singular premise that molecules such as RNA, acetyl-CoA, and NAD are relics of a long line of chemical evolutionary processes preceding the Last Universal Common Ancestor (LUCA). Nevertheless, we assert that prions and prion-like molecules may also be relics of chemical evolutionary processes preceding LUCA. To support this assertion is the observation that prions and prion-like molecules are involved in a plethora of activities in contemporary biology in both complex (eukaryotes) and primitive life forms. Furthermore, a literature survey reveals that small RNA virus genomes harbor information about prions (and amyloids). If, as has been presumed by proponents of the genetics-first hypotheses, small viruses were present during an RNA world era and were involved in some of the earliest evolutionary processes, this places prions and prion-like molecules potentially at the heart of the chemical evolutionary process whose eventual outcome was life. We deliberate on the case for prions and prion-like molecules as the frontier molecules at the dawn of evolution of living systems.
Collapse
|
54
|
Cathcart SJ, Appel SH, Peterson LE, Greene EP, Powell SZ, Arumanayagam AS, Rivera AL, Cykowski MD. Fast Progression in Amyotrophic Lateral Sclerosis Is Associated With Greater TDP-43 Burden in Spinal Cord. J Neuropathol Exp Neurol 2021; 80:754-763. [PMID: 34383907 PMCID: PMC8433592 DOI: 10.1093/jnen/nlab061] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Upper and lower motor neuron pathologies are critical to the autopsy diagnosis of amyotrophic lateral sclerosis (ALS). Further investigation is needed to determine how the relative burden of these pathologies affects the disease course. We performed a blinded, retrospective study of 38 ALS patients, examining the association between pathologic measures in motor cortex, hypoglossal nucleus, and lumbar cord with clinical data, including progression rate and disease duration, site of symptom onset, and upper and lower motor neuron signs. The most critical finding in our study was that TAR DNA-binding protein 43 kDa (TDP-43) pathologic burden in lumbar cord and hypoglossal nucleus was significantly associated with a faster progression rate with reduced survival (p < 0.02). There was no correlation between TDP-43 burden and the severity of cell loss, and no significant clinical associations were identified for motor cortex TDP-43 burden or severity of cell loss in motor cortex. C9orf72 expansion was associated with shorter disease duration (p < 0.001) but was not significantly associated with pathologic measures in these regions. The association between lower motor neuron TDP-43 burden and fast progression with reduced survival in ALS provides further support for the study of TDP-43 as a disease biomarker.
Collapse
Affiliation(s)
- Sahara J Cathcart
- From the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA (SJC, SZP, ASA, ALR, MDC); University of Nebraska Medical Center, Omaha, Nebraska, USA (SJC); Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, MDC); Institute of Academic Medicine at the Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR, MDC); Houston Methodist Neurological Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR); NXG Logic, LLC, Houston, Texas, USA (LEP)
| | - Stanley H Appel
- From the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA (SJC, SZP, ASA, ALR, MDC); University of Nebraska Medical Center, Omaha, Nebraska, USA (SJC); Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, MDC); Institute of Academic Medicine at the Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR, MDC); Houston Methodist Neurological Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR); NXG Logic, LLC, Houston, Texas, USA (LEP)
| | - Leif E Peterson
- From the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA (SJC, SZP, ASA, ALR, MDC); University of Nebraska Medical Center, Omaha, Nebraska, USA (SJC); Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, MDC); Institute of Academic Medicine at the Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR, MDC); Houston Methodist Neurological Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR); NXG Logic, LLC, Houston, Texas, USA (LEP)
| | - Ericka P Greene
- From the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA (SJC, SZP, ASA, ALR, MDC); University of Nebraska Medical Center, Omaha, Nebraska, USA (SJC); Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, MDC); Institute of Academic Medicine at the Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR, MDC); Houston Methodist Neurological Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR); NXG Logic, LLC, Houston, Texas, USA (LEP)
| | - Suzanne Z Powell
- From the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA (SJC, SZP, ASA, ALR, MDC); University of Nebraska Medical Center, Omaha, Nebraska, USA (SJC); Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, MDC); Institute of Academic Medicine at the Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR, MDC); Houston Methodist Neurological Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR); NXG Logic, LLC, Houston, Texas, USA (LEP)
| | - Anithachristy S Arumanayagam
- From the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA (SJC, SZP, ASA, ALR, MDC); University of Nebraska Medical Center, Omaha, Nebraska, USA (SJC); Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, MDC); Institute of Academic Medicine at the Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR, MDC); Houston Methodist Neurological Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR); NXG Logic, LLC, Houston, Texas, USA (LEP)
| | - Andreana L Rivera
- From the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA (SJC, SZP, ASA, ALR, MDC); University of Nebraska Medical Center, Omaha, Nebraska, USA (SJC); Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, MDC); Institute of Academic Medicine at the Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR, MDC); Houston Methodist Neurological Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR); NXG Logic, LLC, Houston, Texas, USA (LEP)
| | - Matthew D Cykowski
- From the Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA (SJC, SZP, ASA, ALR, MDC); University of Nebraska Medical Center, Omaha, Nebraska, USA (SJC); Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, MDC); Institute of Academic Medicine at the Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR, MDC); Houston Methodist Neurological Institute, Houston Methodist Hospital, Houston, Texas, USA (SHA, EPG, SZP, ALR); NXG Logic, LLC, Houston, Texas, USA (LEP)
| |
Collapse
|
55
|
Higher incidence of cervical spinal cord compression in amyotrophic lateral sclerosis: a single-institute cohort study. Neurol Sci 2021; 43:1079-1086. [PMID: 34287724 DOI: 10.1007/s10072-021-05465-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/06/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Although the relationship between amyotrophic lateral sclerosis (ALS) and cervical spondylotic myelopathy (CSM) is important, data relating to CSM complications in ALS remain lacking. PURPOSE We aimed to investigate and validate the spinal cord conditions of ALS patients. MATERIALS AND METHODS We recruited all patients diagnosed with ALS, Parkinson's disease (PD), or chronic inflammatory demyelinating polyneuropathy (CIDP) who were admitted to our department from April 1, 2017, to March 31, 2020. We analyzed the cervical or thoracolumbar magnetic resonance imaging (MRI) scans of these 128 patients. Data relating to spondylosis, cord compression, spinal canal diameter, spinal cord diameter, and the closest distance between the cervical spinal canal and cord were validated using MRI. RESULTS Of the 128 patients, 52 had ALS, 48 had PD, and 28 had CIDP. The proportions of both cervical spondylosis and cervical cord compression were highest in the ALS group compared with the other patient groups (p < 0.05). The proportion of cervical spondylosis in ALS patients reached 38.3%, and that of cervical cord compression reached 53.2%. The closest distance between the cervical spinal canal and cord was also significantly smaller in ALS patients compared with CIDP patients (p < 0.05). In contrast to the cervical cord findings, there were no significant differences in the thoracolumbar cord between ALS patients and the other patient groups. CONCLUSIONS Of the three disease groups, the proportion of CSM was highest in ALS patients. Furthermore, cervical cord conditions were significantly more crowded in the ALS patients than in the other patient groups.
Collapse
|
56
|
Pikatza-Menoio O, Elicegui A, Bengoetxea X, Naldaiz-Gastesi N, López de Munain A, Gerenu G, Gil-Bea FJ, Alonso-Martín S. The Skeletal Muscle Emerges as a New Disease Target in Amyotrophic Lateral Sclerosis. J Pers Med 2021; 11:671. [PMID: 34357138 PMCID: PMC8307751 DOI: 10.3390/jpm11070671] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 01/02/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons (MNs) and severe muscle atrophy without effective treatment. Most research on ALS has been focused on the study of MNs and supporting cells of the central nervous system. Strikingly, the recent observations of pathological changes in muscle occurring before disease onset and independent from MN degeneration have bolstered the interest for the study of muscle tissue as a potential target for delivery of therapies for ALS. Skeletal muscle has just been described as a tissue with an important secretory function that is toxic to MNs in the context of ALS. Moreover, a fine-tuning balance between biosynthetic and atrophic pathways is necessary to induce myogenesis for muscle tissue repair. Compromising this response due to primary metabolic abnormalities in the muscle could trigger defective muscle regeneration and neuromuscular junction restoration, with deleterious consequences for MNs and thereby hastening the development of ALS. However, it remains puzzling how backward signaling from the muscle could impinge on MN death. This review provides a comprehensive analysis on the current state-of-the-art of the role of the skeletal muscle in ALS, highlighting its contribution to the neurodegeneration in ALS through backward-signaling processes as a newly uncovered mechanism for a peripheral etiopathogenesis of the disease.
Collapse
Affiliation(s)
- Oihane Pikatza-Menoio
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Amaia Elicegui
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Xabier Bengoetxea
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
| | - Neia Naldaiz-Gastesi
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Adolfo López de Munain
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
- Department of Neurology, Donostialdea Integrated Health Organization, Osakidetza Basque Health Service, 20014 Donostia/San Sebastián, Spain
- Department of Neurosciences, Faculty of Medicine and Nursery, University of the Basque Country UPV-EHU, 20014 Donostia/San Sebastián, Spain
| | - Gorka Gerenu
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
- Department of Physiology, University of the Basque Country UPV-EHU, 48940 Leioa, Spain
| | - Francisco Javier Gil-Bea
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Sonia Alonso-Martín
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| |
Collapse
|
57
|
Sprunger ML, Jackrel ME. Prion-Like Proteins in Phase Separation and Their Link to Disease. Biomolecules 2021; 11:biom11071014. [PMID: 34356638 PMCID: PMC8301953 DOI: 10.3390/biom11071014] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 02/01/2023] Open
Abstract
Aberrant protein folding underpins many neurodegenerative diseases as well as certain myopathies and cancers. Protein misfolding can be driven by the presence of distinctive prion and prion-like regions within certain proteins. These prion and prion-like regions have also been found to drive liquid-liquid phase separation. Liquid-liquid phase separation is thought to be an important physiological process, but one that is prone to malfunction. Thus, aberrant liquid-to-solid phase transitions may drive protein aggregation and fibrillization, which could give rise to pathological inclusions. Here, we review prions and prion-like proteins, their roles in phase separation and disease, as well as potential therapeutic approaches to counter aberrant phase transitions.
