51
|
The pathogenesis of amyotrophic lateral sclerosis: Mitochondrial dysfunction, protein misfolding and epigenetics. Brain Res 2022; 1786:147904. [PMID: 35390335 DOI: 10.1016/j.brainres.2022.147904] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/24/2022] [Accepted: 04/01/2022] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease with multiple complex mechanisms involved. Among them, mitochondrial dysfunction plays an important role in ALS. Multiple studies have shown that mitochondria are closely associated with reactive oxygen species production and oxidative stress and exhibit different functional states in different genetic backgrounds. In this review we explored the roles of Ca2+, autophagy, mitochondrial quality control in the regulation of mitochondrial homeostasis and their relationship with ALS. In addition, we also summarized and analyzed the roles of protein misfolding and abnormal aggregation in the pathogenesis of ALS. Moreover, we also discussed how epigenetic mechanisms such as DNA methylation and protein post-translational modification affect initiation and progression of ALS. Nevertheless, existing events still cannot fully explain the pathogenesis of ALS at present, more studies are required to explore pathological mechanisms of ALS.
Collapse
|
52
|
Gomes C, Sequeira C, Likhite S, Dennys CN, Kolb SJ, Shaw PJ, Vaz AR, Kaspar BK, Meyer K, Brites D. Neurotoxic Astrocytes Directly Converted from Sporadic and Familial ALS Patient Fibroblasts Reveal Signature Diversities and miR-146a Theragnostic Potential in Specific Subtypes. Cells 2022; 11:cells11071186. [PMID: 35406750 PMCID: PMC8997588 DOI: 10.3390/cells11071186] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/21/2022] [Accepted: 03/28/2022] [Indexed: 12/15/2022] Open
Abstract
A lack of stratification methods in patients with amyotrophic lateral sclerosis (ALS) is likely implicated in therapeutic failures. Regional diversities and pathophysiological abnormalities in astrocytes from mice with SOD1 mutations (mSOD1-ALS) can now be explored in human patients using somatic cell reprogramming. Here, fibroblasts from four sporadic (sALS) and three mSOD1-ALS patients were transdifferentiated into induced astrocytes (iAstrocytes). ALS iAstrocytes were neurotoxic toward HB9-GFP mouse motor neurons (MNs) and exhibited subtype stratification through GFAP, CX43, Ki-67, miR-155 and miR-146a expression levels. Up- (two cases) and down-regulated (three cases) miR-146a values in iAstrocytes were recapitulated in their secretome, either free or as cargo in small extracellular vesicles (sEVs). We previously showed that the neuroprotective phenotype of depleted miR-146 mSOD1 cortical astrocytes was reverted by its mimic. Thus, we tested such modulation in the most miR-146a-depleted patient-iAstrocytes (one sALS and one mSOD1-ALS). The miR-146a mimic in ALS iAstrocytes counteracted their reactive/inflammatory profile and restored miR-146a levels in sEVs. A reduction in lysosomal activity and enhanced synaptic/axonal transport-related genes in NSC-34 MNs occurred after co-culture with miR-146a-modulated iAstrocytes. In summary, the regulation of miR-146a in depleted ALS astrocytes may be key in reestablishing their normal function and in restoring MN lysosomal/synaptic dynamic plasticity in disease sub-groups.
Collapse
Affiliation(s)
- Cátia Gomes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (C.G.); (C.S.); (A.R.V.)
- The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (S.L.); (C.N.D.); (B.K.K.); (K.M.)
| | - Catarina Sequeira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (C.G.); (C.S.); (A.R.V.)
| | - Shibi Likhite
- The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (S.L.); (C.N.D.); (B.K.K.); (K.M.)
| | - Cassandra N. Dennys
- The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (S.L.); (C.N.D.); (B.K.K.); (K.M.)
| | - Stephen J. Kolb
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43214, USA;
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK;
| | - Ana R. Vaz
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (C.G.); (C.S.); (A.R.V.)
- Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Brian K. Kaspar
- The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (S.L.); (C.N.D.); (B.K.K.); (K.M.)
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kathrin Meyer
- The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (S.L.); (C.N.D.); (B.K.K.); (K.M.)
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (C.G.); (C.S.); (A.R.V.)
- Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Correspondence: ; Tel.: +351-217946450
| |
Collapse
|
53
|
Shupik MA, Gutner UA, Ustyugov AA, Rezvykh AP, Funikov SY, Maloshitskaya OA, Sokolov SA, Lebedev AT, Alessenko AV. Changes in the Metabolism of Sphingomyelin and Ceramide in the Brain Structures and Spinal Cord of Transgenic Mice (FUS(1-359)) Modeling Amyotrophic Lateral Sclerosis. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022010137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
54
|
Liu X, Pimm ML, Haarer B, Brawner AT, Henty-Ridilla JL. Biochemical characterization of actin assembly mechanisms with ALS-associated profilin variants. Eur J Cell Biol 2022; 101:151212. [PMID: 35248815 PMCID: PMC10163920 DOI: 10.1016/j.ejcb.2022.151212] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/26/2022] Open
Abstract
Eight separate mutations in the actin-binding protein profilin-1 have been identified as a rare cause of amyotrophic lateral sclerosis (ALS). Profilin is essential for many neuronal cell processes through its regulation of lipids, nuclear signals, and cytoskeletal dynamics, including actin filament assembly. Direct interactions between profilin and actin monomers inhibit actin filament polymerization. In contrast, profilin can also stimulate polymerization by simultaneously binding actin monomers and proline-rich tracts found in other proteins. Whether the ALS-associated mutations in profilin compromise these actin assembly functions is unclear. We performed a quantitative biochemical comparison of the direct and formin mediated impact for the eight ALS-associated profilin variants on actin assembly using classic protein-binding and single-filament microscopy assays. We determined that the binding constant of each profilin for actin monomers generally correlates with the actin nucleation strength associated with each ALS-related profilin. In the presence of formin, the A20T, R136W, Q139L, and C71G variants failed to activate the elongation phase of actin assembly. This diverse range of formin-activities is not fully explained through profilin-poly-L-proline (PLP) interactions, as all ALS-associated variants bind a formin-derived PLP peptide with similar affinities. However, chemical denaturation experiments suggest that the folding stability of these profilins impact some of these effects on actin assembly. Thus, changes in profilin protein stability and alterations in actin filament polymerization may both contribute to the profilin-mediated actin disruptions in ALS.
Collapse
Affiliation(s)
- Xinbei Liu
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Morgan L Pimm
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Brian Haarer
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Andrew T Brawner
- Department of Neuroscience & Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Jessica L Henty-Ridilla
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Neuroscience & Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
55
|
Arjmand B, Kokabi Hamidpour S, Rabbani Z, Tayanloo-Beik A, Rahim F, Aghayan HR, Larijani B. Organ on a Chip: A Novel in vitro Biomimetic Strategy in Amyotrophic Lateral Sclerosis (ALS) Modeling. Front Neurol 2022; 12:788462. [PMID: 35111126 PMCID: PMC8802668 DOI: 10.3389/fneur.2021.788462] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/20/2021] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis is a pernicious neurodegenerative disorder that is associated with the progressive degeneration of motor neurons, the disruption of impulse transmission from motor neurons to muscle cells, and the development of mobility impairments. Clinically, muscle paralysis can spread to other parts of the body. Hence it may have adverse effects on swallowing, speaking, and even breathing, which serves as major problems facing these patients. According to the available evidence, no definite treatment has been found for amyotrophic lateral sclerosis (ALS) that results in a significant outcome, although some pharmacological and non-pharmacological treatments are currently applied that are accompanied by some positive effects. In other words, available therapies are only used to relieve symptoms without any significant treatment effects that highlight the importance of seeking more novel therapies. Unfortunately, the process of discovering new drugs with high therapeutic potential for ALS treatment is fraught with challenges. The lack of a broad view of the disease process from early to late-stage and insufficiency of preclinical studies for providing validated results prior to conducting clinical trials are other reasons for the ALS drug discovery failure. However, increasing the combined application of different fields of regenerative medicine, especially tissue engineering and stem cell therapy can be considered as a step forward to develop more novel technologies. For instance, organ on a chip is one of these technologies that can provide a platform to promote a comprehensive understanding of neuromuscular junction biology and screen candidate drugs for ALS in combination with pluripotent stem cells (PSCs). The structure of this technology is based on the use of essential components such as iPSC- derived motor neurons and iPSC-derived skeletal muscle cells on a single miniaturized chip for ALS modeling. Accordingly, an organ on a chip not only can mimic ALS complexities but also can be considered as a more cost-effective and time-saving disease modeling platform in comparison with others. Hence, it can be concluded that lab on a chip can make a major contribution as a biomimetic micro-physiological system in the treatment of neurodegenerative disorders such as ALS.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Babak Arjmand
| | - Shayesteh Kokabi Hamidpour
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Rabbani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fakher Rahim
- Health Research Institute, Thalassemia, and Hemoglobinopathies Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Bagher Larijani
| |
Collapse
|
56
|
Branchereau P, Cattaert D. Chloride Homeostasis in Developing Motoneurons. ADVANCES IN NEUROBIOLOGY 2022; 28:45-61. [PMID: 36066820 DOI: 10.1007/978-3-031-07167-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Maturation of GABA/Glycine chloride-mediated synaptic inhibitions is crucial for the establishment of a balance between excitation and inhibition. GABA and glycine are excitatory neurotransmitters on immature neurons that exhibit elevated [Cl-]i. Later in development [Cl-]i drops leading to the occurrence of inhibitory synaptic activity. This ontogenic change is closely correlated to a differential expression of two cation-chloride cotransporters that are the Cl- channel K+/Cl- co-transporter type 2 (KCC2) that extrudes Cl- ions and the Na+-K+-2Cl- cotransporter NKCC1 that accumulates Cl- ions. The classical scheme built from studies performed on cortical and hippocampal networks proposes that immature neurons display high [Cl-]i because NKCC1 is overexpressed compared to KCC2 and that the co-transporters ratio reverses in mature neurons, lowering [Cl-]i. In this chapter, we will see that this classical scheme is not true in motoneurons (MNs) and that an early alteration of the chloride homeostasis may be involved in pathological conditions.
Collapse
Affiliation(s)
- Pascal Branchereau
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), Univ. Bordeaux, UMR 5287, CNRS, Bordeaux, France.
| | - Daniel Cattaert
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA), Univ. Bordeaux, UMR 5287, CNRS, Bordeaux, France
| |
Collapse
|
57
|
Russo K, Wharton KA. BMP/TGF-β signaling as a modulator of neurodegeneration in ALS. Dev Dyn 2022; 251:10-25. [PMID: 33745185 PMCID: PMC11929146 DOI: 10.1002/dvdy.333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 12/19/2022] Open
Abstract
This commentary focuses on the emerging intersection between BMP/TGF-β signaling roles in nervous system function and the amyotrophic lateral sclerosis (ALS) disease state. Future research is critical to elucidate the molecular underpinnings of this intersection of the cellular processes disrupted in ALS and those influenced by BMP/TGF-β signaling, including synapse structure, neurotransmission, plasticity, and neuroinflammation. Such knowledge promises to inform us of ideal entry points for the targeted modulation of dysfunctional cellular processes in an effort to abrogate ALS pathologies. It is likely that different interventions are required, either at discrete points in disease progression, or across multiple dysfunctional processes which together lead to motor neuron degeneration and death. We discuss the challenging, but intriguing idea that modulation of the pleiotropic nature of BMP/TGF-β signaling could be advantageous, as a way to simultaneously treat defects in more than one cell process across different forms of ALS.
Collapse
Affiliation(s)
- Kathryn Russo
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island, USA
| | - Kristi A Wharton
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island, USA
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
58
|
Contingent intramuscular boosting of P2XR7 axis improves motor function in transgenic ALS mice. Cell Mol Life Sci 2021; 79:7. [PMID: 34936028 PMCID: PMC8695421 DOI: 10.1007/s00018-021-04070-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/06/2022]
Abstract
Amyotrophic lateral sclerosis is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons and severe muscle atrophy without effective treatment. Most research on the disease has been focused on studying motor neurons and supporting cells of the central nervous system. Strikingly, the recent observations have suggested that morpho-functional alterations in skeletal muscle precede motor neuron degeneration, bolstering the interest in studying muscle tissue as a potential target for the delivery of therapies. We previously showed that the systemic administration of the P2XR7 agonist, 2′(3′)-O‐(4-benzoylbenzoyl) adenosine 5-triphosphate (BzATP), enhanced the metabolism and promoted the myogenesis of new fibres in the skeletal muscles of SOD1G93A mice. Here we further corroborated this evidence showing that intramuscular administration of BzATP improved the motor performance of ALS mice by enhancing satellite cells and the muscle pro-regenerative activity of infiltrating macrophages. The preservation of the skeletal muscle retrogradely propagated along with the motor unit, suggesting that backward signalling from the muscle could impinge on motor neuron death. In addition to providing the basis for a suitable adjunct multisystem therapeutic approach in ALS, these data point out that the muscle should be at the centre of ALS research as a target tissue to address novel therapies in combination with those oriented to the CNS.