Collapse
|
58
|
Singh A, Dawson TM, Kulkarni S. Neurodegenerative disorders and gut-brain interactions. J Clin Invest 2021; 131:e143775. [PMID: 34196307 DOI: 10.1172/jci143775] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative disorders (NDs) affect essential functions not only in the CNS, but also cause persistent gut dysfunctions, suggesting that they have an impact on both CNS and gut-innervating neurons. Although the CNS biology of NDs continues to be well studied, how gut-innervating neurons, including those that connect the gut to the brain, are affected by or involved in the etiology of these debilitating and progressive disorders has been understudied. Studies in recent years have shown how CNS and gut biology, aided by the gut-brain connecting neurons, modulate each other's functions. These studies underscore the importance of exploring the gut-innervating and gut-brain connecting neurons of the CNS and gut function in health, as well as the etiology and progression of dysfunction in NDs. In this Review, we discuss our current understanding of how the various gut-innervating neurons and gut physiology are involved in the etiology of NDs, including Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis, to cause progressive CNS and persistent gut dysfunction.
Collapse
Affiliation(s)
- Alpana Singh
- Center for Neurogastroenterology, Division of Gastroenterology and Hepatology, Department of Medicine
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering.,Department of Neurology.,Solomon H. Snyder Department of Neuroscience, and.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana, USA
| | - Subhash Kulkarni
- Center for Neurogastroenterology, Division of Gastroenterology and Hepatology, Department of Medicine
| |
Collapse
|
59
|
Keskin I, Ekhtiari Bidhendi E, Marklund M, Andersen PM, Brännström T, Marklund SL, Nordström U. Peripheral administration of SOD1 aggregates does not transmit pathogenic aggregation to the CNS of SOD1 transgenic mice. Acta Neuropathol Commun 2021; 9:111. [PMID: 34158126 PMCID: PMC8220797 DOI: 10.1186/s40478-021-01211-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/07/2021] [Indexed: 01/02/2023] Open
Abstract
The deposition of aggregated proteins is a common neuropathological denominator for neurodegenerative disorders. Experimental evidence suggests that disease propagation involves prion-like mechanisms that cause the spreading of template-directed aggregation of specific disease-associated proteins. In transgenic (Tg) mouse models of superoxide dismutase-1 (SOD1)-linked amyotrophic lateral sclerosis (ALS), inoculation of minute amounts of human SOD1 (hSOD1) aggregates into the spinal cord or peripheral nerves induces premature ALS-like disease and template-directed hSOD1 aggregation that spreads along the neuroaxis. This infectious nature of spreading pathogenic aggregates might have implications for the safety of laboratory and medical staff, recipients of donated blood or tissue, or possibly close relatives and caregivers. Here we investigate whether transmission of ALS-like disease is unique to the spinal cord and peripheral nerve inoculations or if hSOD1 aggregation might spread from the periphery into the central nervous system (CNS). We inoculated hSOD1 aggregate seeds into the peritoneal cavity, hindlimb skeletal muscle or spinal cord of adult Tg mice expressing mutant hSOD1. Although we used up to 8000 times higher dose—compared to the lowest dose transmitting disease in spinal cord inoculations—the peripheral inoculations did not transmit seeded aggregation to the CNS or premature ALS-like disease in hSOD1 Tg mice. Nor was any hSOD1 aggregation detected in the liver, kidney, skeletal muscle or sciatic nerve. To explore potential reasons for the lack of disease transmission, we examined the stability of hSOD1 aggregates and found them to be highly vulnerable to both proteases and detergent. Our findings suggest that exposed individuals and personnel handling samples from ALS patients are at low risk of any potential transmission of seeded hSOD1 aggregation.
Collapse
|
60
|
Bacterial Extracellular DNA Promotes β-Amyloid Aggregation. Microorganisms 2021; 9:microorganisms9061301. [PMID: 34203755 PMCID: PMC8232312 DOI: 10.3390/microorganisms9061301] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/05/2021] [Accepted: 06/11/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease is associated with prion-like aggregation of the amyloid β (Aβ) peptide and the subsequent accumulation of misfolded neurotoxic aggregates in the brain. Therefore, it is critical to clearly identify the factors that trigger the cascade of Aβ misfolding and aggregation. Numerous studies have pointed out the association between microorganisms and their virulence factors and Alzheimer’s disease; however, their exact mechanisms of action remain unclear. Recently, we discovered a new pathogenic role of bacterial extracellular DNA, triggering the formation of misfolded Tau aggregates. In this study, we investigated the possible role of DNA extracted from different bacterial and eukaryotic cells in triggering Aβ aggregation in vitro. Interestingly, we found that the extracellular DNA of some, but not all, bacteria is an effective trigger of Aβ aggregation. Furthermore, the acceleration of Aβ nucleation and elongation can vary based on the concentration of the bacterial DNA and the bacterial strain from which this DNA had originated. Our findings suggest that bacterial extracellular DNA might play a previously overlooked role in the Aβ protein misfolding associated with Alzheimer’s disease pathogenesis. Moreover, it highlights a new mechanism of how distantly localized bacteria can remotely contribute to protein misfolding and diseases associated with this process. These findings might lead to the use of bacterial DNA as a novel therapeutic target for the prevention and treatment of Alzheimer’s disease.
Collapse
|
61
|
Spasić S, Nikolić-Kokić A, Miletić S, Oreščanin-Dušić Z, Spasić MB, Blagojević D, Stević Z. Edaravone May Prevent Ferroptosis in ALS. Curr Drug Targets 2021; 21:776-780. [PMID: 32077821 DOI: 10.2174/1389450121666200220123305] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022]
Abstract
Radicava™ (Edaravone) was approved the Food and Drug Administration (FDA) as a new treatment for amyotrophic lateral sclerosis (ALS). Edaravone is a synthetic antioxidant that specifically targets oxidative damage interacting with lipid radicals in the cell. In ALS disease the multiple cell types are involved in devastating loss of motor neurons. Mutations and biochemical changes in various cell types jointly contribute to motor neuron death, disease onset, and disease progression. The overall mechanism of neurodegeneration in ALS is still not completely understood. Dying motor neurons have been reported to exhibit features of apoptosis. However, non-apoptotic features of dying motor neurons have also been reported such as ferroptosis. The role of Edaravone in the prevention of ferroptosis in parallel with other therapeutic approaches to ALS therapy is discussed.
Collapse
Affiliation(s)
- Snežana Spasić
- Department of Chemistry, Institute of Chemistry, Technology and Metallurgy, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Nikolić-Kokić
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Srđan Miletić
- Department of Chemistry, Institute of Chemistry, Technology and Metallurgy, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Zorana Oreščanin-Dušić
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Mihajlo B Spasić
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Duško Blagojević
- Department of Physiology, Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Zorica Stević
- Clinic of Neurology, Clinical Center of Serbia, School of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
62
|
Gonçalves PB, Sodero ACR, Cordeiro Y. Green Tea Epigallocatechin-3-gallate (EGCG) Targeting Protein Misfolding in Drug Discovery for Neurodegenerative Diseases. Biomolecules 2021; 11:767. [PMID: 34065606 PMCID: PMC8160836 DOI: 10.3390/biom11050767] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 12/15/2022] Open
Abstract
The potential to treat neurodegenerative diseases (NDs) of the major bioactive compound of green tea, epigallocatechin-3-gallate (EGCG), is well documented. Numerous findings now suggest that EGCG targets protein misfolding and aggregation, a common cause and pathological mechanism in many NDs. Several studies have shown that EGCG interacts with misfolded proteins such as amyloid beta-peptide (Aβ), linked to Alzheimer's disease (AD), and α-synuclein, linked to Parkinson's disease (PD). To date, NDs constitute a serious public health problem, causing a financial burden for health care systems worldwide. Although current treatments provide symptomatic relief, they do not stop or even slow the progression of these devastating disorders. Therefore, there is an urgent need to develop effective drugs for these incurable ailments. It is expected that targeting protein misfolding can serve as a therapeutic strategy for many NDs since protein misfolding is a common cause of neurodegeneration. In this context, EGCG may offer great potential opportunities in drug discovery for NDs. Therefore, this review critically discusses the role of EGCG in NDs drug discovery and provides updated information on the scientific evidence that EGCG can potentially be used to treat many of these fatal brain disorders.
Collapse
Affiliation(s)
| | | | - Yraima Cordeiro
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro 21949-900, Brazil; (P.B.G.); (A.C.R.S.)
| |
Collapse
|
63
|
Ediriweera GR, Chen L, Yerbury JJ, Thurecht KJ, Vine KL. Non-Viral Vector-Mediated Gene Therapy for ALS: Challenges and Future Perspectives. Mol Pharm 2021; 18:2142-2160. [PMID: 34010004 DOI: 10.1021/acs.molpharmaceut.1c00297] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease, for which no effective treatment is yet available to either slow or terminate it. Recent advances in gene therapy renew hope for developing an effective approach to control this disease. Non-viral vectors, such as lipid- and polymer-based nanoparticles, cationic polymers, and exosomes, can effectively transfer genes into primary neurons. The resulting gene expression can be long-term, stable, and without immunological complications, which is essential for the effective management of neurological disorders. This Review will first describe the current research and clinical stage of novel therapies for ALS. It will then touch on the journey of non-viral vector use in ALS, subsequently highlighting the application of non-viral vector-mediated gene therapy. The bottlenecks in the translation of non-viral vectors for ALS treatment are also discussed, including the biological barriers of systemic administration and the issues of "when, where, and how much?" for effective gene delivery. The prospect of employing emerging techniques, such as CRISPR-Cas9 gene editing, stem cell methodology, and low-intensity focused ultrasound for fueling the transport of non-viral vectors to the central nervous system for personalized gene therapy, is briefly discussed in the context of ALS. Despite the challenging road that lies ahead, with the current expansion in interest and technological advancement in non-viral vector-delivered gene therapy for ALS, we hold hope that the field is headed toward a positive future.