Collapse
|
59
|
Bonifacino T, Zerbo RA, Balbi M, Torazza C, Frumento G, Fedele E, Bonanno G, Milanese M. Nearly 30 Years of Animal Models to Study Amyotrophic Lateral Sclerosis: A Historical Overview and Future Perspectives. Int J Mol Sci 2021; 22:ijms222212236. [PMID: 34830115 PMCID: PMC8619465 DOI: 10.3390/ijms222212236] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, multigenic, multifactorial, and non-cell autonomous neurodegenerative disease characterized by upper and lower motor neuron loss. Several genetic mutations lead to ALS development and many emerging gene mutations have been discovered in recent years. Over the decades since 1990, several animal models have been generated to study ALS pathology including both vertebrates and invertebrates such as yeast, worms, flies, zebrafish, mice, rats, guinea pigs, dogs, and non-human primates. Although these models show different peculiarities, they are all useful and complementary to dissect the pathological mechanisms at the basis of motor neuron degeneration and ALS progression, thus contributing to the development of new promising therapeutics. In this review, we describe the up to date and available ALS genetic animal models, classified by the different genetic mutations and divided per species, pointing out their features in modeling, the onset and progression of the pathology, as well as their specific pathological hallmarks. Moreover, we highlight similarities, differences, advantages, and limitations, aimed at helping the researcher to select the most appropriate experimental animal model, when designing a preclinical ALS study.
Collapse
Affiliation(s)
- Tiziana Bonifacino
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Genoa, Italy
| | - Roberta Arianna Zerbo
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Matilde Balbi
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Carola Torazza
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Giulia Frumento
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
| | - Ernesto Fedele
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence:
| | - Giambattista Bonanno
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Marco Milanese
- Pharmacology and Toxicology Unit, Department of Pharmacy, University of Genoa, 16148 Genoa, Italy; (T.B.); (R.A.Z.); (M.B.); (C.T.); (G.F.); (G.B.); (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Genoa, Italy
| |
Collapse
|
60
|
Thompson AG, Oeckl P, Feneberg E, Bowser R, Otto M, Fischer R, Kessler B, Turner MR. Advancing mechanistic understanding and biomarker development in amyotrophic lateral sclerosis. Expert Rev Proteomics 2021; 18:977-994. [PMID: 34758687 DOI: 10.1080/14789450.2021.2004890] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Proteomic analysis has contributed significantly to the study of the neurodegenerative disease amyotrophic lateral sclerosis (ALS). It has helped to define the pathological change common to nearly all cases, namely intracellular aggregates of phosphorylated TDP-43, shifting the focus of pathogenesis in ALS toward RNA biology. Proteomics has also uniquely underpinned the delineation of disease mechanisms in model systems and has been central to recent advances in human ALS biomarker development. AREAS COVERED The contribution of proteomics to understanding the cellular pathological changes, disease mechanisms, and biomarker development in ALS are covered. EXPERT OPINION Proteomics has delivered unique insights into the pathogenesis of ALS and advanced the goal of objective measurements of disease activity to improve therapeutic trials. Further developments in sensitivity and quantification are expected, with application to the presymptomatic phase of human disease offering the hope of prevention strategies.
Collapse
Affiliation(s)
| | - Patrick Oeckl
- Department of Neurology, University of Ulm, Ulm, Germany.,German Center for Neurodegenerative Diseases (Dzne e.V.), Ulm, Germany
| | - Emily Feneberg
- Department of Neurology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Robert Bowser
- Departments of Neurology and Translational Neuroscience, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany.,Department of Neurology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Benedikt Kessler
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
61
|
Moreno-Jiménez L, Benito-Martín M, Sanclemente-Alamán I, Matías-Guiu J, Sancho-Bielsa F, Canales-Aguirre A, Mateos-Díaz J, Matías-Guiu J, Aguilar J, Gómez-Pinedo U. Modelos experimentales murinos en la esclerosis lateral amiotrófica. Puesta al día. Neurologia 2021. [DOI: 10.1016/j.nrl.2021.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
62
|
Tejido C, Pakravan D, Bosch LVD. Potential Therapeutic Role of HDAC Inhibitors in FUS-ALS. Front Mol Neurosci 2021; 14:686995. [PMID: 34434087 PMCID: PMC8380926 DOI: 10.3389/fnmol.2021.686995] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/29/2021] [Indexed: 01/05/2023] Open
Abstract
Mutations in the FUS gene cause amyotrophic lateral sclerosis (ALS-FUS). However, the exact pathogenic mechanism of mutant fused in sarcoma (FUS) protein is not completely understood. FUS is an RNA binding protein (RBP) localized predominantly in the nucleus, but ALS-linked FUS mutations can affect its nuclear localization signal impairing its import into the nucleus. This mislocalization to the cytoplasm facilitates FUS aggregation in cytoplasmic inclusions. Therapies targeting post translational modifications are rising as new treatments for ALS, in particular acetylation which could have a role in the dynamics of RBPs. Research using histone deacetylase (HDAC) inhibitors in FUS-ALS models showed that HDACs can influence cytoplasmic FUS localization. Inhibition of HDACs could promote acetylation of the FUS RNA binding domain (RRM) and altering its RNA interactions resulting in FUS maintenance in the nucleus. In addition, acetylation of FUS RRMs might also favor or disfavor its incorporation into pathological inclusions. In this review, we summarize and discuss the evidence for the potential role of HDACs in the context of FUS-ALS and we propose a new hypothesis based on this overview.
Collapse
Affiliation(s)
- Clara Tejido
- Vlaams Instituut voor Biotechnologie (VIB), Center for Brain and Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Donya Pakravan
- Vlaams Instituut voor Biotechnologie (VIB), Center for Brain and Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurosciences, Experimental Neurology, Leuven Brain Institute, Katholieke Universiteit Leuven (KU Leuven)-University of Leuven, Leuven, Belgium
| | - Ludo Van Den Bosch
- Vlaams Instituut voor Biotechnologie (VIB), Center for Brain and Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,Department of Neurosciences, Experimental Neurology, Leuven Brain Institute, Katholieke Universiteit Leuven (KU Leuven)-University of Leuven, Leuven, Belgium
| |
Collapse
|
63
|
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
|
64
|
Wang X, Zhang JB, He KJ, Wang F, Liu CF. Advances of Zebrafish in Neurodegenerative Disease: From Models to Drug Discovery. Front Pharmacol 2021; 12:713963. [PMID: 34335276 PMCID: PMC8317260 DOI: 10.3389/fphar.2021.713963] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disease (NDD), including Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis, are characterized by the progressive loss of neurons which leads to the decline of motor and/or cognitive function. Currently, the prevalence of NDD is rapidly increasing in the aging population. However, valid drugs or treatment for NDD are still lacking. The clinical heterogeneity and complex pathogenesis of NDD pose a great challenge for the development of disease-modifying therapies. Numerous animal models have been generated to mimic the pathological conditions of these diseases for drug discovery. Among them, zebrafish (Danio rerio) models are progressively emerging and becoming a powerful tool for in vivo study of NDD. Extensive use of zebrafish in pharmacology research or drug screening is due to the high conserved evolution and 87% homology to humans. In this review, we summarize the zebrafish models used in NDD studies, and highlight the recent findings on pharmacological targets for NDD treatment. As high-throughput platforms in zebrafish research have rapidly developed in recent years, we also discuss the application prospects of these new technologies in future NDD research.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jin-Bao Zhang
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Kai-Jie He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Fen Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, Suqian First Hospital, Suqian, China
| |
Collapse
|
65
|
Lotz SK, Blackhurst BM, Reagin KL, Funk KE. Microbial Infections Are a Risk Factor for Neurodegenerative Diseases. Front Cell Neurosci 2021; 15:691136. [PMID: 34305533 PMCID: PMC8292681 DOI: 10.3389/fncel.2021.691136] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, comprise a family of disorders characterized by progressive loss of nervous system function. Neuroinflammation is increasingly recognized to be associated with many neurodegenerative diseases but whether it is a cause or consequence of the disease process is unclear. Of growing interest is the role of microbial infections in inciting degenerative neuroinflammatory responses and genetic factors that may regulate those responses. Microbial infections cause inflammation within the central nervous system through activation of brain-resident immune cells and infiltration of peripheral immune cells. These responses are necessary to protect the brain from lethal infections but may also induce neuropathological changes that lead to neurodegeneration. This review discusses the molecular and cellular mechanisms through which microbial infections may increase susceptibility to neurodegenerative diseases. Elucidating these mechanisms is critical for developing targeted therapeutic approaches that prevent the onset and slow the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Kristen E. Funk
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| |
Collapse
|
66
|
Fernández-Ruiz J, de Lago E, Rodríguez-Cueto C, Moro MA. Recent advances in the pathogenesis and therapeutics of amyotrophic lateral sclerosis. Br J Pharmacol 2021; 178:1253-1256. [PMID: 33638898 DOI: 10.1111/bph.15348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Javier Fernández-Ruiz
- IUIN, CIBERNED and IRYCIS, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Eva de Lago
- IUIN, CIBERNED and IRYCIS, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Carmen Rodríguez-Cueto
- IUIN, CIBERNED and IRYCIS, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - María A Moro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Instituto de Salud Carlos III, IUIN and I+12, Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| |
Collapse
|
67
|
Tau and TDP-43 synergy: a novel therapeutic target for sporadic late-onset Alzheimer's disease. GeroScience 2021; 43:1627-1634. [PMID: 34185246 PMCID: PMC8492812 DOI: 10.1007/s11357-021-00407-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/16/2021] [Indexed: 11/10/2022] Open
Abstract
Alzheimer’s disease (AD) is traditionally defined by the presence of two types of protein aggregates in the brain: amyloid plaques comprised of the protein amyloid-β (Aβ) and neurofibrillary tangles containing the protein tau. However, a large proportion (up to 57%) of AD patients also have TDP-43 aggregates present as an additional comorbid pathology. The presence of TDP-43 aggregates in AD correlates with hippocampal sclerosis, worse brain atrophy, more severe cognitive impairment, and more rapid cognitive decline. In patients with mixed Aβ, tau, and TDP-43 pathology, TDP-43 may interact with neurodegenerative processes in AD, worsening outcomes. While considerable progress has been made to characterize TDP-43 pathology in AD and late-onset dementia, there remains a critical need for mechanistic studies to understand underlying disease biology and develop therapeutic interventions. This perspectives article reviews the current understanding of these processes from autopsy cohort studies and model organism-based research, and proposes targeting neurotoxic synergies between tau and TDP-43 as a new therapeutic strategy for AD with comorbid TDP-43 pathology.
Collapse
|
68
|
Braems E, Tziortzouda P, Van Den Bosch L. Exploring the alternative: Fish, flies and worms as preclinical models for ALS. Neurosci Lett 2021; 759:136041. [PMID: 34118308 DOI: 10.1016/j.neulet.2021.136041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 04/15/2021] [Accepted: 06/01/2021] [Indexed: 12/22/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disorder characterized by the loss of upper and lower motor neurons. In general, patients succumb to respiratory insufficiency due to respiratory muscle weakness. Despite many promising therapeutic strategies primarily identified in rodent models, patient trials remain rather unsuccessful. There is a clear need for alternative approaches, which could provide directions towards the justified use of rodents and which increase the likelihood to identify new promising clinical candidates. In the last decades, the use of fast genetic approaches and the development of high-throughput screening platforms in the nematode Caenorhabditis elegans, in the fruit fly (Drosophila melanogaster) and in zebrafish (Danio rerio) have contributed to new insights into ALS pathomechanisms, disease modifiers and therapeutic targets. In this mini-review, we provide an overview of these alternative small animal studies, modeling the most common ALS genes and discuss the most recent preclinical discoveries. We conclude that small animal models will not replace rodent models, yet they clearly represent an important asset for preclinical studies.
Collapse
Affiliation(s)
- Elke Braems
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Paraskevi Tziortzouda
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.
| |
Collapse
|
69
|
Overexpression of miR-124 in Motor Neurons Plays a Key Role in ALS Pathological Processes. Int J Mol Sci 2021; 22:ijms22116128. [PMID: 34200161 PMCID: PMC8201298 DOI: 10.3390/ijms22116128] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
miRNA(miR)-124 is an important regulator of neurogenesis, but its upregulation in SOD1G93A motor neurons (mSOD1 MNs) was shown to associate with neurodegeneration and microglia activation. We used pre-miR-124 in wild-type (WT) MNs and anti-miR-124 in mSOD1 MNs to characterize the miR-124 pathological role. miR-124 overexpression in WT MNs produced a miRNA profile like that of mSOD1 MNs (high miR-125b; low miR-146a and miR-21), and similarly led to early apoptosis. Alterations in mSOD1 MNs were abrogated with anti-miR-124 and changes in their miRNAs mostly recapitulated by their secretome. Normalization of miR-124 levels in mSOD1 MNs prevented the dysregulation of neurite network, mitochondria dynamics, axonal transport, and synaptic signaling. Same alterations were observed in WT MNs after pre-miR-124 transfection. Secretome from mSOD1 MNs triggered spinal microglia activation, which was unno-ticed with that from anti-miR-124-modulated cells. Secretome from such modulated MNs, when added to SC organotypic cultures from mSOD1 mice in the early symptomatic stage, also coun-teracted the pathology associated to GFAP decrease, PSD-95 and CX3CL1-CX3CR1 signaling im-pairment, neuro-immune homeostatic imbalance, and enhanced miR-124 expression levels. Data suggest that miR-124 is implicated in MN degeneration and paracrine-mediated pathogenicity. We propose miR-124 as a new therapeutic target and a promising ALS biomarker in patient sub-populations.
Collapse
|
70
|
Chua JP, De Calbiac H, Kabashi E, Barmada SJ. Autophagy and ALS: mechanistic insights and therapeutic implications. Autophagy 2021; 18:254-282. [PMID: 34057020 PMCID: PMC8942428 DOI: 10.1080/15548627.2021.1926656] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mechanisms of protein homeostasis are crucial for overseeing the clearance of misfolded and toxic proteins over the lifetime of an organism, thereby ensuring the health of neurons and other cells of the central nervous system. The highly conserved pathway of autophagy is particularly necessary for preventing and counteracting pathogenic insults that may lead to neurodegeneration. In line with this, mutations in genes that encode essential autophagy factors result in impaired autophagy and lead to neurodegenerative conditions such as amyotrophic lateral sclerosis (ALS). However, the mechanistic details underlying the neuroprotective role of autophagy, neuronal resistance to autophagy induction, and the neuron-specific effects of autophagy-impairing mutations remain incompletely defined. Further, the manner and extent to which non-cell autonomous effects of autophagy dysfunction contribute to ALS pathogenesis are not fully understood. Here, we review the current understanding of the interplay between autophagy and ALS pathogenesis by providing an overview of critical steps in the autophagy pathway, with special focus on pivotal factors impaired by ALS-causing mutations, their physiologic effects on autophagy in disease models, and the cell type-specific mechanisms regulating autophagy in non-neuronal cells which, when impaired, can contribute to neurodegeneration. This review thereby provides a framework not only to guide further investigations of neuronal autophagy but also to refine therapeutic strategies for ALS and related neurodegenerative diseases.Abbreviations: ALS: amyotrophic lateral sclerosis; Atg: autophagy-related; CHMP2B: charged multivesicular body protein 2B; DPR: dipeptide repeat; FTD: frontotemporal dementia; iPSC: induced pluripotent stem cell; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; PINK1: PTEN induced kinase 1; RNP: ribonuclear protein; sALS: sporadic ALS; SPHK1: sphingosine kinase 1; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK-binding kinase 1; TFEB: transcription factor EB; ULK: unc-51 like autophagy activating kinase; UPR: unfolded protein response; UPS: ubiquitin-proteasome system; VCP: valosin containing protein.
Collapse
Affiliation(s)
- Jason P Chua
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Hortense De Calbiac
- Recherche translationnelle sur les maladies neurologiques, Institut Imagine, UMR-1163 INSERM et Université Paris Descartes, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Edor Kabashi
- Recherche translationnelle sur les maladies neurologiques, Institut Imagine, UMR-1163 INSERM et Université Paris Descartes, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Sami J Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
71
|
Plesia M, Stevens OA, Lloyd GR, Kendall CA, Coldicott I, Kennerley AJ, Miller G, Shaw PJ, Mead RJ, Day JCC, Alix JJP. In Vivo Fiber Optic Raman Spectroscopy of Muscle in Preclinical Models of Amyotrophic Lateral Sclerosis and Duchenne Muscular Dystrophy. ACS Chem Neurosci 2021; 12:1768-1776. [PMID: 33950665 PMCID: PMC8154326 DOI: 10.1021/acschemneuro.0c00794] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/30/2021] [Indexed: 12/13/2022] Open
Abstract
Neuromuscular diseases result in muscle weakness, disability, and, in many instances, death. Preclinical models form the bedrock of research into these disorders, and the development of in vivo and potentially translational biomarkers for the accurate identification of disease is crucial. Spontaneous Raman spectroscopy can provide a rapid, label-free, and highly specific molecular fingerprint of tissue, making it an attractive potential biomarker. In this study, we have developed and tested an in vivo intramuscular fiber optic Raman technique in two mouse models of devastating human neuromuscular diseases, amyotrophic lateral sclerosis, and Duchenne muscular dystrophy (SOD1G93A and mdx, respectively). The method identified diseased and healthy muscle with high classification accuracies (area under the receiver operating characteristic curves (AUROC): 0.76-0.92). In addition, changes in diseased muscle over time were also identified (AUROCs 0.89-0.97). Key spectral changes related to proteins and the loss of α-helix protein structure. Importantly, in vivo recording did not cause functional motor impairment and only a limited, resolving tissue injury was seen on high-resolution magnetic resonance imaging. Lastly, we demonstrate that ex vivo muscle from human patients with these conditions produced similar spectra to those observed in mice. We conclude that spontaneous Raman spectroscopy of muscle shows promise as a translational research tool.
Collapse
Affiliation(s)
- Maria Plesia
- Sheffield
Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Oliver A. Stevens
- Interface
Analysis Centre, School of Physics, University
of Bristol, Bristol BS8 1TL, UK
| | - Gavin R. Lloyd
- Phenome
Centre Birmingham, University of Birmingham, Birmingham B15 2TT, UK
- Biophotonics
Research Unit, Gloucestershire Hospitals
NHS Foundation Trust, Gloucester GL1 3NN, UK
| | - Catherine A. Kendall
- Biophotonics
Research Unit, Gloucestershire Hospitals
NHS Foundation Trust, Gloucester GL1 3NN, UK
| | - Ian Coldicott
- Sheffield
Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | | | - Gaynor Miller
- Department
of Oncology and Metabolism, University of
Sheffield, Sheffield S10 2RX, UK
| | - Pamela J. Shaw
- Sheffield
Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Cross-Faculty
Neuroscience Institute, University of Sheffield, Sheffield S10 2HQ, UK
| | - Richard J. Mead
- Sheffield
Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Cross-Faculty
Neuroscience Institute, University of Sheffield, Sheffield S10 2HQ, UK
| | - John C. C. Day
- Interface
Analysis Centre, School of Physics, University
of Bristol, Bristol BS8 1TL, UK
| | - James J. P. Alix
- Sheffield
Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Cross-Faculty
Neuroscience Institute, University of Sheffield, Sheffield S10 2HQ, UK
| |
Collapse
|
72
|
Rodríguez-Cueto C, Gómez-Almería M, García Toscano L, Romero J, Hillard CJ, de Lago E, Fernández-Ruiz J. Inactivation of the CB 2 receptor accelerated the neuropathological deterioration in TDP-43 transgenic mice, a model of amyotrophic lateral sclerosis. Brain Pathol 2021; 31:e12972. [PMID: 33983653 PMCID: PMC8549023 DOI: 10.1111/bpa.12972] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/29/2021] [Accepted: 04/21/2021] [Indexed: 11/29/2022] Open
Abstract
The activation of the cannabinoid receptor type‐2 (CB2) afforded neuroprotection in amyotrophic lateral sclerosis (ALS) models. The objective of this study was to further investigate the relevance of the CB2 receptor through investigating the consequences of its inactivation. TDP‐43(A315T) transgenic mice were crossed with CB2 receptor knock‐out mice to generate double mutants. Temporal and qualitative aspects of the pathological phenotype of the double mutants were compared to TDP‐43 transgenic mice expressing the CB2 receptor. The double mutants exhibited significantly accelerated neurological decline, such that deteriorated rotarod performance was visible at 7 weeks, whereas rotarod performance was normal up to 11 weeks in transgenic mice with intact expression of the CB2 receptor. A morphological analysis of spinal cords confirmed an earlier death (visible at 65 days) of motor neurons labelled with Nissl staining and ChAT immunofluorescence in double mutants compared to TDP‐43 transgenic mice expressing the CB2 receptor. Evidence of glial reactivity, measured using GFAP and Iba‐1 immunostaining, was seen in double mutants at 65 days, but not in TDP‐43 transgenic mice expressing the CB2 receptor. However, at 90 days, both genotypes exhibited similar changes for all these markers, although surviving motor neurons of transgenic mice presented some morphological abnormalities in absence of the CB2 receptor that were not as evident in the presence of this receptor. This faster deterioration seen in double mutants led to premature mortality compared with TDP‐43 transgenic mice expressing the CB2 receptor. We also investigated the consequences of a pharmacological inactivation of the CB2 receptor using the selective antagonist AM630 in TDP‐43 transgenic mice, but results showed only subtle trends towards a greater deterioration. In summary, our results confirmed the potential of the CB2 receptor agonists as a neuroprotective therapy in ALS and strongly support the need to progress towards an evaluation of this potential in patients.
Collapse
Affiliation(s)
- Carmen Rodríguez-Cueto
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Marta Gómez-Almería
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain
| | - Laura García Toscano
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Julián Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Eva de Lago
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Javier Fernández-Ruiz
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| |
Collapse
|
73
|
Region-specific vulnerability in neurodegeneration: lessons from normal ageing. Ageing Res Rev 2021; 67:101311. [PMID: 33639280 PMCID: PMC8024744 DOI: 10.1016/j.arr.2021.101311] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/22/2021] [Indexed: 02/07/2023]
Abstract
Why neurodegenerative disease pathology is regionally restricted remains elusive. Regions selectively prone to neurodegeneration are also vulnerable to normal ageing. Nervous system tissue, cellular and molecular ageing may determine regional vulnerability. Differential ageing can conceptually extend from an individual to subcellular scale. An understanding of region-specific vulnerability might guide therapeutic advances.
A number of age-associated neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD) and amyotrophic lateral sclerosis (ALS), possess a shared characteristic of region-specific neurodegeneration. However, the mechanisms which determine why particular regions within the nervous system are selectively vulnerable to neurodegeneration, whilst others remain relatively unaffected throughout disease progression, remain elusive. Here, we review how regional susceptibility to the ubiquitous physiological phenomenon of normal ageing might underlie the vulnerability of these same regions to neurodegeneration, highlighting three regions archetypally associated with AD, PD and ALS (the hippocampus, substantia nigra pars compacta and ventral spinal cord, respectively), as especially prone to age-related alterations. Placing particular emphasis on these three regions, we comprehensively explore differential regional susceptibility to nervous system tissue, cellular and molecular level ageing to provide an integrated perspective on why age-related neurodegenerative diseases exhibit region-selective vulnerability. Combining these principles with increasingly recognised differences between chronological and biological ageing (termed differential or ‘delta’ ageing) might ultimately guide therapeutic approaches for these devastating neurodegenerative diseases, for which a paucity of disease modifying and/or life promoting treatments currently exist.
Collapse
|
74
|
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
|
75
|
Abstract
Cigarette smoke (CS) has been consistently demonstrated to be an environmental risk factor for amyotrophic lateral sclerosis (ALS), although the molecular pathogenic mechanisms involved are yet to be elucidated. Here, we propose different mechanisms by which CS exposure can cause sporadic ALS pathogenesis. Oxidative stress and neuroinflammation are widely implicated in ALS pathogenesis, with blood–spinal cord barrier disruption also recognised to be involved in the disease process. In addition, immunometabolic, epigenetic and microbiome alterations have been implicated in ALS recently. Identification of the underlying pathophysiological mechanisms that underpin CS-associated ALS will drive future research to be conducted into new targets for treatment.