Collapse
Affiliation(s)
- Gayathri R Ediriweera
- Centre for Advanced Imaging and Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Liyu Chen
- Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Justin J Yerbury
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,School of Chemistry and Molecular Bioscience, Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging and Australian Institute for Bioengineering & Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kara L Vine
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,School of Chemistry and Molecular Bioscience, Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
64
|
Cicardi ME, Marrone L, Azzouz M, Trotti D. Proteostatic imbalance and protein spreading in amyotrophic lateral sclerosis. EMBO J 2021; 40:e106389. [PMID: 33792056 PMCID: PMC8126909 DOI: 10.15252/embj.2020106389] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/18/2020] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder whose exact causative mechanisms are still under intense investigation. Several lines of evidence suggest that the anatomical and temporal propagation of pathological protein species along the neural axis could be among the main driving mechanisms for the fast and irreversible progression of ALS pathology. Many ALS-associated proteins form intracellular aggregates as a result of their intrinsic prion-like properties and/or following impairment of the protein quality control systems. During the disease course, these mutated proteins and aberrant peptides are released in the extracellular milieu as soluble or aggregated forms through a variety of mechanisms. Internalization by recipient cells may seed further aggregation and amplify existing proteostatic imbalances, thus triggering a vicious cycle that propagates pathology in vulnerable cells, such as motor neurons and other susceptible neuronal subtypes. Here, we provide an in-depth review of ALS pathology with a particular focus on the disease mechanisms of seeding and transmission of the most common ALS-associated proteins, including SOD1, FUS, TDP-43, and C9orf72-linked dipeptide repeats. For each of these proteins, we report historical, biochemical, and pathological evidence of their behaviors in ALS. We further discuss the possibility to harness pathological proteins as biomarkers and reflect on the implications of these findings for future research.
Collapse
Affiliation(s)
- Maria Elena Cicardi
- Department of NeuroscienceWeinberg ALS CenterVickie and Jack Farber Institute for NeuroscienceThomas Jefferson UniversityPhiladelphiaPAUSA
| | - Lara Marrone
- Department of NeuroscienceSheffield Institute for Translational Neuroscience (SITraN)University of SheffieldSheffieldUK
| | - Mimoun Azzouz
- Department of NeuroscienceSheffield Institute for Translational Neuroscience (SITraN)University of SheffieldSheffieldUK
| | - Davide Trotti
- Department of NeuroscienceWeinberg ALS CenterVickie and Jack Farber Institute for NeuroscienceThomas Jefferson UniversityPhiladelphiaPAUSA
| |
Collapse
|
65
|
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: 1.3] [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
|
66
|
Wood A, Gurfinkel Y, Polain N, Lamont W, Lyn Rea S. Molecular Mechanisms Underlying TDP-43 Pathology in Cellular and Animal Models of ALS and FTLD. Int J Mol Sci 2021; 22:4705. [PMID: 33946763 PMCID: PMC8125728 DOI: 10.3390/ijms22094705] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/22/2021] [Accepted: 04/28/2021] [Indexed: 02/03/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are neurodegenerative disorders that exist on a disease spectrum due to pathological, clinical and genetic overlap. In up to 97% of ALS cases and ~50% of FTLD cases, the primary pathological protein observed in affected tissues is TDP-43, which is hyperphosphorylated, ubiquitinated and cleaved. The TDP-43 is observed in aggregates that are abnormally located in the cytoplasm. The pathogenicity of TDP-43 cytoplasmic aggregates may be linked with both a loss of nuclear function and a gain of toxic functions. The cellular processes involved in ALS and FTLD disease pathogenesis include changes to RNA splicing, abnormal stress granules, mitochondrial dysfunction, impairments to axonal transport and autophagy, abnormal neuromuscular junctions, endoplasmic reticulum stress and the subsequent induction of the unfolded protein response. Here, we review and discuss the evidence for alterations to these processes that have been reported in cellular and animal models of TDP-43 proteinopathy.
Collapse
Affiliation(s)
- Alistair Wood
- School of Molecular Science, University of Western Australia, Nedlands 6009, Australia;
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Health Research Building, Discovery Way, Murdoch 6150, Australia; (Y.G.); (N.P.)
| | - Yuval Gurfinkel
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Health Research Building, Discovery Way, Murdoch 6150, Australia; (Y.G.); (N.P.)
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands 6009, Australia;
| | - Nicole Polain
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Health Research Building, Discovery Way, Murdoch 6150, Australia; (Y.G.); (N.P.)
| | - Wesley Lamont
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Nedlands 6009, Australia;
| | - Sarah Lyn Rea
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Health Research Building, Discovery Way, Murdoch 6150, Australia; (Y.G.); (N.P.)
- Hub for Immersive Visualisation and eResearch, Curtin University, Bentley 6102, Australia
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands 6009, Australia
| |
Collapse
|
67
|
Cheng F, De Luca A, Hogan AL, Rayner SL, Davidson JM, Watchon M, Stevens CH, Muñoz SS, Ooi L, Yerbury JJ, Don EK, Fifita JA, Villalva MD, Suddull H, Chapman TR, Hedl TJ, Walker AK, Yang S, Morsch M, Shi B, Blair IP, Laird AS, Chung RS, Lee A. Unbiased Label-Free Quantitative Proteomics of Cells Expressing Amyotrophic Lateral Sclerosis (ALS) Mutations in CCNF Reveals Activation of the Apoptosis Pathway: A Workflow to Screen Pathogenic Gene Mutations. Front Mol Neurosci 2021; 14:627740. [PMID: 33986643 PMCID: PMC8111008 DOI: 10.3389/fnmol.2021.627740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
The past decade has seen a rapid acceleration in the discovery of new genetic causes of ALS, with more than 20 putative ALS-causing genes now cited. These genes encode proteins that cover a diverse range of molecular functions, including free radical scavenging (e.g., SOD1), regulation of RNA homeostasis (e.g., TDP-43 and FUS), and protein degradation through the ubiquitin-proteasome system (e.g., ubiquilin-2 and cyclin F) and autophagy (TBK1 and sequestosome-1/p62). It is likely that the various initial triggers of disease (either genetic, environmental and/or gene-environment interaction) must converge upon a common set of molecular pathways that underlie ALS pathogenesis. Given the complexity, it is not surprising that a catalog of molecular pathways and proteostasis dysfunctions have been linked to ALS. One of the challenges in ALS research is determining, at the early stage of discovery, whether a new gene mutation is indeed disease-specific, and if it is linked to signaling pathways that trigger neuronal cell death. We have established a proof-of-concept proteogenomic workflow to assess new gene mutations, using CCNF (cyclin F) as an example, in cell culture models to screen whether potential gene candidates fit the criteria of activating apoptosis. This can provide an informative and time-efficient output that can be extended further for validation in a variety of in vitro and in vivo models and/or for mechanistic studies. As a proof-of-concept, we expressed cyclin F mutations (K97R, S195R, S509P, R574Q, S621G) in HEK293 cells for label-free quantitative proteomics that bioinformatically predicted activation of the neuronal cell death pathways, which was validated by immunoblot analysis. Proteomic analysis of induced pluripotent stem cells (iPSCs) derived from patient fibroblasts bearing the S621G mutation showed the same activation of these pathways providing compelling evidence for these candidate gene mutations to be strong candidates for further validation and mechanistic studies (such as E3 enzymatic activity assays, protein-protein and protein-substrate studies, and neuronal apoptosis and aberrant branching measurements in zebrafish). Our proteogenomics approach has great utility and provides a relatively high-throughput screening platform to explore candidate gene mutations for their propensity to cause neuronal cell death, which will guide a researcher for further experimental studies.
Collapse
Affiliation(s)
- Flora Cheng
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Alana De Luca
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Alison L Hogan
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Stephanie L Rayner
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Jennilee M Davidson
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Maxinne Watchon
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Claire H Stevens
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia.,School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Sonia Sanz Muñoz
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia.,School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia.,School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Justin J Yerbury
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia.,School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Emily K Don
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Jennifer A Fifita
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Maria D Villalva
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Hannah Suddull
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Tyler R Chapman
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Thomas J Hedl
- Neurodegeneration Pathobiology Laboratory, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Adam K Walker
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia.,Neurodegeneration Pathobiology Laboratory, Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Shu Yang
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Marco Morsch
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Bingyang Shi
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Ian P Blair
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Angela S Laird
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Roger S Chung
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Albert Lee
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| |
Collapse
|
68
|
Jin Y, Vadukul DM, Gialama D, Ge Y, Thrush R, White JT, Aprile FA. The Diagnostic Potential of Amyloidogenic Proteins. Int J Mol Sci 2021; 22:4128. [PMID: 33923609 PMCID: PMC8074075 DOI: 10.3390/ijms22084128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative disorders are a highly prevalent class of diseases, whose pathological mechanisms start before the appearance of any clear symptoms. This fact has prompted scientists to search for biomarkers that could aid early treatment. These currently incurable pathologies share the presence of aberrant aggregates called amyloids in the nervous system, which are composed of specific proteins. In this review, we discuss how these proteins, their conformations and modifications could be exploited as biomarkers for diagnostic purposes. We focus on proteins that are associated with the most prevalent neurodegenerative disorders, including Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, and frontotemporal dementia. We also describe current challenges in detection, the most recent techniques with diagnostic potentials and possible future developments in diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Francesco Antonio Aprile
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK; (Y.J.); (D.M.V.); (D.G.); (Y.G.); (R.T.); (J.T.W.)
| |
Collapse
|
69
|
Sannigrahi A, Chowdhury S, Das B, Banerjee A, Halder A, Kumar A, Saleem M, Naganathan AN, Karmakar S, Chattopadhyay K. The metal cofactor zinc and interacting membranes modulate SOD1 conformation-aggregation landscape in an in vitro ALS model. eLife 2021; 10:e61453. [PMID: 33825682 PMCID: PMC8087447 DOI: 10.7554/elife.61453] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 04/01/2021] [Indexed: 12/22/2022] Open
Abstract
Aggregation of Cu-Zn superoxide dismutase (SOD1) is implicated in the motor neuron disease, amyotrophic lateral sclerosis (ALS). Although more than 140 disease mutations of SOD1 are available, their stability or aggregation behaviors in membrane environment are not correlated with disease pathophysiology. Here, we use multiple mutational variants of SOD1 to show that the absence of Zn, and not Cu, significantly impacts membrane attachment of SOD1 through two loop regions facilitating aggregation driven by lipid-induced conformational changes. These loop regions influence both the primary (through Cu intake) and the gain of function (through aggregation) of SOD1 presumably through a shared conformational landscape. Combining experimental and theoretical frameworks using representative ALS disease mutants, we develop a 'co-factor derived membrane association model' wherein mutational stress closer to the Zn (but not to the Cu) pocket is responsible for membrane association-mediated toxic aggregation and survival time scale after ALS diagnosis.