Collapse
|
76
|
Moisse M, Zwamborn RAJ, van Vugt J, van der Spek R, van Rheenen W, Kenna B, Van Eijk K, Kenna K, Corcia P, Couratier P, Vourc'h P, Hardiman O, McLaughin R, Gotkine M, Drory V, Ticozzi N, Silani V, de Carvalho M, Mora Pardina JS, Povedano M, Andersen PM, Weber M, Başak NA, Chen X, Eberle MA, Al‐Chalabi A, Shaw C, Shaw PJ, Morrison KE, Landers JE, Glass JD, Robberecht W, van Es M, van den Berg L, Veldink J, Van Damme P. The Effect of SMN Gene Dosage on ALS Risk and Disease Severity. Ann Neurol 2021; 89:686-697. [PMID: 33389754 PMCID: PMC8048961 DOI: 10.1002/ana.26009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 02/01/2023]
Abstract
OBJECTIVE The role of the survival of motor neuron (SMN) gene in amyotrophic lateral sclerosis (ALS) is unclear, with several conflicting reports. A decisive result on this topic is needed, given that treatment options are available now for SMN deficiency. METHODS In this largest multicenter case control study to evaluate the effect of SMN1 and SMN2 copy numbers in ALS, we used whole genome sequencing data from Project MinE data freeze 2. SMN copy numbers of 6,375 patients with ALS and 2,412 controls were called from whole genome sequencing data, and the reliability of the calls was tested with multiplex ligation-dependent probe amplification data. RESULTS The copy number distribution of SMN1 and SMN2 between cases and controls did not show any statistical differences (binomial multivariate logistic regression SMN1 p = 0.54 and SMN2 p = 0.49). In addition, the copy number of SMN did not associate with patient survival (Royston-Parmar; SMN1 p = 0.78 and SMN2 p = 0.23) or age at onset (Royston-Parmar; SMN1 p = 0.75 and SMN2 p = 0.63). INTERPRETATION In our well-powered study, there was no association of SMN1 or SMN2 copy numbers with the risk of ALS or ALS disease severity. This suggests that changing SMN protein levels in the physiological range may not modify ALS disease course. This is an important finding in the light of emerging therapies targeted at SMN deficiencies. ANN NEUROL 2021;89:686-697.
Collapse
Affiliation(s)
- Matthieu Moisse
- Departments of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain and Disease ResearchLeuvenBelgium
| | - Ramona A. J. Zwamborn
- Department of NeurologyUMC Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| | - Joke van Vugt
- Department of NeurologyUMC Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| | - Rick van der Spek
- Department of NeurologyUMC Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| | - Wouter van Rheenen
- Department of NeurologyUMC Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| | - Brendan Kenna
- Department of NeurologyUMC Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| | - Kristel Van Eijk
- Department of NeurologyUMC Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| | - Kevin Kenna
- Department of NeurologyUMC Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| | - Philippe Corcia
- Centre SLA, CHRU de ToursToursFrance
- UMR 1253, iBrain, Université de Tours, InsermToursFrance
| | | | | | - Orla Hardiman
- Academic Unit of NeurologyTrinity College Dublin, Trinity Biomedical Sciences InstituteDublinRepublic of Ireland
| | - Russell McLaughin
- Complex Trait Genomics LaboratorySmurfit Institute of Genetics, Trinity College DublinDublinRepublic of Ireland
| | - Marc Gotkine
- The Agnes Ginges Center for Human NeurogeneticsHadassah‐Hebrew University Medical CenterJerusalemIsrael
| | - Vivian Drory
- Department of NeurologyTel‐Aviv Sourasky Medical CentreTel AvivIsrael
| | - Nicola Ticozzi
- Department of Neurology, Stroke Unit and Laboratory of NeuroscienceIRCCS Istituto Auxologico ItalianoMilanoItaly
- Department of Pathophysiology and Transplantation“Dino Ferrari” Center, Università degli Studi di MilanoMilanItaly
| | - Vincenzo Silani
- Department of Neurology, Stroke Unit and Laboratory of NeuroscienceIRCCS Istituto Auxologico ItalianoMilanoItaly
- Department of Pathophysiology and Transplantation“Dino Ferrari” Center, Università degli Studi di MilanoMilanItaly
| | - Mamede de Carvalho
- Instituto de Fisiologia, Instituto de Medicina Molecular, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
| | | | | | - Peter M. Andersen
- Department of Clinical Science, NeurosciencesUmeå UniversityUmeåSweden
| | - Markus Weber
- Neuromuscular Diseases Unit/ALS ClinicSt. GallenSwitzerland
| | - Nazli A. Başak
- Koç University, School of Medicine, KUTTAM‐NDALIstanbulTurkey
| | | | | | - Ammar Al‐Chalabi
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience Institute, King's College LondonLondonUK
| | - Chris Shaw
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience Institute, King's College LondonLondonUK
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience (SITraN)University of SheffieldSheffieldUK
| | - Karen E. Morrison
- School of Medicine, Dentistry, and Biomedical SciencesQueen's University BelfastBelfastUK
| | - John E. Landers
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMA
| | | | - Wim Robberecht
- Departments of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Department of NeurologyUniversity Hospitals LeuvenLeuvenBelgium
| | - Michael van Es
- Department of NeurologyUMC Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| | - Leonard van den Berg
- Department of NeurologyUMC Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| | - Jan Veldink
- Department of NeurologyUMC Utrecht Brain Center, Utrecht UniversityUtrechtThe Netherlands
| | - Philip Van Damme
- Departments of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain and Disease ResearchLeuvenBelgium
- Department of NeurologyUniversity Hospitals LeuvenLeuvenBelgium
| | | |
Collapse
|
77
|
Bovio F, Sciandrone B, Urani C, Fusi P, Forcella M, Regonesi ME. Superoxide dismutase 1 (SOD1) and cadmium: A three models approach to the comprehension of its neurotoxic effects. Neurotoxicology 2021; 84:125-135. [PMID: 33774064 DOI: 10.1016/j.neuro.2021.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/19/2021] [Accepted: 03/19/2021] [Indexed: 12/13/2022]
Abstract
Cadmium (Cd) is a widespread toxic environmental contaminant, released by anthropogenic activities. It interferes with essential metal ions homeostasis and affects protein structures and functions by substituting zinc, copper and iron. In this study, the effect of cadmium on SOD1, a CuZn metalloenzyme catalyzing superoxide conversion into hydrogen peroxide, has been investigated in three different biological models. We first evaluated the effects of cadmium combined with copper and/or zinc on the recombinant GST-SOD1, expressed in E. coli BL21. The enzyme activity and expression were investigated in the presence of fixed copper and/or zinc doses with different cadmium concentrations, in the cellular medium. Cadmium caused a dose-dependent reduction in SOD1 activity, while the expression remains constant. Similar results were obtained in the cellular model represented by the human SH-SY5Y neuronal cell line. After cadmium treatment for 24 and 48 h, SOD1 enzymatic activity decreased in a dose- and time-dependent way, while the protein expression remained constant. Finally, a 16 h cadmium treatment caused a 25 % reduction of CuZn-SOD activity without affecting the protein expression in the Caenorhabditis elegans model. Taken together our results show an inhibitory effect of cadmium on SOD1 enzymatic activity, without affecting the protein expression, in all the biological models used, suggesting that cadmium can displace zinc from the enzyme catalytic site.
Collapse
Affiliation(s)
- Federica Bovio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy
| | - Barbara Sciandrone
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy
| | - Chiara Urani
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126, Milan, Italy; Integrated Models for Prevention and Protection in Environmental and Occupational Health, (MISTRAL), Interuniversity Research Center, Italy
| | - Paola Fusi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy; Integrated Models for Prevention and Protection in Environmental and Occupational Health, (MISTRAL), Interuniversity Research Center, Italy.
| | - Matilde Forcella
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy.
| | - Maria Elena Regonesi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza, 2, 20126, Milan, Italy; Milan Center of Neuroscience (NeuroMI), 20126, Milan, Italy
| |
Collapse
|
78
|
Bedlack R, Barkhaus P, Barnes B, Bereman M, Bertorini T, Carter G, Crayle J, Kihuwa-Mani S, Bowser R, Kittrell P, McDermott C, Pattee G, Salmon K, Wicks P. ALSUntangled #60: light therapy. Amyotroph Lateral Scler Frontotemporal Degener 2021; 23:315-319. [PMID: 33683159 DOI: 10.1080/21678421.2021.1883668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
ALSUntangled reviews alternative and off-label treatments for people with ALS. Here we review light therapy. We show that it has theoretically plausible mechanisms, three flawed pre-clinical data, studies, and one incompletely documented case report supporting its use. We explain why further studies are needed to determine whether any specific light therapy protocol can help people with ALS.
Collapse
Affiliation(s)
| | - Paul Barkhaus
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ben Barnes
- Department of Neurology, Augusta University Medical Center, Augusta, GA, USA
| | - Michael Bereman
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA
| | - Tulio Bertorini
- Department of Neurology, Methodist University Hospital, Memphis, TN, USA
| | - Gregory Carter
- Department of Neurology, St Lukes Rehabilitation Hospital, Chesterfield, MO, USA
| | - Jesse Crayle
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Sky Kihuwa-Mani
- University of North Carolina at Greensboro, Lloyd International Honors College, Greensboro, NC, USA
| | - Robert Bowser
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Pamela Kittrell
- Department of Neurology, University of Texas Health Science Center, Houston, TX, USA
| | | | - Gary Pattee
- Department of Neurology, Nebraska Medicine, Omaha, NE, USA
| | - Kristiana Salmon
- Department of Neurology, McGill Centre for Research in Neuroscience, Montreal, QC, Canada
| | | |
Collapse
|
79
|
Klingl YE, Pakravan D, Van Den Bosch L. Opportunities for histone deacetylase inhibition in amyotrophic lateral sclerosis. Br J Pharmacol 2021; 178:1353-1372. [PMID: 32726472 PMCID: PMC9327724 DOI: 10.1111/bph.15217] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease. ALS patients suffer from a progressive loss of motor neurons, leading to respiratory failure within 3 to 5 years after diagnosis. Available therapies only slow down the disease progression moderately or extend the lifespan by a few months. Epigenetic hallmarks have been linked to the disease, creating an avenue for potential therapeutic approaches. Interference with one class of epigenetic enzymes, histone deacetylases, has been shown to affect neurodegeneration in many preclinical models. Consequently, it is crucial to improve our understanding about histone deacetylases and their inhibitors in (pre)clinical models of ALS. We conclude that selective inhibitors with high tolerability and safety and sufficient blood-brain barrier permeability will be needed to interfere with both epigenetic and non-epigenetic targets of these enzymes. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.
Collapse
Affiliation(s)
- Yvonne E. Klingl
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI)KU Leuven‐University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
| | - Donya Pakravan
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI)KU Leuven‐University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI)KU Leuven‐University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
| |
Collapse
|
80
|
Beckers J, Tharkeshwar AK, Van Damme P. C9orf72 ALS-FTD: recent evidence for dysregulation of the autophagy-lysosome pathway at multiple levels. Autophagy 2021; 17:3306-3322. [PMID: 33632058 PMCID: PMC8632097 DOI: 10.1080/15548627.2021.1872189] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two clinically distinct classes of neurodegenerative disorders. Yet, they share a range of genetic, cellular, and molecular features. Hexanucleotide repeat expansions (HREs) in the C9orf72 gene and the accumulation of toxic protein aggregates in the nervous systems of the affected individuals are among such common features. Though the mechanisms by which HREs cause toxicity is not clear, the toxic gain of function due to transcribed HRE RNA or dipeptide repeat proteins (DPRs) produced by repeat-associated non-AUG translation together with a reduction in C9orf72 expression are proposed as the contributing factors for disease pathogenesis in ALS and FTD. In addition, several recent studies point toward alterations in protein homeostasis as one of the root causes of the disease pathogenesis. In this review, we discuss the effects of the C9orf72 HRE in the autophagy-lysosome pathway based on various recent findings. We suggest that dysfunction of the autophagy-lysosome pathway synergizes with toxicity from C9orf72 repeat RNA and DPRs to drive disease pathogenesis. Abbreviation: ALP: autophagy-lysosome pathway; ALS: amyotrophic lateral sclerosis; AMPK: AMP-activated protein kinase; ATG: autophagy-related; ASO: antisense oligonucleotide; C9orf72: C9orf72-SMCR8 complex subunit; DENN: differentially expressed in normal and neoplastic cells; DPR: dipeptide repeat protein; EIF2A/eIF2α: eukaryotic translation initiation factor 2A; ER: endoplasmic reticulum; FTD: frontotemporal dementia; GAP: GTPase-activating protein; GEF: guanine nucleotide exchange factor; HRE: hexanucleotide repeat expansion; iPSC: induced pluripotent stem cell; ISR: integrated stress response; M6PR: mannose-6-phosphate receptor, cation dependent; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MN: motor neuron; MTORC1: mechanistic target of rapamycin kinase complex 1; ND: neurodegenerative disorder; RAN: repeat-associated non-ATG; RB1CC1/FIP200: RB1 inducible coiled-coil 1; SLC66A1/PQLC2: solute carrier family 66 member 1; SMCR8: SMCR8-C9orf72 complex subunit; SQSTM1/p62: sequestosome 1; STX17: syntaxin 17; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK binding kinase 1; TFEB: transcription factor EB; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system; WDR41: WD repeat domain 41.
Collapse
Affiliation(s)
- Jimmy Beckers
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Arun Kumar Tharkeshwar
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.,University Hospitals Leuven, Department of Neurology, Leuven, Belgium
| |
Collapse
|
81
|
Rodríguez-Cueto C, García-Toscano L, Santos-García I, Gómez-Almería M, Gonzalo-Consuegra C, Espejo-Porras F, Fernández-Ruiz J, de Lago E. Targeting the CB 2 receptor and other endocannabinoid elements to delay disease progression in amyotrophic lateral sclerosis. Br J Pharmacol 2021; 178:1373-1387. [PMID: 33486755 DOI: 10.1111/bph.15386] [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: 10/19/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 02/06/2023] Open
Abstract
Cannabinoids form a singular group of plant-derived compounds, endogenous lipids and synthetic derivatives with multiple therapeutic effects exerted by targeting different elements of the endocannabinoid system. One of their therapeutic applications is the preservation of neuronal integrity exerted by attenuating the multiple neurotoxic events that kill neurons in neurodegenerative disorders. In this review, we will address the potential of cannabinoids as neuroprotective agents in amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disorder characterized by muscle denervation, atrophy and paralysis, and progressive deterioration in upper and/or lower motor neurons. The emphasis will be paid on the cannabinoid type 2 (CB2 ) receptor, whose activation limits glial reactivity, but the potential of additional endocannabinoid-related targets will be also addressed. The evidence accumulated so far at the preclinical level supports the need to soon move towards the patients and initiate clinical trials to confirm the potential of cannabinoid-based medicines as disease modifiers in ALS. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.
Collapse
Affiliation(s)
- Carmen Rodríguez-Cueto
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.,Area 1 - Neurociencias y Organos de los Sentidos, IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Laura García-Toscano
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.,Area 1 - Neurociencias y Organos de los Sentidos, IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Irene Santos-García
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.,Area 1 - Neurociencias y Organos de los Sentidos, IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Marta Gómez-Almería
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.,Area 1 - Neurociencias y Organos de los Sentidos, IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Claudia Gonzalo-Consuegra
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.,Area 1 - Neurociencias y Organos de los Sentidos, IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Francisco Espejo-Porras
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.,Area 1 - Neurociencias y Organos de los Sentidos, IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Javier Fernández-Ruiz
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.,Area 1 - Neurociencias y Organos de los Sentidos, IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Eva de Lago
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.,Area 1 - Neurociencias y Organos de los Sentidos, IRYCIS, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| |
Collapse
|
82
|
Johnson MA, Deng Q, Taylor G, McEachin ZT, Chan AWS, Root J, Bassell GJ, Kukar T. Divergent FUS phosphorylation in primate and mouse cells following double-strand DNA damage. Neurobiol Dis 2020; 146:105085. [PMID: 32950644 PMCID: PMC8064403 DOI: 10.1016/j.nbd.2020.105085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022] Open
Abstract
Fused in sarcoma (FUS) is a RNA/DNA protein involved in multiple nuclear and cytoplasmic functions including transcription, splicing, mRNA trafficking, and stress granule formation. To accomplish these many functions, FUS must shuttle between cellular compartments in a highly regulated manner. When shuttling is disrupted, FUS abnormally accumulates into cytoplasmic inclusions that can be toxic. Disrupted shuttling of FUS into the nucleus is a hallmark of ~10% of frontotemporal lobar degeneration (FTLD) cases, the neuropathology that underlies frontotemporal dementia (FTD). Multiple pathways are known to disrupt nuclear/cytoplasmic shuttling of FUS. In earlier work, we discovered that double-strand DNA breaks (DSBs) trigger DNA-dependent protein kinase (DNA-PK) to phosphorylate FUS (p-FUS) at N-terminal residues leading to the cytoplasmic accumulation of FUS. Therefore, DNA damage may contribute to the development of FTLD pathology with FUS inclusions. In the present study, we examined how DSBs effect FUS phosphorylation in various primate and mouse cellular models. All cell lines derived from human and non-human primates exhibit N-terminal FUS phosphorylation following calicheamicin γ1 (CLM) induced DSBs. In contrast, we were unable to detect FUS phosphorylation in mouse-derived primary neurons or immortalized cell lines regardless of CLM treatment, duration, or concentration. Despite DNA damage induced by CLM treatment, we find that mouse cells do not phosphorylate FUS, likely due to reduced levels and activity of DNA-PK compared to human cells. Taken together, our work reveals that mouse-derived cellular models regulate FUS in an anomalous manner compared to primate cells. This raises the possibility that mouse models may not fully recapitulate the pathogenic cascades that lead to FTLD with FUS pathology.
Collapse
Affiliation(s)
- Michelle A Johnson
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta, GA, United States of America; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta, GA, United States of America
| | - Qiudong Deng
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta, GA, United States of America; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta, GA, United States of America
| | - Georgia Taylor
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta, GA, United States of America; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta, GA, United States of America
| | - Zachary T McEachin
- Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta, GA, United States of America; Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, United States of America; Laboratory of Translational Cell Biology, Emory University, School of Medicine, Atlanta, GA, United States of America
| | - Anthony W S Chan
- Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta, GA, United States of America; Department of Human Genetics, Emory University, School of Medicine, Atlanta, GA, United States of America; Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, 954 Gatewood Rd, NE, Atlanta, GA, United States of America
| | - Jessica Root
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta, GA, United States of America; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta, GA, United States of America
| | - Gary J Bassell
- Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta, GA, United States of America; Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, United States of America; Laboratory of Translational Cell Biology, Emory University, School of Medicine, Atlanta, GA, United States of America
| | - Thomas Kukar
- Department of Pharmacology and Chemical Biology, Emory University, School of Medicine, Atlanta, GA, United States of America; Center for Neurodegenerative Disease, Emory University, School of Medicine, Atlanta, GA, United States of America; Department of Neurology, Emory University, School of Medicine, Atlanta, GA, United States of America.
| |
Collapse
|
83
|
Eleutherio ECA, Silva Magalhães RS, de Araújo Brasil A, Monteiro Neto JR, de Holanda Paranhos L. SOD1, more than just an antioxidant. Arch Biochem Biophys 2020; 697:108701. [PMID: 33259795 DOI: 10.1016/j.abb.2020.108701] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022]
Abstract
During cellular respiration, radicals, such as superoxide, are produced, and in a large concentration, they may cause cell damage. To combat this threat, the cell employs the enzyme Cu/Zn Superoxide Dismutase (SOD1), which converts the radical superoxide into molecular oxygen and hydrogen peroxide, through redox reactions. Although this is its main function, recent studies have shown that the SOD1 has other functions that deviates from its original one including activation of nuclear gene transcription or as an RNA binding protein. This comprehensive review looks at the most important aspects of human SOD1 (hSOD1), including the structure, properties, and characteristics as well as transcriptional and post-translational modifications (PTM) that the enzyme can receive and their effects, and its many functions. We also discuss the strategies currently used to analyze it to better understand its participation in diseases linked to hSOD1 including Amyotrophic Lateral Sclerosis (ALS), cancer, and Parkinson.
Collapse
|
84
|
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
|
85
|
Caldwell KA, Willicott CW, Caldwell GA. Modeling neurodegeneration in Caenorhabditis elegans. Dis Model Mech 2020; 13:13/10/dmm046110. [PMID: 33106318 PMCID: PMC7648605 DOI: 10.1242/dmm.046110] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The global burden of neurodegenerative diseases underscores the urgent need for innovative strategies to define new drug targets and disease-modifying factors. The nematode Caenorhabditis elegans has served as the experimental subject for multiple transformative discoveries that have redefined our understanding of biology for ∼60 years. More recently, the considerable attributes of C. elegans have been applied to neurodegenerative diseases, including amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease and Huntington's disease. Transgenic nematodes with genes encoding normal and disease variants of proteins at the single- or multi-copy level under neuronal-specific promoters limits expression to select neuronal subtypes. The anatomical transparency of C. elegans affords the use of co-expressed fluorescent proteins to follow the progression of neurodegeneration as the animals age. Significantly, a completely defined connectome facilitates detailed understanding of the impact of neurodegeneration on organismal health and offers a unique capacity to accurately link cell death with behavioral dysfunction or phenotypic variation in vivo. Moreover, chemical treatments, as well as forward and reverse genetic screening, hasten the identification of modifiers that alter neurodegeneration. When combined, these chemical-genetic analyses establish critical threshold states to enhance or reduce cellular stress for dissecting associated pathways. Furthermore, C. elegans can rapidly reveal whether lifespan or healthspan factor into neurodegenerative processes. Here, we outline the methodologies employed to investigate neurodegeneration in C. elegans and highlight numerous studies that exemplify its utility as a pre-clinical intermediary to expedite and inform mammalian translational research. Summary: While unsurpassed as an experimental system for fundamental biology, Caenorhabditis elegans remains undervalued for its translational potential. Here, we highlight significant outcomes from, and resources available for, C. elegans-based research into neurodegenerative disorders.
Collapse
Affiliation(s)
- Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA .,Departments of Neurobiology, Neurology, Center for Neurodegeneration and Experimental Therapeutics, and Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corey W Willicott
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA.,Departments of Neurobiology, Neurology, Center for Neurodegeneration and Experimental Therapeutics, and Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
86
|
Pasteuning-Vuhman S, de Jongh R, Timmers A, Pasterkamp RJ. Towards Advanced iPSC-based Drug Development for Neurodegenerative Disease. Trends Mol Med 2020; 27:263-279. [PMID: 33121873 DOI: 10.1016/j.molmed.2020.09.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases (NDDs) are a heterogeneous group of diseases that are characterized by the progressive loss of neurons leading to motor, sensory, and/or cognitive defects. Currently, NDDs are not curable and treatment focuses on alleviating symptoms and halting disease progression. Phenotypic heterogeneity between individual NDD patients, lack of robust biomarkers, the limited translational potential of experimental models, and other factors have hampered drug development for the treatment of NDDs. This review summarizes and discusses the use of induced pluripotent stem cell (iPSC) approaches for improving drug discovery and testing. It highlights challenges associated with iPSC modeling and also discusses innovative approaches such as brain organoids and microfluidic-based technology which will improve drug development for NDDs.
Collapse
Affiliation(s)
- Svetlana Pasteuning-Vuhman
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Rianne de Jongh
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Annabel Timmers
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
87
|
Mechanistic insights into the loss-of-function mechanisms of rare human D-amino acid oxidase variants implicated in amyotrophic lateral sclerosis. Sci Rep 2020; 10:17146. [PMID: 33051492 PMCID: PMC7555490 DOI: 10.1038/s41598-020-74048-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Impaired enzymatic activity in D-amino acid oxidase (DAAO) caused by missense mutations has been shown to trigger amyotrophic lateral sclerosis (ALS) through an abnormal accumulation of D-serine in the spinal cord. While loss of enzymatic functions of certain ALS-causing DAAO variants have been studied before, a detailed understanding of structure-dynamics-function relationship of the rare DAAO variants has not been investigated hitherto. To address this, we carried out a comprehensive study of all the reported rare DAAO variants. By employing a spectrum of bioinformatics analyses along with extensive structural dynamics simulations, we show that certain rare variants disrupted key interactions with the active site and decreased the conformational flexibility of active site loop comprising residues 216-228, which is essential for substrate binding and product release. Moreover, these variants lost crucial interactions with the cofactor flavin-adenine-dinucleotide, resulting in weaker binding affinity. A detailed inspection revealed that these variants exhibited such characteristics due to the abrogation of specific salt bridges. Taken together, our study provides a gateway into the structural-dynamic features of the rare DAAO variants and highlights the importance of informatics-based integrated analyses in the screening and prioritization of variants a priori to the clinical-functional characterization.