Collapse
Affiliation(s)
- Achinta Sannigrahi
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical BiologyKolkataIndia
| | - Sourav Chowdhury
- Chemistry and Chemical Biology, Harvard UniversityCambridgeUnited States
| | - Bidisha Das
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical BiologyKolkataIndia
- Academy of Scientific and Innovative Research (AcSIR), CSIR- Human Resource development Centre CampusGhaziabadIndia
| | | | | | - Amaresh Kumar
- School of Biological Sciences, National Institute of Science Education and Research (NISER)BhubaneswarIndia
| | - Mohammed Saleem
- School of Biological Sciences, National Institute of Science Education and Research (NISER)BhubaneswarIndia
| | - Athi N Naganathan
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology MadrasChennaiIndia
| | | | - Krishnananda Chattopadhyay
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical BiologyKolkataIndia
- Academy of Scientific and Innovative Research (AcSIR), CSIR- Human Resource development Centre CampusGhaziabadIndia
| |
Collapse
|
70
|
Marafon G, Crisma M, Masato A, Plotegher N, Bubacco L, Moretto A. Photoresponsive Prion‐Mimic Foldamer to Induce Controlled Protein Aggregation. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202012995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Giulia Marafon
- Department of Chemical Sciences University of Padova 35131 Padova Italy
| | - Marco Crisma
- Institute of Biomolecular Chemistry Padova Unit CNR 35131 Padova Italy
| | - Anna Masato
- Department of Biology University of Padova 35131 Padova Italy
| | | | - Luigi Bubacco
- Department of Biology University of Padova 35131 Padova Italy
| | - Alessandro Moretto
- Department of Chemical Sciences University of Padova 35131 Padova Italy
- Institute of Biomolecular Chemistry Padova Unit CNR 35131 Padova Italy
| |
Collapse
|
71
|
Marafon G, Crisma M, Masato A, Plotegher N, Bubacco L, Moretto A. Photoresponsive Prion-Mimic Foldamer to Induce Controlled Protein Aggregation. Angew Chem Int Ed Engl 2021; 60:5173-5178. [PMID: 33180342 DOI: 10.1002/anie.202012995] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Indexed: 12/30/2022]
Abstract
Proteins reconfigure their 3D-structure, and consequently their function, under the control of specific molecular interactions that sense, process and transmit information from the surrounding environment. When this fundamental process is hampered, many pathologies occur as in the case of protein misfolding diseases. In this work, we follow the early steps of α-synuclein (aS) aggregation, a process associated with Parkinson's disease etiopathogenesis, that is promptly promoted by a light-mediated binding between the protein and a photoactive foldamer. The latter can switch between two conformations, one of which generates supramolecular fibrillar seeds that act as molecular templates able to induce a fast β-sheet transition for aS monomers that successively undergo fibrillar polymerization. The proposed method represents a powerful tool to study protein aggregation relevant to misfolding diseases in a controlled and inducible system.
Collapse
Affiliation(s)
- Giulia Marafon
- Department of Chemical Sciences, University of Padova, 35131, Padova, Italy
| | - Marco Crisma
- Institute of Biomolecular Chemistry, Padova Unit, CNR, 35131, Padova, Italy
| | - Anna Masato
- Department of Biology, University of Padova, 35131, Padova, Italy
| | | | - Luigi Bubacco
- Department of Biology, University of Padova, 35131, Padova, Italy
| | - Alessandro Moretto
- Department of Chemical Sciences, University of Padova, 35131, Padova, Italy.,Institute of Biomolecular Chemistry, Padova Unit, CNR, 35131, Padova, Italy
| |
Collapse
|
72
|
Shafiq K, Sanghai N, Guo Y, Kong J. Implication of post-translationally modified SOD1 in pathological aging. GeroScience 2021; 43:507-515. [PMID: 33608813 PMCID: PMC8110659 DOI: 10.1007/s11357-021-00332-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/31/2021] [Indexed: 12/18/2022] Open
Abstract
Why certain people relish healthy aging throughout their life span while others suffer pathological consequences? In this review, we focus on some of the dominant paradigms of pathological aging, such as amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), and Parkinson's disease (PD), and predict that the antioxidant superoxide dismutase 1 (SOD1), when post-translationally modified by aging-associated oxidative stress, acts as a mechanism to accelerated aging in these age-related neurodegenerative diseases. Oxidative modifications of natively reduced SOD1 induce pathological confirmations such as misfolding, leading to a subsequent formation of monomeric, oligomeric, and multimeric aggregates. Misfolded SOD1 propagates like prions from cell to cell. These modified conformations are detected in brain tissues in ALS, AD, and PD, and are considered a contributing factor to their initial pathogenesis. We have also elaborated on oxidative stress-induced non-native modifications of SOD1 and offered a logistic argument on their global implication in accelerated or pathological aging in the context of ALS, AD, and PD.
Collapse
Affiliation(s)
- Kashfia Shafiq
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, 27, King's College Cir, Toronto, ON, M5S, Canada
| | - Nitesh Sanghai
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada
| | - Ying Guo
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada.,Pathological Department, Hebei North University, Zhangjiakou, Hebei, China
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada.
| |
Collapse
|
73
|
Genç B, Gautam M, Gözütok Ö, Dervishi I, Sanchez S, Goshu GM, Koçak N, Xie E, Silverman RB, Özdinler PH. Improving mitochondria and ER stability helps eliminate upper motor neuron degeneration that occurs due to mSOD1 toxicity and TDP-43 pathology. Clin Transl Med 2021; 11:e336. [PMID: 33634973 PMCID: PMC7898037 DOI: 10.1002/ctm2.336] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Upper motor neurons (UMNs) are a key component of motor neuron circuitry. Their degeneration is a hallmark for diseases, such as hereditary spastic paraplegia (HSP), primary lateral sclerosis (PLS), and amyotrophic lateral sclerosis (ALS). Currently there are no preclinical assays investigating cellular responses of UMNs to compound treatment, even for diseases of the UMNs. The basis of UMN vulnerability is not fully understood, and no compound has yet been identified to improve the health of diseased UMNs: two major roadblocks for building effective treatment strategies. METHODS Novel UMN reporter models, in which UMNs that are diseased because of misfolded superoxide dismutase protein (mSOD1) toxicity and TDP-43 pathology are labeled with eGFP expression, allow direct assessment of UMN response to compound treatment. Electron microscopy reveals very precise aspects of endoplasmic reticulum (ER) and mitochondrial damage. Administration of NU-9, a compound initially identified based on its ability to reduce mSOD1 toxicity, has profound impact on improving the health and stability of UMNs, as identified by detailed cellular and ultrastructural analyses. RESULTS Problems with mitochondria and ER are conserved in diseased UMNs among different species. NU-9 has drug-like pharmacokinetic properties. It lacks toxicity and crosses the blood brain barrier. NU-9 improves the structural integrity of mitochondria and ER, reduces levels of mSOD1, stabilizes degenerating UMN apical dendrites, improves motor behavior measured by the hanging wire test, and eliminates ongoing degeneration of UMNs that become diseased both because of mSOD1 toxicity and TDP-43 pathology, two distinct and important overarching causes of motor neuron degeneration. CONCLUSIONS Mechanism-focused and cell-based drug discovery approaches not only addressed key cellular defects responsible for UMN loss, but also identified NU-9, the first compound to improve the health of diseased UMNs, neurons that degenerate in ALS, HSP, PLS, and ALS/FTLD patients.
Collapse
Affiliation(s)
- Barış Genç
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Mukesh Gautam
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Öge Gözütok
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Ina Dervishi
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Santana Sanchez
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Gashaw M. Goshu
- Department of ChemistryNorthwestern UniversityEvanstonIllinoisUSA
| | - Nuran Koçak
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Edward Xie
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Richard B. Silverman
- Department of ChemistryNorthwestern UniversityEvanstonIllinoisUSA
- Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental TherapeuticsNorthwestern UniversityEvanstonIllinoisUSA
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Chemistry of Life Processes InstituteNorthwestern UniversityEvanstonIL60208
| | - P. Hande Özdinler
- Department of Neurology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental TherapeuticsNorthwestern UniversityEvanstonIllinoisUSA
- Chemistry of Life Processes InstituteNorthwestern UniversityEvanstonIL60208
- Mesulam Center for Cognitive Neurology and Alzheimer's DiseaseNorthwestern University, Feinberg School of MedicineChicagoIL60611
- Les Turner ALS CenterNorthwestern University, Feinberg School of MedicineChicagoIL60611
| |
Collapse
|
74
|
Choi ES, Dokholyan NV. SOD1 oligomers in amyotrophic lateral sclerosis. Curr Opin Struct Biol 2021; 66:225-230. [PMID: 33465527 DOI: 10.1016/j.sbi.2020.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 11/18/2022]
Abstract
Identifying nonnative, trimeric forms of SOD1 trimers as the toxic species, rather than large aggregates revolutionizes our understanding of ALS pathophysiology. Large protein aggregates, what was previously thought as the central cause of neurodegeneration, play protective role and are not responsible for neuronal death. SOD1 trimers are implicated at the molecular, cellular, and organismal level. Understanding the formation of the nonnative trimer and its role in the cell, leading to cell death, holds the key to developing a new standard of therapeutics for ALS and for other neurodegenerative diseases. This review highlights recent advances of knowledge for the role of SOD1 oligomers in ALS.
Collapse
Affiliation(s)
- Esther S Choi
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA.