Collapse
|
88
|
Lattante S, Marangi G, Doronzio PN, Conte A, Bisogni G, Zollino M, Sabatelli M. High-Throughput Genetic Testing in ALS: The Challenging Path of Variant Classification Considering the ACMG Guidelines. Genes (Basel) 2020; 11:genes11101123. [PMID: 32987860 PMCID: PMC7600768 DOI: 10.3390/genes11101123] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/15/2020] [Accepted: 09/22/2020] [Indexed: 12/17/2022] Open
Abstract
The development of high-throughput sequencing technologies and screening of big patient cohorts with familial and sporadic amyotrophic lateral sclerosis (ALS) led to the identification of a significant number of genetic variants, which are sometimes difficult to interpret. The American College of Medical Genetics and Genomics (ACMG) provided guidelines to help molecular geneticists and pathologists to interpret variants found in laboratory testing. We assessed the application of the ACMG criteria to ALS-related variants, combining data from literature with our experience. We analyzed a cohort of 498 ALS patients using massive parallel sequencing of ALS-associated genes and identified 280 variants with a minor allele frequency < 1%. Examining all variants using the ACMG criteria, thus considering the type of variant, inheritance, familial segregation, and possible functional studies, we classified 20 variants as “pathogenic”. In conclusion, ALS’s genetic complexity, such as oligogenic inheritance, presence of genes acting as risk factors, and reduced penetrance, needs to be considered when interpreting variants. The goal of this work is to provide helpful suggestions to geneticists and clinicians dealing with ALS.
Collapse
Affiliation(s)
- Serena Lattante
- Section of Genomic Medicine, Department of Life Sciences and Public Health, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Roma, Italy; (S.L.); (P.N.D.); (M.Z.)
- Complex Operational Unit of Medical Genetics, Department of Laboratory and Infectious Disease Sciences, A. Gemelli University Hospital Foundation IRCCS, 00168 Roma, Italy
| | - Giuseppe Marangi
- Section of Genomic Medicine, Department of Life Sciences and Public Health, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Roma, Italy; (S.L.); (P.N.D.); (M.Z.)
- Complex Operational Unit of Medical Genetics, Department of Laboratory and Infectious Disease Sciences, A. Gemelli University Hospital Foundation IRCCS, 00168 Roma, Italy
- Correspondence: ; Tel.: +39-0630154606
| | - Paolo Niccolò Doronzio
- Section of Genomic Medicine, Department of Life Sciences and Public Health, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Roma, Italy; (S.L.); (P.N.D.); (M.Z.)
- Complex Operational Unit of Medical Genetics, Department of Laboratory and Infectious Disease Sciences, A. Gemelli University Hospital Foundation IRCCS, 00168 Roma, Italy
| | - Amelia Conte
- Adult NEMO Clinical Center, Complex Operational Unit of Neurology, Department of Aging, Neurological, Orthopedic and Head-Neck Sciences, A. Gemelli University Hospital Foundation IRCCS, 00168 Roma, Italy; (A.C.); (G.B.); (M.S.)
| | - Giulia Bisogni
- Adult NEMO Clinical Center, Complex Operational Unit of Neurology, Department of Aging, Neurological, Orthopedic and Head-Neck Sciences, A. Gemelli University Hospital Foundation IRCCS, 00168 Roma, Italy; (A.C.); (G.B.); (M.S.)
| | - Marcella Zollino
- Section of Genomic Medicine, Department of Life Sciences and Public Health, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Roma, Italy; (S.L.); (P.N.D.); (M.Z.)
- Complex Operational Unit of Medical Genetics, Department of Laboratory and Infectious Disease Sciences, A. Gemelli University Hospital Foundation IRCCS, 00168 Roma, Italy
| | - Mario Sabatelli
- Adult NEMO Clinical Center, Complex Operational Unit of Neurology, Department of Aging, Neurological, Orthopedic and Head-Neck Sciences, A. Gemelli University Hospital Foundation IRCCS, 00168 Roma, Italy; (A.C.); (G.B.); (M.S.)
- Section of Neurology, Department of Neuroscience, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Roma, Italy
| |
Collapse
|
89
|
Soo SK, Rudich PD, Traa A, Harris-Gauthier N, Shields HJ, Van Raamsdonk JM. Compounds that extend longevity are protective in neurodegenerative diseases and provide a novel treatment strategy for these devastating disorders. Mech Ageing Dev 2020; 190:111297. [PMID: 32610099 PMCID: PMC7484136 DOI: 10.1016/j.mad.2020.111297] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
While aging is the greatest risk factor for the development of neurodegenerative disease, the role of aging in these diseases is poorly understood. In the inherited forms of these diseases, the disease-causing mutation is present from birth but symptoms appear decades later. This indicates that these mutations are well tolerated in younger individuals but not in older adults. Based on this observation, we hypothesized that changes taking place during normal aging make the cells in the brain (and elsewhere) susceptible to the disease-causing mutations. If so, then delaying some of these age-related changes may be beneficial in the treatment of neurodegenerative disease. In this review, we examine the effects of five compounds that have been shown to extend longevity (metformin, rapamycin, resveratrol, N-acetyl-l-cysteine, curcumin) in four of the most common neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis). While not all investigations observe a beneficial effect of these compounds, there are multiple studies that show a protective effect of each of these lifespan-extending compounds in animal models of neurodegenerative disease. Combined with genetic studies, this suggests the possibility that targeting the aging process may be an effective strategy to treat neurodegenerative disease.
Collapse
Affiliation(s)
- Sonja K Soo
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H4A 3J1, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Paige D Rudich
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H4A 3J1, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Annika Traa
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H4A 3J1, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Namasthée Harris-Gauthier
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H4A 3J1, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Hazel J Shields
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H4A 3J1, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Jeremy M Van Raamsdonk
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, H4A 3J1, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, H4A 3J1, Canada; Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, H4A 3J1, Canada; Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
90
|
Younas N, Zafar S, Shafiq M, Noor A, Siegert A, Arora AS, Galkin A, Zafar A, Schmitz M, Stadelmann C, Andreoletti O, Ferrer I, Zerr I. SFPQ and Tau: critical factors contributing to rapid progression of Alzheimer's disease. Acta Neuropathol 2020; 140:317-339. [PMID: 32577828 PMCID: PMC7423812 DOI: 10.1007/s00401-020-02178-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/10/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022]
Abstract
Dysfunctional RNA-binding proteins (RBPs) have been implicated in several neurodegenerative disorders. Recently, this paradigm of RBPs has been extended to pathophysiology of Alzheimer’s disease (AD). Here, we identified disease subtype specific variations in the RNA-binding proteome (RBPome) of sporadic AD (spAD), rapidly progressive AD (rpAD), and sporadic Creutzfeldt Jakob disease (sCJD), as well as control cases using RNA pull-down assay in combination with proteomics. We show that one of these identified proteins, splicing factor proline and glutamine rich (SFPQ), is downregulated in the post-mortem brains of rapidly progressive AD patients, sCJD patients and 3xTg mice brain at terminal stage of the disease. In contrast, the expression of SFPQ was elevated at early stage of the disease in the 3xTg mice, and in vitro after oxidative stress stimuli. Strikingly, in rpAD patients’ brains SFPQ showed a significant dislocation from the nucleus and cytoplasmic colocalization with TIA-1. Furthermore, in rpAD brain lesions, SFPQ and p-tau showed extranuclear colocalization. Of note, association between SFPQ and tau-oligomers in rpAD brains suggests a possible role of SFPQ in oligomerization and subsequent misfolding of tau protein. In line with the findings from the human brain, our in vitro study showed that SFPQ is recruited into TIA-1-positive stress granules (SGs) after oxidative stress induction, and colocalizes with tau/p-tau in these granules, providing a possible mechanism of SFPQ dislocation through pathological SGs. Furthermore, the expression of human tau in vitro induced significant downregulation of SFPQ, suggesting a causal role of tau in the downregulation of SFPQ. The findings from the current study indicate that the dysregulation and dislocation of SFPQ, the subsequent DNA-related anomalies and aberrant dynamics of SGs in association with pathological tau represents a critical pathway which contributes to rapid progression of AD.
Collapse
Affiliation(s)
- Neelam Younas
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Saima Zafar
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany.
- Biomedical Engineering and Sciences Department, School of Mechanical and Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Islamabad, Pakistan.
| | - Mohsin Shafiq
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Aneeqa Noor
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Anna Siegert
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Amandeep Singh Arora
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
- Institute for Behavioral Medicine Research, The Ohio State University, 460 Medical Center Dr, Columbus, OH, 43210, USA
| | - Alexey Galkin
- St. Petersburg Branch, Vavilov Institute of General Genetics, St. Petersburg, Russia
| | - Ayesha Zafar
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- College of Medicine Center for Pharmacogenomics, The Ohio State University, 460 W 12th Avenue, Columbus, OH, 1004 BRT, USA
| | - Mathias Schmitz
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | | | - Olivier Andreoletti
- UMR INRA ENVT 1225- Interactions Hôte Agent Pathogène-École Nationale Vétérinaire de Toulouse, Toulouse, France
| | - Isidre Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
- Bellvitge University Hospital-IDIBELL, Barcelona, Spain
- CIBERNED, Barcelona, Spain
- Hospitalet de Llobregat, Barcelona, Spain
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen and the German Center for Neurodegenerative Diseases (DZNE), Robert-Koch-Straße 40, 37075, Göttingen, Germany.
| |
Collapse
|
91
|
Spencer KR, Foster ZW, Rauf NA, Guilderson L, Collins D, Averill JG, Walker SE, Robey I, Cherry JD, Alvarez VE, Huber BR, McKee AC, Kowall NW, Brady CB, Stein TD. Neuropathological profile of long-duration amyotrophic lateral sclerosis in military Veterans. Brain Pathol 2020; 30:1028-1040. [PMID: 32633852 PMCID: PMC8018169 DOI: 10.1111/bpa.12876] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder affecting both the upper and lower motor neurons. Although ALS typically leads to death within 3 to 5 years after initial symptom onset, approximately 10% of patients with ALS live more than 10 years after symptom onset. We set out to determine similarities and differences in clinical presentation and neuropathology in persons with ALS with long vs. those with standard duration. Participants were United States military Veterans with a pathologically confirmed diagnosis of ALS (n = 179), dichotomized into standard duration (<10 years) and long-duration (≥10 years). The ALS Functional Rating Scale-Revised (ALSFRS-R) was administered at study entry and semi-annually thereafter until death. Microglial density was determined in a subset of participants. long-duration ALS occurred in 76 participants (42%) with a mean disease duration of 16.3 years (min/max = 10.1/42.2). Participants with long-duration ALS were younger at disease onset (P = 0.002), had a slower initial ALS symptom progression on the ALSFRS-R (P < 0.001) and took longer to diagnose (P < 0.002) than standard duration ALS. Pathologically, long-duration ALS was associated with less frequent TDP-43 pathology (P < 0.001). Upper motor neuron degeneration was similar; however, long-duration ALS participants had less severe lower motor neuron degeneration at death (P < 0.001). In addition, the density of microglia was decreased in the corticospinal tract (P = 0.017) and spinal cord anterior horn (P = 0.009) in long-duration ALS. Notably, many neuropathological markers of ALS were similar between the standard and long-duration groups and there was no difference in the frequency of known ALS genetic mutations. These findings suggest that the lower motor neuron system is relatively spared in long-duration ALS and that pathological progression is likely slowed by as yet unknown genetic and environmental modifiers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ian Robey
- Southern Arizona VA Healthcare SystemTucsonAZ
| | - Jonathan D. Cherry
- VA Boston Healthcare SystemBostonMA,Boston University Alzheimer's Disease and CTE Center, Boston University School of MedicineBostonMA,Department of Pathology and Laboratory MedicineBoston University School of MedicineBostonMA
| | - Victor E. Alvarez
- VA Boston Healthcare SystemBostonMA,Boston University Alzheimer's Disease and CTE Center, Boston University School of MedicineBostonMA,Department of NeurologyBoston University School of MedicineBostonMA,Department of Veterans Affairs Medical CenterBedfordMA
| | - Bertrand R. Huber
- VA Boston Healthcare SystemBostonMA,Boston University Alzheimer's Disease and CTE Center, Boston University School of MedicineBostonMA,Department of Veterans Affairs Medical CenterBedfordMA
| | - Ann C. McKee
- VA Boston Healthcare SystemBostonMA,Boston University Alzheimer's Disease and CTE Center, Boston University School of MedicineBostonMA,Department of NeurologyBoston University School of MedicineBostonMA,Department of Veterans Affairs Medical CenterBedfordMA
| | - Neil W. Kowall
- VA Boston Healthcare SystemBostonMA,Boston University Alzheimer's Disease and CTE Center, Boston University School of MedicineBostonMA,Department of NeurologyBoston University School of MedicineBostonMA
| | - Christopher B. Brady
- VA Boston Healthcare SystemBostonMA,Department of NeurologyBoston University School of MedicineBostonMA,Division of AgingBrigham and Women's Hospital, Harvard Medical SchoolBostonMA
| | - Thor D. Stein
- VA Boston Healthcare SystemBostonMA,Boston University Alzheimer's Disease and CTE Center, Boston University School of MedicineBostonMA,Department of Pathology and Laboratory MedicineBoston University School of MedicineBostonMA,Department of Veterans Affairs Medical CenterBedfordMA
| |
Collapse
|
92
|
Clark CM, Clark RM, Hoyle JA, Dickson TC. Pathogenic or protective? Neuropeptide Y in amyotrophic lateral sclerosis. J Neurochem 2020; 156:273-289. [PMID: 32654149 DOI: 10.1111/jnc.15125] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/16/2020] [Accepted: 06/24/2020] [Indexed: 12/13/2022]
Abstract
Neuropeptide Y (NPY) is an endogenous peptide of the central and enteric nervous systems which has gained significant interest as a potential neuroprotective agent for treatment of neurodegenerative disease. Amyotrophic lateral sclerosis (ALS) is an aggressive and fatal neurodegenerative disease characterized by motor deficits and motor neuron loss. In ALS, recent evidence from ALS patients and animal models has indicated that NPY may have a role in the disease pathogenesis. Increased NPY levels were found to correlate with disease progression in ALS patients. Similarly, NPY expression is increased in the motor cortex of ALS mice by end stages of the disease. Although the functional consequence of increased NPY levels in ALS is currently unknown, NPY has been shown to exert a diverse range of neuroprotective roles in other neurodegenerative diseases; through modulation of potassium channel activity, increased production of neurotrophins, inhibition of endoplasmic reticulum stress and autophagy, reduction of excitotoxicity, oxidative stress, neuroinflammation and hyperexcitability. Several of these mechanisms and signalling pathways are heavily implicated in the pathogenesis of ALS. Therefore, in this review, we discuss possible effects of NPY and NPY-receptor signalling in the ALS disease context, as determining NPY's contribution to, or impact on, ALS disease mechanisms will be essential for future studies investigating the NPY system as a therapeutic strategy in this devastating disease.
Collapse
Affiliation(s)
- Courtney M Clark
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Rosemary M Clark
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Joshua A Hoyle
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Tracey C Dickson
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
93
|
Salazar JL, Yang SA, Yamamoto S. Post-Developmental Roles of Notch Signaling in the Nervous System. Biomolecules 2020; 10:biom10070985. [PMID: 32630239 PMCID: PMC7408554 DOI: 10.3390/biom10070985] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Since its discovery in Drosophila, the Notch signaling pathway has been studied in numerous developmental contexts in diverse multicellular organisms. The role of Notch signaling in nervous system development has been extensively investigated by numerous scientists, partially because many of the core Notch signaling components were initially identified through their dramatic ‘neurogenic’ phenotype of developing fruit fly embryos. Components of the Notch signaling pathway continue to be expressed in mature neurons and glia cells, which is suggestive of a role in the post-developmental nervous system. The Notch pathway has been, so far, implicated in learning and memory, social behavior, addiction, and other complex behaviors using genetic model organisms including Drosophila and mice. Additionally, Notch signaling has been shown to play a modulatory role in several neurodegenerative disease model animals and in mediating neural toxicity of several environmental factors. In this paper, we summarize the knowledge pertaining to the post-developmental roles of Notch signaling in the nervous system with a focus on discoveries made using the fruit fly as a model system as well as relevant studies in C elegans, mouse, rat, and cellular models. Since components of this pathway have been implicated in the pathogenesis of numerous psychiatric and neurodegenerative disorders in human, understanding the role of Notch signaling in the mature brain using model organisms will likely provide novel insights into the mechanisms underlying these diseases.
Collapse
Affiliation(s)
- Jose L. Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Sheng-An Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
- Department of Neuroscience, BCM, Houston, TX 77030, USA
- Program in Developmental Biology, BCM, Houston, TX 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-832-824-8119
| |
Collapse
|
94
|
Bendotti C, Bonetto V, Pupillo E, Logroscino G, Al-Chalabi A, Lunetta C, Riva N, Mora G, Lauria G, Weishaupt JH, Agosta F, Malaspina A, Basso M, Greensmith L, Van Den Bosch L, Ratti A, Corbo M, Hardiman O, Chiò A, Silani V, Beghi E. Focus on the heterogeneity of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:485-495. [PMID: 32583689 DOI: 10.1080/21678421.2020.1779298] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The clinical manifestations of amyotrophic lateral sclerosis (ALS) are variable in terms of age at disease onset, site of onset, progression of symptoms, motor neuron involvement, and the occurrence of cognitive and behavioral changes. Genetic background is a key determinant of the ALS phenotype. The mortality of the disease also varies with the ancestral origin of the affected population and environmental factors are likely to be associated with ALS at least within some cohorts. Disease heterogeneity is likely underpinned by the presence of different pathogenic mechanisms. A variety of ALS animal models can be informative about the heterogeneity of the neuropathological or genetic aspects of the disease and can support the development of new therapeutic intervention. Evolving biomarkers can contribute to the identification of differing genotypes and phenotypes, and can be used to explore whether genotypic and phenotypic differences in animal models might help to provide a better definition of the heterogeneity of ALS in humans. These include neurofilaments, peripheral blood mononuclear cells, extracellular vesicles, microRNA and imaging findings. These biomarkers might predict not only the development of the disease, but also the variability in progression, although robust validation is required. A promising area of progress in modeling the heterogeneity of human ALS is represented by the use of human induced pluripotent stem cell (iPSCs)-derived motor neurons. Although the translational value of iPSCs remains unclear, this model is attractive in the perspective of replicating the heterogeneity of sporadic ALS as a first step toward a personalized medicine strategy.
Collapse
Affiliation(s)
- Caterina Bendotti
- Mario Negri-ALS Study Group, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Valentina Bonetto
- Mario Negri-ALS Study Group, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Elisabetta Pupillo
- Mario Negri-ALS Study Group, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Giancarlo Logroscino
- Department of Neurosciences and Sense Organs, Center for Neurodegenerative Diseases and the Aging Brain Università degli Studi di Bari, Bari; Fondazione Giovanni Panico Tricase, Lecce, Italy
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Christian Lunetta
- NEuroMuscular Omnicentre (NEMO), Serena Onlus Foundation, Milano, Italy
| | - Nilo Riva
- Neuroimaging Research Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milano, Italy
| | - Gabriela Mora
- Department of Neurorehabilitation, ICS Maugeri IRCCS, Milano, Italy
| | - Giuseppe Lauria
- Unit of Neurology, Motor Neuron Disease Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy.,Department of Biomedical and Clinical Sciences "Lduigi Sacco", University of Milan, Milan, Italy
| | | | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milano, Italy
| | | | - Manuela Basso
- Mario Negri-ALS Study Group, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.,Department of Cellular, Computational and Integrative Biology (CIBIO), Università degli Studi di Trento, Trento, Italy
| | - Linda Greensmith
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Ludo Van Den Bosch
- Center for Brain & Disease Research (VIB) and Laboratory of Neurobiology (KU Leuven), Leuven, Belgium
| | - Antonia Ratti
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, Milano, Italy.,Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico (CCP), Milano, Italy
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Adriano Chiò
- "Rita Levi Montalcini" Department of Neuroscience, Università degli Studi di Torino, Torino, Italy
| | - Vincenzo Silani
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, Milano, Italy.,Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milano, Italy
| | - Ettore Beghi
- Mario Negri-ALS Study Group, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| |
Collapse
|
95
|
Shimizu Y, Ishii C, Yanobu-Takanashi R, Nakano K, Imaike A, Mita M, Hamase K, Okamura T. d-Amino acid oxidase deficiency is caused by a large deletion in the Dao gene in LEA rats. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140463. [PMID: 32512180 DOI: 10.1016/j.bbapap.2020.140463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/15/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022]
Abstract
d-Amino acids, enantiomers of l-amino acids, are increasingly recognized as physiologically active molecules as well as potential biomarkers for diseases. d-Amino acid oxidase (DAO) catalyzes the oxidative deamination of d-amino acids and is present in a wide variety of organisms from yeasts to humans. Previous studies indicated that LEA rats lacked DAO activity, and levels of d-Ser and d-Ala were markedly increased in their tissues, suggesting a mutated locus responsible for the lack of Dao activity (ldao) existed in the LEA genome. Sequence analysis identified deletion breakpoints located in intron 4-5 of the Dao gene and intron 1-2 of the Svop gene, resulting in a 54.1-kb deletion which encompassed exons 5-12 of the Dao gene and exons 2-16 of the Svop gene. We developed a novel congenic rat strain, F344-Daoldao, harboring the Daoldao mutation from LEA rats delivered onto the F344 genetic background. Compared to the parental F344 strain, in F344-Daoldao rats d-Ala was markedly increased in both cerebrum and cerebellum, while d-Ser content was increased in cerebellum but not cerebrum. d-Ala, d-Ser, d-Pro and d-Leu levels were also elevated in F344-Daoldao plasma. F344-Daoldao rats represent a novel model system that will aid in elucidating the physiological functions of d-amino acids in vivo. (203 words).
Collapse
Affiliation(s)
- Yukiko Shimizu
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjuku, Tokyo 162-8655, Japan; Department of Pediatrics, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo 113-8421, Japan
| | - Chiharu Ishii
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi, Fukuoka 812-8582, Japan
| | - Rieko Yanobu-Takanashi
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjuku, Tokyo 162-8655, Japan
| | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjuku, Tokyo 162-8655, Japan
| | - Akio Imaike
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjuku, Tokyo 162-8655, Japan; Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo 113-8421, Japan
| | - Masashi Mita
- KAGAMI INC., 7-7-15 Saito-asagi, Ibaraki, Osaka 567-0085, Japan
| | - Kenji Hamase
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi, Fukuoka 812-8582, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjuku, Tokyo 162-8655, Japan; Section of Animal Models, Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine (NCGM), 1-21-1 Toyama, Shinjuku, Tokyo 162-8655, Japan.
| |
Collapse
|
96
|
Pham J, Keon M, Brennan S, Saksena N. Connecting RNA-Modifying Similarities of TDP-43, FUS, and SOD1 with MicroRNA Dysregulation Amidst A Renewed Network Perspective of Amyotrophic Lateral Sclerosis Proteinopathy. Int J Mol Sci 2020; 21:ijms21103464. [PMID: 32422969 PMCID: PMC7278980 DOI: 10.3390/ijms21103464] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
Beyond traditional approaches in understanding amyotrophic lateral sclerosis (ALS), multiple recent studies in RNA-binding proteins (RBPs)-including transactive response DNA-binding protein (TDP-43) and fused in sarcoma (FUS)-have instigated an interest in their function and prion-like properties. Given their prominence as hallmarks of a highly heterogeneous disease, this prompts a re-examination of the specific functional interrelationships between these proteins, especially as pathological SOD1-a non-RBP commonly associated with familial ALS (fALS)-exhibits similar properties to these RBPs including potential RNA-regulatory capabilities. Moreover, the cytoplasmic mislocalization, aggregation, and co-aggregation of TDP-43, FUS, and SOD1 can be identified as proteinopathies akin to other neurodegenerative diseases (NDs), eliciting strong ties to disrupted RNA splicing, transport, and stability. In recent years, microRNAs (miRNAs) have also been increasingly implicated in the disease, and are of greater significance as they are the master regulators of RNA metabolism in disease pathology. However, little is known about the role of these proteins and how they are regulated by miRNA, which would provide mechanistic insights into ALS pathogenesis. This review seeks to discuss current developments across TDP-43, FUS, and SOD1 to build a detailed snapshot of the network pathophysiology underlying ALS while aiming to highlight possible novel therapeutic targets to guide future research.
Collapse
Affiliation(s)
- Jade Pham
- Faculty of Medicine, The University of New South Wales, Kensington, Sydney, NSW 2033, Australia;
| | - Matt Keon
- Iggy Get Out, Neurodegenerative Disease Section, Darlinghurst, Sydney, NSW 2010, Australia; (M.K.); (S.B.)
| | - Samuel Brennan
- Iggy Get Out, Neurodegenerative Disease Section, Darlinghurst, Sydney, NSW 2010, Australia; (M.K.); (S.B.)
| | - Nitin Saksena
- Iggy Get Out, Neurodegenerative Disease Section, Darlinghurst, Sydney, NSW 2010, Australia; (M.K.); (S.B.)