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
75
|
Vasta R, Torrieri MC, D'Ovidio F, Circiello A, De Mattei F, Manera U, Canosa A, Calvo A, Chiò A, Moglia C. Neck flexor weakness at diagnosis predicts respiratory impairment in amyotrophic lateral sclerosis. Eur J Neurol 2020; 28:1181-1187. [PMID: 33314477 DOI: 10.1111/ene.14676] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/03/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE The purpose was to assess the prognostic role of neck muscle weakness at diagnosis in amyotrophic lateral sclerosis (ALS) patients with respect to survival and respiratory impairment. METHODS A retrospective cohort study was conducted. All ALS patients seen in the Turin ALS Centre from 2007 to 2014 were included. Muscle strength at diagnosis was evaluated using the Medical Research Council (MRC) scale. Survival was considered as the time from diagnosis to death or tracheostomy; time to respiratory impairment was considered as the interval from diagnosis to the first event amongst an ALS Functional Rating Scale revised item 10 <4, forced vital capacity <70%, start of non-invasive ventilation or tracheostomy. Time from diagnosis to dysarthria, dysphagia and walking impairment were considered as secondary outcomes. Cox proportional hazard regression models adjusted for sex, age at diagnosis, diagnostic delay, onset site, genetics status and the MRC scores of other muscle groups were used to assess the prognostic role of neck muscles. RESULTS A total of 370 patients were included in the study. Fifty-nine (15.9%) patients showed neck flexor weakness at diagnosis; MRC values were mostly in agreement for neck extensors. Neck flexors were the only muscles able to predict survival (hazard ratio 0.49, 95% confidence interval 0.28-0.86; p = 0.01). Furthermore, neck flexor normal strength decreased the risk of respiratory impairment (hazard ratio 0.46, 95% confidence interval 0.22-0.96; p = 0.04) but did not influence any secondary outcomes. DISCUSSION Neck flexor weakness at diagnosis predicts survival and respiratory impairment in ALS. This result could be valuable for both planning of patients' interventions and clinical trials' design.
Collapse
Affiliation(s)
- Rosario Vasta
- ALS Centre, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Maria Claudia Torrieri
- ALS Centre, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Fabrizio D'Ovidio
- ALS Centre, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Alberta Circiello
- ALS Centre, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Filippo De Mattei
- ALS Centre, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Umberto Manera
- ALS Centre, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Antonio Canosa
- ALS Centre, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy.,Neurology 1, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Andrea Calvo
- ALS Centre, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy.,Neurology 1, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Adriano Chiò
- ALS Centre, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy.,Neurology 1, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Cristina Moglia
- ALS Centre, Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy.,Neurology 1, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| |
Collapse
|
76
|
Evidence that corticofugal propagation of ALS pathology is not mediated by prion-like mechanism. Prog Neurobiol 2020; 200:101972. [PMID: 33309802 DOI: 10.1016/j.pneurobio.2020.101972] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/27/2020] [Accepted: 12/06/2020] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) arises from the combined degeneration of motor neurons (MN) and corticospinal neurons (CSN). Recent clinical and pathological studies suggest that ALS might start in the motor cortex and spread along the corticofugal axonal projections (including the CSN), either via altered cortical excitability and activity or via prion-like propagation of misfolded proteins. Using mouse genetics, we recently provided the first experimental arguments in favour of the corticofugal hypothesis, but the mechanism of propagation remained an open question. To gain insight into this matter, we tested here the possibility that the toxicity of the corticofugal projection neurons (CFuPN) to their targets could be mediated by their cell autonomous-expression of an ALS causing transgene and possible diffusion of toxic misfolded proteins to their spinal targets. We generated a Crym-CreERT2 mouse line to ablate the SOD1G37R transgene selectively in CFuPN. This was sufficient to fully rescue the CSN and to limit spasticity, but had no effect on the burden of misfolded SOD1 protein in the spinal cord, MN survival, disease onset and progression. The data thus indicate that in ALS corticofugal propagation is likely not mediated by prion-like mechanisms, but could possibly rather rely on cortical hyperexcitability.
Collapse
|
77
|
HuD regulates SOD1 expression during oxidative stress in differentiated neuroblastoma cells and sporadic ALS motor cortex. Neurobiol Dis 2020; 148:105211. [PMID: 33271327 DOI: 10.1016/j.nbd.2020.105211] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/09/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
The neuronal RNA-binding protein (RBP) HuD plays an important role in brain development, synaptic plasticity and neurodegenerative diseases such as Parkinson's (PD) and Alzheimer's (AD). Bioinformatics analysis of the human SOD1 mRNA 3' untranslated region (3'UTR) demonstrated the presence of HuD binding adenine-uridine (AU)-rich instability-conferring elements (AREs). Using differentiated SH-SY5Y cells along with brain tissues from sporadic amyotrophic lateral sclerosis (sALS) patients, we assessed HuD-dependent regulation of SOD1 mRNA. In vitro binding and mRNA decay assays demonstrate that HuD specifically binds to SOD1 ARE motifs promoting mRNA stabilization. In SH-SY5Y cells, overexpression of full-length HuD increased SOD1 mRNA and protein levels while a dominant negative form of the RBP downregulated its expression. HuD regulation of SOD1 mRNA was also found to be oxidative stress (OS)-dependent, as shown by the increased HuD binding and upregulation of this mRNA after H2O2 exposure. This treatment also induced a shift in alternative polyadenylation (APA) site usage in SOD1 3'UTR, increasing the levels of a long variant bearing HuD binding sites. The requirement of HuD for SOD1 upregulation during oxidative damage was validated using a specific siRNA that downregulated HuD protein levels to 36% and prevented upregulation of SOD1 and 91 additional genes. In the motor cortex from sALS patients, we found increases in SOD1 and HuD mRNAs and proteins, accompanied by greater HuD binding to this mRNA as confirmed by RNA-immunoprecipitation (RIP) assays. Altogether, our results suggest a role of HuD in the post-transcriptional regulation of SOD1 expression during ALS pathogenesis.
Collapse
|
78
|
Abstract
Though primarily a sporadic condition, Parkinson’s disease is increasingly recognized to be a multifactorial disease with a strong genetic component. At a cellular level, disruptions of protein trafficking and recycling, particularly by misfolding, accumulation, and aggregation of α-synuclein, mitochondrial dysfunction, oxidative stress, and other etiopathogenic mechanisms, have been found to result in the death of vulnerable neuronal populations and appear to drive the neurodegeneration underlying Parkinson’s disease. The improved understanding of these mechanisms has led to the development of novel pathogenesis-targeted and potentially disease-modifying therapeutic approaches in Parkinson’s disease. Until these treatments are fully developed and approved, clinicians must rely on therapies designed to improve quality of life of patients by treating various motor and non-motor symptoms of the disease.
Collapse
Affiliation(s)
- Arjun Tarakad
- Parkinson’s Disease Center and Movement Disorder Clinic, Department of Neurology, Baylor College of Medicine, 7200 Cambridge, Suite 9A, Houston, TX 77030-4202, USA
| | - Joseph Jankovic
- Parkinson’s Disease Center and Movement Disorder Clinic, Department of Neurology, Baylor College of Medicine, 7200 Cambridge, Suite 9A, Houston, TX 77030-4202, USA
| |
Collapse
|
79
|
McAlary L, Chew YL, Lum JS, Geraghty NJ, Yerbury JJ, Cashman NR. Amyotrophic Lateral Sclerosis: Proteins, Proteostasis, Prions, and Promises. Front Cell Neurosci 2020; 14:581907. [PMID: 33328890 PMCID: PMC7671971 DOI: 10.3389/fncel.2020.581907] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the progressive degeneration of the motor neurons that innervate muscle, resulting in gradual paralysis and culminating in the inability to breathe or swallow. This neuronal degeneration occurs in a spatiotemporal manner from a point of onset in the central nervous system (CNS), suggesting that there is a molecule that spreads from cell-to-cell. There is strong evidence that the onset and progression of ALS pathology is a consequence of protein misfolding and aggregation. In line with this, a hallmark pathology of ALS is protein deposition and inclusion formation within motor neurons and surrounding glia of the proteins TAR DNA-binding protein 43, superoxide dismutase-1, or fused in sarcoma. Collectively, the observed protein aggregation, in conjunction with the spatiotemporal spread of symptoms, strongly suggests a prion-like propagation of protein aggregation occurs in ALS. In this review, we discuss the role of protein aggregation in ALS concerning protein homeostasis (proteostasis) mechanisms and prion-like propagation. Furthermore, we examine the experimental models used to investigate these processes, including in vitro assays, cultured cells, invertebrate models, and murine models. Finally, we evaluate the therapeutics that may best prevent the onset or spread of pathology in ALS and discuss what lies on the horizon for treating this currently incurable disease.
Collapse
Affiliation(s)
- Luke McAlary
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Yee Lian Chew
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Jeremy Stephen Lum
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Nicholas John Geraghty
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Justin John Yerbury
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Neil R. Cashman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
80
|
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: 3.2] [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
|
81
|
Jo M, Lee S, Jeon YM, Kim S, Kwon Y, Kim HJ. The role of TDP-43 propagation in neurodegenerative diseases: integrating insights from clinical and experimental studies. Exp Mol Med 2020; 52:1652-1662. [PMID: 33051572 PMCID: PMC8080625 DOI: 10.1038/s12276-020-00513-7] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/21/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
TAR DNA-binding protein 43 (TDP-43) is a highly conserved nuclear RNA/DNA-binding protein involved in the regulation of RNA processing. The accumulation of TDP-43 aggregates in the central nervous system is a common feature of many neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Alzheimer's disease (AD), and limbic predominant age-related TDP-43 encephalopathy (LATE). Accumulating evidence suggests that prion-like spreading of aberrant protein aggregates composed of tau, amyloid-β, and α-synuclein is involved in the progression of neurodegenerative diseases such as AD and PD. Similar to those of prion-like proteins, pathological aggregates of TDP-43 can be transferred from cell-to-cell in a seed-dependent and self-templating manner. Here, we review clinical and experimental studies supporting the prion-like spreading of misfolded TDP-43 and discuss the molecular mechanisms underlying the propagation of these pathological aggregated proteins. The idea that misfolded TDP-43 spreads in a prion-like manner between cells may guide novel therapeutic strategies for TDP-43-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Myungjin Jo
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
| | - Shinrye Lee
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
| | - Yu-Mi Jeon
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea
| | - Seyeon Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea.,Department of Brain & Cognitive Sciences, DGIST, Daegu, 42988, South Korea
| | - Younghwi Kwon
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea.,Department of Brain & Cognitive Sciences, DGIST, Daegu, 42988, South Korea
| | - Hyung-Jun Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, South Korea.
| |
Collapse
|
82
|
Plotkin SS, Cashman NR. Passive immunotherapies targeting Aβ and tau in Alzheimer's disease. Neurobiol Dis 2020; 144:105010. [PMID: 32682954 PMCID: PMC7365083 DOI: 10.1016/j.nbd.2020.105010] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 07/01/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022] Open
Abstract
Amyloid-β (Aβ) and tau proteins currently represent the two most promising targets to treat Alzheimer's disease. The most extensively developed method to treat the pathologic forms of these proteins is through the administration of exogenous antibodies, or passive immunotherapy. In this review, we discuss the molecular-level strategies that researchers are using to design an effective therapeutic antibody, given the challenges in treating this disease. These challenges include selectively targeting a protein that has misfolded or is pathological rather than the more abundant, healthy protein, designing strategic constructs for immunizing an animal to raise an antibody that has the appropriate conformational selectivity to achieve this end, and clearing the pathological protein species before prion-like cell-to-cell spread of misfolded protein has irreparably damaged neurons, without invoking damaging inflammatory responses in the brain that naturally arise when the innate immune system is clearing foreign agents. The various solutions to these problems in current clinical trials will be discussed.
Collapse
Affiliation(s)
- Steven S Plotkin
- University of British Columbia, Department of Physics and Astronomy and Genome Sciences and Technology Program, Vancouver, BC V6T 1Z1, Canada.
| | - Neil R Cashman
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 2B5, Canada.
| |
Collapse
|
83
|
Lauwers E, Lalli G, Brandner S, Collinge J, Compernolle V, Duyckaerts C, Edgren G, Haïk S, Hardy J, Helmy A, Ivinson AJ, Jaunmuktane Z, Jucker M, Knight R, Lemmens R, Lin IC, Love S, Mead S, Perry VH, Pickett J, Poppy G, Radford SE, Rousseau F, Routledge C, Schiavo G, Schymkowitz J, Selkoe DJ, Smith C, Thal DR, Theys T, Tiberghien P, van den Burg P, Vandekerckhove P, Walton C, Zaaijer HL, Zetterberg H, De Strooper B. Potential human transmission of amyloid β pathology: surveillance and risks. Lancet Neurol 2020; 19:872-878. [PMID: 32949547 DOI: 10.1016/s1474-4422(20)30238-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 01/05/2023]
Abstract
Studies in experimental animals show transmissibility of amyloidogenic proteins associated with prion diseases, Alzheimer's disease, Parkinson's disease, and other neurodegenerative diseases. Although these data raise potential concerns for public health, convincing evidence for human iatrogenic transmission only exists for prions and amyloid β after systemic injections of contaminated growth hormone extracts or dura mater grafts derived from cadavers. Even though these procedures are now obsolete, some reports raise the possibility of iatrogenic transmission of amyloid β through putatively contaminated neurosurgical equipment. Iatrogenic transmission of amyloid β might lead to amyloid deposition in the brain parenchyma and blood vessel walls, potentially resulting in cerebral amyloid angiopathy after several decades. Cerebral amyloid angiopathy can cause life-threatening brain haemorrhages; yet, there is no proof that the transmission of amyloid β can also lead to Alzheimer's dementia. Large, long-term epidemiological studies and sensitive, cost-efficient tools to detect amyloid are needed to better understand any potential routes of amyloid β transmission and to clarify whether other similar proteopathic seeds, such as tau or α-synuclein, can also be transferred iatrogenically.
Collapse
Affiliation(s)
- Elsa Lauwers
- VIB-KU Leuven Center for Brain and Disease Research, KU Leuven, Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Giovanna Lalli
- UK Dementia Research Institute, University College London, London, UK
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK; Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London National Health Service Foundation Trust, London, UK
| | - John Collinge
- Medical Research Council Prion Unit at UCL, Institute of Prion Diseases, University College London, London, UK
| | - Veerle Compernolle
- Blood Services, Belgian Red Cross-Flanders, Mechelen, Belgium; Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Charles Duyckaerts
- Institut du Cerveau et de la Moelle épinière, Sorbonne University, INSERM, CNRS UMR, Paris, France; Laboratoire de Neuropathologie Raymond Escourolle, Hôpital de la Pitié-Salpêtrière, Assistance Publique- Hôpitaux de Paris, Paris, France
| | - Gustaf Edgren
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Cardiology, Södersjukhuset, Stockholm, Sweden
| | - Stéphane Haïk
- Institut du Cerveau et de la Moelle épinière, Sorbonne University, INSERM, CNRS UMR, Paris, France; Laboratoire de Neuropathologie Raymond Escourolle, Hôpital de la Pitié-Salpêtrière, Assistance Publique- Hôpitaux de Paris, Paris, France; Cellule Nationale de Référence des maladies de Creutzfeldt-Jakob, Hôpital de la Pitié-Salpêtrière, Assistance Publique- Hôpitaux de Paris, Paris, France
| | - John Hardy
- UK Dementia Research Institute, University College London, London, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK; Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, University College London, London, UK; National Institute for Health Research University College London Hospitals Biomedical Research Centre, London, UK; Institute for Advanced Study, The Hong Kong University of Science and Technology, Hong Kong Special Administrative Region, China
| | - Adel Helmy
- Department of Clinical Neuroscience, Division of Neurosurgery, University of Cambridge, Cambridge, UK
| | - Adrian J Ivinson
- UK Dementia Research Institute, University College London, London, UK
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK; Queen Square Brain Bank for Neurological Disorders, Queen Square Institute of Neurology, University College London, London, UK; Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London National Health Service Foundation Trust, London, UK
| | - Mathias Jucker
- Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Richard Knight
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK; National Creutzfeldt-Jakob Disease Research and Surveillance Unit, Western General Hospital, Edinburgh, UK
| | - Robin Lemmens
- VIB-KU Leuven Center for Brain and Disease Research, KU Leuven, Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - I-Chun Lin
- UK Dementia Research Institute, University College London, London, UK
| | - Seth Love
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Simon Mead
- Medical Research Council Prion Unit at UCL, Institute of Prion Diseases, University College London, London, UK
| | - V Hugh Perry
- UK Dementia Research Institute, University College London, London, UK
| | - James Pickett
- Alzheimer's Society, London, London, UK; Epilepsy Research UK, London, UK
| | - Guy Poppy
- Biological Sciences, University of Southampton, Southampton, UK
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Frederic Rousseau
- VIB-KU Leuven Center for Brain and Disease Research, KU Leuven, Leuven, Belgium; Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | | | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, UK; Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Joost Schymkowitz
- VIB-KU Leuven Center for Brain and Disease Research, KU Leuven, Leuven, Belgium; Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Harvard University, Boston, MA, USA
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Dietmar R Thal
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium; Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Tom Theys
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
| | - Pierre Tiberghien
- Etablissement Français du Sang, La Plaine St Denis, France; Unité Mixte de Recherche, INSERM, Université de Franche-Comté, Besançon, France
| | - Peter van den Burg
- European Blood Alliance, Brussels, Belgium; Department of Transfusion Medicine, Sanquin, Amsterdam, Netherlands
| | - Philippe Vandekerckhove
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium; Blood Services, Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Clare Walton
- Alzheimer's Society, London, London, UK; Multiple Sclerosis International Federation, London, UK
| | - Hans L Zaaijer
- Department of Blood-borne Infections, Sanquin, Amsterdam, Netherlands
| | - Henrik Zetterberg
- UK Dementia Research Institute, University College London, London, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK; Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bart De Strooper
- VIB-KU Leuven Center for Brain and Disease Research, KU Leuven, Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium; UK Dementia Research Institute, University College London, London, UK.
| |
Collapse
|
84
|
From Seeds to Fibrils and Back: Fragmentation as an Overlooked Step in the Propagation of Prions and Prion-Like Proteins. Biomolecules 2020; 10:biom10091305. [PMID: 32927676 PMCID: PMC7563560 DOI: 10.3390/biom10091305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Many devastating neurodegenerative diseases are driven by the misfolding of normal proteins into a pathogenic abnormal conformation. Examples of such protein misfolding diseases include Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and prion diseases. The misfolded proteins involved in these diseases form self-templating oligomeric assemblies that recruit further correctly folded protein and induce their conversion. Over time, this leads to the formation of high molecular and mostly fibrillar aggregates that are increasingly inefficient at converting normal protein. Evidence from a multitude of in vitro models suggests that fibrils are fragmented to form new seeds, which can convert further normal protein and also spread to neighboring cells as observed in vivo. While fragmentation and seed generation were suggested as crucial steps in aggregate formation decades ago, the biological pathways involved remain largely unknown. Here, we show that mechanisms of aggregate clearance—namely the mammalian Hsp70–Hsp40–Hsp110 tri-chaperone system, macro-autophagy, and the proteasome system—may not only be protective, but also play a role in fragmentation. We further review the challenges that exist in determining the precise contribution of these mechanisms to protein misfolding diseases and suggest future directions to resolve these issues.
Collapse
|
85
|
McAlary L, Yerbury JJ, Cashman NR. The prion-like nature of amyotrophic lateral sclerosis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:261-296. [PMID: 32958236 DOI: 10.1016/bs.pmbts.2020.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The misfolding, aggregation, and deposition of specific proteins is the key hallmark of most progressive neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis (ALS). ALS is characterized by the rapid and progressive degenerations of motor neurons in the spinal cord and motor cortex, resulting in paralysis of those who suffer from it. Pathologically, there are three major aggregating proteins associated with ALS, including TAR DNA-binding protein of 43kDa (TDP-43), superoxide dismutase-1 (SOD1), and fused in sarcoma (FUS). While there are ALS-associated mutations found in each of these proteins, the most prevalent aggregation pathology is that of wild-type TDP-43 (97% of cases), with the remaining split between mutant forms of SOD1 (~2%) and FUS (~1%). Considering the progressive nature of ALS and its association with the aggregation of specific proteins, a growing notion is that the spread of pathology and symptoms can be explained by a prion-like mechanism. Prion diseases are a group of highly infectious neurodegenerative disorders caused by the misfolding, aggregation, and spread of a transmissible conformer of prion protein (PrP). Pathogenic PrP is capable of converting healthy PrP into a toxic form through template-directed misfolding. Application of this finding to other neurodegenerative disorders, and in particular ALS, has revolutionized our understanding of cause and progression of these disorders. In this chapter, we first provide a background on ALS pathology and genetic origin. We then detail and discuss the evidence supporting a prion-like propagation of protein misfolding and aggregation in ALS with a particular focus on SOD1 and TDP-43 as these are the most well-established models in the field.
Collapse
Affiliation(s)
- L McAlary
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia; Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - J J Yerbury
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia; Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - N R Cashman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
86
|
van Es MA, Goedee HS, Westeneng HJ, Nijboer TCW, van den Berg LH. Is it accurate to classify ALS as a neuromuscular disorder? Expert Rev Neurother 2020; 20:895-906. [PMID: 32749157 DOI: 10.1080/14737175.2020.1806061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a fatal disorder characterized by the progressive loss of upper and lower motor neurons. ALS has traditionally been classified within the domain of neuromuscular diseases, which are a unique spectrum of disorders that predominantly affect the peripheral nervous system. However, over the past decades compounding evidence has emerged that there is extensive involvement of the central nervous system. Therefore, one can question whether it remains accurate to classify ALS as a neuromuscular disorder. AREAS COVERED In this review, the authors sought to discuss current approaches toward disease classification and how we should classify ALS based on novel insights from clinical, imaging, pathophysiological, neuropathological and genetic studies. EXPERT OPINION ALS exhibits the cardinal features of a neurodegenerative disease. Therefore, classifying ALS as a neuromuscular disease in the strict sense has become untenable. Diagnosing ALS however does require significant neuromuscular expertise and therefore neuromuscular specialists remain best equipped to evaluate this category of patients. Designating motor neuron diseases as a separate category in the ICD-11 is justified and adequately deals with this issue. However, to drive effective therapy development the fields of motor neuron disease and neurodegenerative disorders must come together.
Collapse
Affiliation(s)
- Michael A van Es
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht , Utrecht, The Netherlands
| | - H Stephan Goedee
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Henk-Jan Westeneng
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Tanja C W Nijboer
- Center of Excellence for Rehabilitation Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, and De Hoogstraat Rehabilitation , Utrecht, Netherlands.,Department of Experimental Psychology, Helmholtz Institute, Utrecht University , Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht , Utrecht, The Netherlands
| |
Collapse
|
87
|
Kukharsky MS, Skvortsova VI, Bachurin SO, Buchman VL. In a search for efficient treatment for amyotrophic lateral sclerosis: Old drugs for new approaches. Med Res Rev 2020; 41:2804-2822. [DOI: 10.1002/med.21725] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/23/2020] [Accepted: 08/08/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Michail S. Kukharsky
- Faculty of Medical Biology Pirogov Russian National Research Medical University Moscow Russian Federation
- Institute of Physiologically Active Compounds Russian Academy of Sciences Moscow Region Russian Federation
| | - Veronika I. Skvortsova
- Faculty of Medical Biology Pirogov Russian National Research Medical University Moscow Russian Federation
| | - Sergey O. Bachurin
- Institute of Physiologically Active Compounds Russian Academy of Sciences Moscow Region Russian Federation
| | - Vladimir L. Buchman
- Institute of Physiologically Active Compounds Russian Academy of Sciences Moscow Region Russian Federation
- School of Biosciences Cardiff University Cardiff United Kingdom
| |
Collapse
|
88
|
San Gil R, Cox D, McAlary L, Berg T, Walker AK, Yerbury JJ, Ooi L, Ecroyd H. Neurodegenerative disease-associated protein aggregates are poor inducers of the heat shock response in neuronal cells. J Cell Sci 2020; 133:jcs.243709. [PMID: 32661089 DOI: 10.1242/jcs.243709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022] Open
Abstract
Protein aggregates that result in inclusion formation are a pathological hallmark common to many neurodegenerative diseases, including amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. Under conditions of cellular stress, activation of the heat shock response (HSR) results in an increase in the levels of molecular chaperones and is a first line of cellular defence against inclusion formation. It remains to be established whether neurodegenerative disease-associated proteins and inclusions are themselves capable of inducing an HSR in neuronal cells. To address this, we generated a neuroblastoma cell line that expresses a fluorescent reporter protein under conditions of heat shock transcription factor 1 (HSF1)-mediated HSR induction. We show that the HSR is not induced by exogenous treatment with aggregated forms of recombinant α-synuclein or the G93A mutant of superoxide dismutase-1 (SOD1G93A) nor intracellular expression of SOD1G93A or a pathogenic form of polyglutamine-expanded huntingtin (Htt72Q). These results suggest that pathogenic proteins evade detection or impair induction of the HSR in neuronal cells. A failure of protein aggregation to induce an HSR might contribute to the development of inclusion pathology in neurodegenerative diseases.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Rebecca San Gil
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia.,Neurodegeneration Pathobiology Laboratory, Queensland Brain Institute, University of Queensland, St Lucia, QLD 4072, Australia
| | - Dezerae Cox
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia
| | - Luke McAlary
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Tracey Berg
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Adam K Walker
- Neurodegeneration Pathobiology Laboratory, Queensland Brain Institute, University of Queensland, St Lucia, QLD 4072, Australia
| | - Justin J Yerbury
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Heath Ecroyd
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia .,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
89
|
Aydemir D, Ulusu NN. Importance of the serum biochemical parameters as potential biomarkers for rapid diagnosis and evaluating preclinical stage of ALS. Med Hypotheses 2020; 141:109736. [DOI: 10.1016/j.mehy.2020.109736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 02/07/2023]
|
90
|
Izrael M, Slutsky SG, Revel M. Rising Stars: Astrocytes as a Therapeutic Target for ALS Disease. Front Neurosci 2020; 14:824. [PMID: 32848579 PMCID: PMC7399224 DOI: 10.3389/fnins.2020.00824] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a multifactorial disease, characterized by a progressive loss of motor neurons that eventually leads to paralysis and death. The current ALS-approved drugs modestly change the clinical course of the disease. The mechanism by which motor neurons progressively degenerate remains unclear but entails a non-cell autonomous process. Astrocytes impaired biological functionality were implicated in multiple neurodegenerative diseases, including ALS, frontotemporal dementia (FTD), Parkinson’s disease (PD), and Alzheimer disease (AD). In ALS disease patients, A1 reactive astrocytes were found to play a key role in the pathology of ALS disease and death of motor neurons, via loss or gain of function or acquired toxicity. The contribution of astrocytes to the maintenance of motor neurons by diverse mechanisms makes them a promising therapeutic candidate for the treatment of ALS. Therapeutic approaches targeting at modulating the function of endogenous astrocytes or replacing lost functionality by transplantation of healthy astrocytes, may contribute to the development of therapies which might slow down or even halt the progression ALS diseases. The proposed mechanisms by which astrocytes can potentially ameliorate ALS progression and the status of ALS clinical studies involving astrocytes are discussed.
Collapse
Affiliation(s)
- Michal Izrael
- Neurodegenerative Diseases Department at Kadimastem Ltd., Nes-Ziona, Israel
| | - Shalom Guy Slutsky
- Neurodegenerative Diseases Department at Kadimastem Ltd., Nes-Ziona, Israel
| | - Michel Revel
- Neurodegenerative Diseases Department at Kadimastem Ltd., Nes-Ziona, Israel.,Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
91
|
Chatterjee S, Salimi A, Lee JY. Insights into amyotrophic lateral sclerosis linked Pro525Arg mutation in the fused in sarcoma protein through in silico analysis and molecular dynamics simulation. J Biomol Struct Dyn 2020; 39:5963-5976. [DOI: 10.1080/07391102.2020.1794967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
| | - Abbas Salimi
- Department of Chemistry, Sungkyunkwan University, Suwon, Korea
| | - Jin Yong Lee
- Department of Chemistry, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
92
|
Kim Y, Connor JR. The roles of iron and HFE genotype in neurological diseases. Mol Aspects Med 2020; 75:100867. [PMID: 32654761 DOI: 10.1016/j.mam.2020.100867] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/21/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022]
Abstract
Iron accumulation is a recurring pathological phenomenon in many neurological diseases including Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, and others. Iron is essential for normal development and functions of the brain; however, excess redox-active iron can also lead to oxidative damage and cell death. Especially for terminally differentiated cells like neurons, regulation of reactive oxygen species is critical for cell viability. As a result, cellular iron level is tightly regulated. Although iron accumulation related to neurological diseases has been well documented, the pathoetiological contributions of the homeostatic iron regulator (HFE), which controls cellular iron uptake, is less understood. Furthermore, a common HFE variant, H63D HFE, has been identified as a modifier of multiple neurological diseases. This review will discuss the roles of iron and HFE in the brain as well as their impact on various disease processes.
Collapse
Affiliation(s)
- Yunsung Kim
- Penn State College of Medicine, Department of Neurosurgery, Hershey, PA, USA
| | - James R Connor
- Penn State College of Medicine, Department of Neurosurgery, Hershey, PA, USA.
| |
Collapse
|
93
|
Affiliation(s)
- Orla Hardiman
- From Trinity College and Beaumont Hospital, Dublin (O.H.); and University Medical Center, Utrecht, the Netherlands (L.H.B.)
| | - Leonard H van den Berg
- From Trinity College and Beaumont Hospital, Dublin (O.H.); and University Medical Center, Utrecht, the Netherlands (L.H.B.)
| |
Collapse
|
94
|
McCombe PA, Garton FC, Katz M, Wray NR, Henderson RD. What do we know about the variability in survival of patients with amyotrophic lateral sclerosis? Expert Rev Neurother 2020; 20:921-941. [PMID: 32569484 DOI: 10.1080/14737175.2020.1785873] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION ALS is a fatal neurodegenerative disease. However, patients show variability in the length of survival after symptom onset. Understanding the mechanisms of long survival could lead to possible avenues for therapy. AREAS COVERED This review surveys the reported length of survival in ALS, the clinical features that predict survival in individual patients, and possible factors, particularly genetic factors, that could cause short or long survival. The authors also speculate on possible mechanisms. EXPERT OPINION a small number of known factors can explain some variability in ALS survival. However, other disease-modifying factors likely exist. Factors that alter motor neurone vulnerability and immune, metabolic, and muscle function could affect survival by modulating the disease process. Knowing these factors could lead to interventions to change the course of the disease. The authors suggest a broad approach is needed to quantify the proportion of variation survival attributable to genetic and non-genetic factors and to identify and estimate the effect size of specific factors. Studies of this nature could not only identify novel avenues for therapeutic research but also play an important role in clinical trial design and personalized medicine.
Collapse
Affiliation(s)
- Pamela A McCombe
- Centre for Clinical Research, The University of Queensland , Brisbane, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital , Brisbane, Australia
| | - Fleur C Garton
- Institute for Molecular Biosciences, The University of Queensland , Brisbane, Australia
| | - Matthew Katz
- Department of Neurology, Royal Brisbane and Women's Hospital , Brisbane, Australia
| | - Naomi R Wray
- Institute for Molecular Biosciences, The University of Queensland , Brisbane, Australia.,Queensland Brain Institute, The University of Queensland , Brisbane, Australia
| | - Robert D Henderson
- Centre for Clinical Research, The University of Queensland , Brisbane, Australia
| |
Collapse
|
95
|
Malik R, Wiedau M. Therapeutic Approaches Targeting Protein Aggregation in Amyotrophic Lateral Sclerosis. Front Mol Neurosci 2020; 13:98. [PMID: 32581709 PMCID: PMC7296057 DOI: 10.3389/fnmol.2020.00098] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/08/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a debilitating neurodegenerative disease that targets motor neurons (MNs) in the brain and spinal cord. It leads to gradual loss of motor signals to muscles leading to atrophy and weakness. Most patients do not survive for more than 3–5 years after disease onset. Current ALS treatments provide only a small delay of disease progression. Therefore, it is of utmost importance to explore new therapeutic approaches. One of the major hindrances in achieving this goal is poor understanding of causes of the disease. ALS has complex pathophysiological mechanisms in its genetic and sporadic forms. Protein aggregates are a common hallmark of ALS regardless of cause making protein pathways attractive therapeutic targets in ALS. Here, we provide an overview of compounds in different stages of pharmacological development and their protein pathway targets.
Collapse
Affiliation(s)
- Ravinder Malik
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Martina Wiedau
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
96
|
Barp A, Gerardi F, Lizio A, Sansone VA, Lunetta C. Emerging Drugs for the Treatment of Amyotrophic Lateral Sclerosis: A Focus on Recent Phase 2 Trials. Expert Opin Emerg Drugs 2020; 25:145-164. [PMID: 32456491 DOI: 10.1080/14728214.2020.1769067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disease involving both upper and lower motor neurons and resulting in increasing disability and death 3-5 years after onset of symptoms. Over 40 large clinical trials for ALS have been negative, except for Riluzole that offers a modest survival benefit, and Edaravone that modestly reduces disease progression in patients with specific characteristics. Thus, the discovery of efficient disease modifying therapy is an urgent need. AREAS COVERED Although the cause of ALS remains unclear, many studies have demonstrated that neuroinflammation, proteinopathies, glutamate-induced excitotoxicity, microglial activation, oxidative stress, and mitochondrial dysfunction may play a key role in the pathogenesis. This review highlights recent discoveries relating to these diverse mechanisms and their implications for the development of therapy. Ongoing phase 2 clinical trials aimed to interfere with these pathophysiological mechanisms are discussed. EXPERT OPINION This review describes the challenges that the discovery of an efficient drug therapy faces and how these issues may be addressed. With the continuous advances coming from basic research, we provided possible suggestions that may be considered to improve performance of clinical trials and turn ALS research into a 'fertile ground' for drug development for this devastating disease.
Collapse
Affiliation(s)
- Andrea Barp
- NEuroMuscular Omnicentre, Fondazione Serena Onlus , Milan, Italy.,Dept. Biomedical Sciences of Health, University of Milan , Milan, Italy
| | | | - Andrea Lizio
- NEuroMuscular Omnicentre, Fondazione Serena Onlus , Milan, Italy
| | - Valeria Ada Sansone
- NEuroMuscular Omnicentre, Fondazione Serena Onlus , Milan, Italy.,Dept. Biomedical Sciences of Health, University of Milan , Milan, Italy
| | | |
Collapse
|
97
|
Chernoff YO, Grizel AV, Rubel AA, Zelinsky AA, Chandramowlishwaran P, Chernova TA. Application of yeast to studying amyloid and prion diseases. ADVANCES IN GENETICS 2020; 105:293-380. [PMID: 32560789 PMCID: PMC7527210 DOI: 10.1016/bs.adgen.2020.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyloids are fibrous cross-β protein aggregates that are capable of proliferation via nucleated polymerization. Amyloid conformation likely represents an ancient protein fold and is linked to various biological or pathological manifestations. Self-perpetuating amyloid-based protein conformers provide a molecular basis for transmissible (infectious or heritable) protein isoforms, termed prions. Amyloids and prions, as well as other types of misfolded aggregated proteins are associated with a variety of devastating mammalian and human diseases, such as Alzheimer's, Parkinson's and Huntington's diseases, transmissible spongiform encephalopathies (TSEs), amyotrophic lateral sclerosis (ALS) and transthyretinopathies. In yeast and fungi, amyloid-based prions control phenotypically detectable heritable traits. Simplicity of cultivation requirements and availability of powerful genetic approaches makes yeast Saccharomyces cerevisiae an excellent model system for studying molecular and cellular mechanisms governing amyloid formation and propagation. Genetic techniques allowing for the expression of mammalian or human amyloidogenic and prionogenic proteins in yeast enable researchers to capitalize on yeast advantages for characterization of the properties of disease-related proteins. Chimeric constructs employing mammalian and human aggregation-prone proteins or domains, fused to fluorophores or to endogenous yeast proteins allow for cytological or phenotypic detection of disease-related protein aggregation in yeast cells. Yeast systems are amenable to high-throughput screening for antagonists of amyloid formation, propagation and/or toxicity. This review summarizes up to date achievements of yeast assays in application to studying mammalian and human disease-related aggregating proteins, and discusses both limitations and further perspectives of yeast-based strategies.
Collapse
Affiliation(s)
- Yury O Chernoff
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States; Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg, Russia.
| | - Anastasia V Grizel
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg, Russia
| | - Aleksandr A Rubel
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg, Russia; Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg, Russia; Sirius University of Science and Technology, Sochi, Russia
| | - Andrew A Zelinsky
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg, Russia
| | | | - Tatiana A Chernova
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
98
|
Yerbury JJ, Farrawell NE, McAlary L. Proteome Homeostasis Dysfunction: A Unifying Principle in ALS Pathogenesis. Trends Neurosci 2020; 43:274-284. [PMID: 32353332 DOI: 10.1016/j.tins.2020.03.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/18/2020] [Accepted: 03/01/2020] [Indexed: 02/08/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common motor neuron disease but currently has no effective treatment. Growing evidence suggests that proteome homeostasis underlies ALS pathogenesis. Protein production, trafficking, and degradation all shape the proteome. We present a hypothesis that proposes all genetic lesions associated with ALS (including in mRNA-binding proteins) cause widespread imbalance to an already metastable proteome. The impact of such dysfunction is felt across the entire proteome and is not restricted to a small subset of proteins. Proteome imbalance may cause functional defects, such as excitability alterations, and eventually cell death. While this idea is a unifying principle for all of ALS, we propose that stratification will appear that might dictate the efficacy of therapeutics based on the proteostasis network.
Collapse
Affiliation(s)
- Justin J Yerbury
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons and School of Chemistry and Molecular Biosciences, University of Wollongong, Wollongong, NSW 2522, Australia.
| | - Natalie E Farrawell
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons and School of Chemistry and Molecular Biosciences, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Luke McAlary
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons and School of Chemistry and Molecular Biosciences, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
99
|
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: 59] [Impact Index Per Article: 11.8] [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
|
100
|
The Timing and Extent of Motor Neuron Vulnerability in ALS Correlates with Accumulation of Misfolded SOD1 Protein in the Cortex and in the Spinal Cord. Cells 2020; 9:cells9020502. [PMID: 32098365 PMCID: PMC7072754 DOI: 10.3390/cells9020502] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 12/11/2022] Open
Abstract
Understanding the cellular and molecular basis of selective vulnerability has been challenging, especially for motor neuron diseases. Developing drugs that improve the health of neurons that display selective vulnerability relies on in vivo cell-based models and quantitative readout measures that translate to patient outcome. We initially developed and characterized UCHL1-eGFP mice, in which motor neurons are labeled with eGFP that is stable and long-lasting. By crossing UCHL1-eGFP to amyotrophic lateral sclerosis (ALS) disease models, we generated ALS mouse models with fluorescently labeled motor neurons. Their examination over time began to reveal the cellular basis of selective vulnerability even within the related motor neuron pools. Accumulation of misfolded SOD1 protein both in the corticospinal and spinal motor neurons over time correlated with the timing and extent of degeneration. This further proved simultaneous degeneration of both upper and lower motor neurons, and the requirement to consider both upper and lower motor neuron populations in drug discovery efforts. Demonstration of the direct correlation between misfolded SOD1 accumulation and motor neuron degeneration in both cortex and spinal cord is important for building cell-based assays in vivo. Our report sets the stage for shifting focus from mice to diseased neurons for drug discovery efforts, especially for motor neuron diseases.
Collapse
|