- Correspondence:
| |
Collapse
|
97
|
Sanna S, Esposito S, Masala A, Sini P, Nieddu G, Galioto M, Fais M, Iaccarino C, Cestra G, Crosio C. HDAC1 inhibition ameliorates TDP-43-induced cell death in vitro and in vivo. Cell Death Dis 2020; 11:369. [PMID: 32409664 PMCID: PMC7224392 DOI: 10.1038/s41419-020-2580-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
TDP-43 pathology is a disease hallmark that characterizes both amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD-TDP). TDP-43 undergoes several posttranslational modifications that can change its biological activities and its aggregative propensity, which is a common hallmark of different neurodegenerative conditions. New evidence is provided by the current study pointing at TDP-43 acetylation in ALS cellular models. Using both in vitro and in vivo approaches, we demonstrate that TDP-43 interacts with histone deacetylase 1 (HDAC1) via RRM1 and RRM2 domains, that are known to contain the two major TDP-43 acetylation sites, K142 and K192. Moreover, we show that TDP-43 is a direct transcriptional activator of CHOP promoter and this activity is regulated by acetylation. Finally and most importantly, we observe both in cell culture and in Drosophila that a HDCA1 reduced level (genomic inactivation or siRNA) or treatment with pan-HDAC inhibitors exert a protective role against WT or pathological mutant TDP-43 toxicity, suggesting TDP-43 acetylation as a new potential therapeutic target. HDAC inhibition efficacy in neurodegeneration has long been debated, but future investigations are warranted in this area. Selection of more specific HDAC inhibitors is still a promising option for neuronal protection especially as HDAC1 appears as a downstream target of both TDP- 43 and FUS, another ALS-related gene.
Collapse
Affiliation(s)
- Simona Sanna
- Department of Biomedical Sciences, University of Sassari, Via Muroni 25, I-07100, Sassari, Italy
| | - Sonia Esposito
- Department of Biomedical Sciences, University of Sassari, Via Muroni 25, I-07100, Sassari, Italy
| | - Alessandra Masala
- Department of Biomedical Sciences, University of Sassari, Via Muroni 25, I-07100, Sassari, Italy
| | - Paola Sini
- Department of Biomedical Sciences, University of Sassari, Via Muroni 25, I-07100, Sassari, Italy
| | - Gabriele Nieddu
- Department of Biomedical Sciences, University of Sassari, Via Muroni 25, I-07100, Sassari, Italy
| | - Manuela Galioto
- Department of Biomedical Sciences, University of Sassari, Via Muroni 25, I-07100, Sassari, Italy
| | - Milena Fais
- Department of Biomedical Sciences, University of Sassari, Via Muroni 25, I-07100, Sassari, Italy
| | - Ciro Iaccarino
- Department of Biomedical Sciences, University of Sassari, Via Muroni 25, I-07100, Sassari, Italy
| | - Gianluca Cestra
- Istitute of Molecular Biology and Pathology-National Research Council at Department of Biology and Biotechnology-Charles Darwin, Sapienza University of Rome, P.Le A.Moro 5, I-00185, Rome, Italy
| | - Claudia Crosio
- Department of Biomedical Sciences, University of Sassari, Via Muroni 25, I-07100, Sassari, Italy.
| |
Collapse
|
98
|
Olesen MN, Wuolikainen A, Nilsson AC, Wirenfeldt M, Forsberg K, Madsen JS, Lillevang ST, Brandslund I, Andersen PM, Asgari N. Inflammatory profiles relate to survival in subtypes of amyotrophic lateral sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:e697. [PMID: 32123048 PMCID: PMC7136052 DOI: 10.1212/nxi.0000000000000697] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate inflammatory cytokines in patients with motor neuron disease (MND) evaluating the putative contribution of amyotrophic lateral sclerosis (ALS)-causing gene variants. METHODS This study is a retrospective case series with prospective follow-up (1994-2016) of 248 patients with MND, of whom 164 had ALS who were screened for mutations in the genes for SOD1 and C9orf72. Paired CSF and plasma were collected at the diagnostic evaluation before treatment. A panel of cytokines were measured blindly via digital ELISA on the Simoa platform. RESULTS Time from disease onset to death was longer for patients with ALS-causing SOD1 mutations (mSOD1, n = 24) than those with C9orf72 hexanucleotide repeat expansion (C9orf72HRE) ALS (n = 19; q = 0.001) and other ALS (OALS) (n = 119; q = 0.0008). Patients with OALS had higher CSF tumor necrosis factor alpha (TNF-α) compared with those with C9orf72HRE ALS (q = 0.014). Patients with C9orf72HRE ALS had higher CSF interferon alpha compared with those with OALS and mSOD1 ALS (q = 0.042 and q = 0.042). In patients with ALS, the survival was negatively correlated with plasma interleukin (IL) 10 (hazard ratio [HR] 1.17, 95% CI 1.05-1.30). Plasma TNF-α, IL-10, and TNF-related apoptosis-inducing ligand (TRAIL) (HR 1.01 [1.00-1.02], 1.15 [1.02-1.30], and 1.01 [1.00-1.01], respectively) of patients with OALS, plasma IL-1β (HR 5.90 [1.27-27.5]) of patients with C9orf72HRE ALS, and CSF TRAIL (10.5 [1.12-98.6]) of patients with mSOD1 ALS all correlated negatively with survival. CONCLUSIONS Differences in survival times in ALS subtypes were correlated with cytokine levels, suggesting specific immune responses related to ALS genetic variants.
Collapse
Affiliation(s)
- Mads Nikolaj Olesen
- From the Institutes of Regional Health Research and Molecular Medicine (M.N.O., N.A.), University of Southern Denmark; Departments of Neurology (M.N.O.), Slagelse Hospital & Biochemistry & Immunology, Lillebaelt Hospital, Vejle, Denmark; Department of Pharmacology and Clinical Neuroscience (A.W., K.F., P.M.A.), Umeå University, Sweden; Department of Clinical Immunology (A.C.N., S.T.L.), Odense University Hospital, Denmark; Department of Pathology (M.W.), Odense University Hospital, Denmark; Biochemistry & Immunology (J.S.M., I.B.), Lillebaelt Hospital, Vejle, Denmark; Institute of Regional Health Research (J.S.M., I.B.), University of Southern Denmark, Odense; Department of Neurology (N.A.), Slagelse Hospital; and OPEN, Odense Patient Data Explorative Network (N.A.), Odense University Hospital, Denmark
| | - Anna Wuolikainen
- From the Institutes of Regional Health Research and Molecular Medicine (M.N.O., N.A.), University of Southern Denmark; Departments of Neurology (M.N.O.), Slagelse Hospital & Biochemistry & Immunology, Lillebaelt Hospital, Vejle, Denmark; Department of Pharmacology and Clinical Neuroscience (A.W., K.F., P.M.A.), Umeå University, Sweden; Department of Clinical Immunology (A.C.N., S.T.L.), Odense University Hospital, Denmark; Department of Pathology (M.W.), Odense University Hospital, Denmark; Biochemistry & Immunology (J.S.M., I.B.), Lillebaelt Hospital, Vejle, Denmark; Institute of Regional Health Research (J.S.M., I.B.), University of Southern Denmark, Odense; Department of Neurology (N.A.), Slagelse Hospital; and OPEN, Odense Patient Data Explorative Network (N.A.), Odense University Hospital, Denmark
| | - Anna Christine Nilsson
- From the Institutes of Regional Health Research and Molecular Medicine (M.N.O., N.A.), University of Southern Denmark; Departments of Neurology (M.N.O.), Slagelse Hospital & Biochemistry & Immunology, Lillebaelt Hospital, Vejle, Denmark; Department of Pharmacology and Clinical Neuroscience (A.W., K.F., P.M.A.), Umeå University, Sweden; Department of Clinical Immunology (A.C.N., S.T.L.), Odense University Hospital, Denmark; Department of Pathology (M.W.), Odense University Hospital, Denmark; Biochemistry & Immunology (J.S.M., I.B.), Lillebaelt Hospital, Vejle, Denmark; Institute of Regional Health Research (J.S.M., I.B.), University of Southern Denmark, Odense; Department of Neurology (N.A.), Slagelse Hospital; and OPEN, Odense Patient Data Explorative Network (N.A.), Odense University Hospital, Denmark
| | - Martin Wirenfeldt
- From the Institutes of Regional Health Research and Molecular Medicine (M.N.O., N.A.), University of Southern Denmark; Departments of Neurology (M.N.O.), Slagelse Hospital & Biochemistry & Immunology, Lillebaelt Hospital, Vejle, Denmark; Department of Pharmacology and Clinical Neuroscience (A.W., K.F., P.M.A.), Umeå University, Sweden; Department of Clinical Immunology (A.C.N., S.T.L.), Odense University Hospital, Denmark; Department of Pathology (M.W.), Odense University Hospital, Denmark; Biochemistry & Immunology (J.S.M., I.B.), Lillebaelt Hospital, Vejle, Denmark; Institute of Regional Health Research (J.S.M., I.B.), University of Southern Denmark, Odense; Department of Neurology (N.A.), Slagelse Hospital; and OPEN, Odense Patient Data Explorative Network (N.A.), Odense University Hospital, Denmark
| | - Karin Forsberg
- From the Institutes of Regional Health Research and Molecular Medicine (M.N.O., N.A.), University of Southern Denmark; Departments of Neurology (M.N.O.), Slagelse Hospital & Biochemistry & Immunology, Lillebaelt Hospital, Vejle, Denmark; Department of Pharmacology and Clinical Neuroscience (A.W., K.F., P.M.A.), Umeå University, Sweden; Department of Clinical Immunology (A.C.N., S.T.L.), Odense University Hospital, Denmark; Department of Pathology (M.W.), Odense University Hospital, Denmark; Biochemistry & Immunology (J.S.M., I.B.), Lillebaelt Hospital, Vejle, Denmark; Institute of Regional Health Research (J.S.M., I.B.), University of Southern Denmark, Odense; Department of Neurology (N.A.), Slagelse Hospital; and OPEN, Odense Patient Data Explorative Network (N.A.), Odense University Hospital, Denmark
| | - Jonna Skov Madsen
- From the Institutes of Regional Health Research and Molecular Medicine (M.N.O., N.A.), University of Southern Denmark; Departments of Neurology (M.N.O.), Slagelse Hospital & Biochemistry & Immunology, Lillebaelt Hospital, Vejle, Denmark; Department of Pharmacology and Clinical Neuroscience (A.W., K.F., P.M.A.), Umeå University, Sweden; Department of Clinical Immunology (A.C.N., S.T.L.), Odense University Hospital, Denmark; Department of Pathology (M.W.), Odense University Hospital, Denmark; Biochemistry & Immunology (J.S.M., I.B.), Lillebaelt Hospital, Vejle, Denmark; Institute of Regional Health Research (J.S.M., I.B.), University of Southern Denmark, Odense; Department of Neurology (N.A.), Slagelse Hospital; and OPEN, Odense Patient Data Explorative Network (N.A.), Odense University Hospital, Denmark
| | - Soeren Thue Lillevang
- From the Institutes of Regional Health Research and Molecular Medicine (M.N.O., N.A.), University of Southern Denmark; Departments of Neurology (M.N.O.), Slagelse Hospital & Biochemistry & Immunology, Lillebaelt Hospital, Vejle, Denmark; Department of Pharmacology and Clinical Neuroscience (A.W., K.F., P.M.A.), Umeå University, Sweden; Department of Clinical Immunology (A.C.N., S.T.L.), Odense University Hospital, Denmark; Department of Pathology (M.W.), Odense University Hospital, Denmark; Biochemistry & Immunology (J.S.M., I.B.), Lillebaelt Hospital, Vejle, Denmark; Institute of Regional Health Research (J.S.M., I.B.), University of Southern Denmark, Odense; Department of Neurology (N.A.), Slagelse Hospital; and OPEN, Odense Patient Data Explorative Network (N.A.), Odense University Hospital, Denmark
| | - Ivan Brandslund
- From the Institutes of Regional Health Research and Molecular Medicine (M.N.O., N.A.), University of Southern Denmark; Departments of Neurology (M.N.O.), Slagelse Hospital & Biochemistry & Immunology, Lillebaelt Hospital, Vejle, Denmark; Department of Pharmacology and Clinical Neuroscience (A.W., K.F., P.M.A.), Umeå University, Sweden; Department of Clinical Immunology (A.C.N., S.T.L.), Odense University Hospital, Denmark; Department of Pathology (M.W.), Odense University Hospital, Denmark; Biochemistry & Immunology (J.S.M., I.B.), Lillebaelt Hospital, Vejle, Denmark; Institute of Regional Health Research (J.S.M., I.B.), University of Southern Denmark, Odense; Department of Neurology (N.A.), Slagelse Hospital; and OPEN, Odense Patient Data Explorative Network (N.A.), Odense University Hospital, Denmark
| | - Peter Munch Andersen
- From the Institutes of Regional Health Research and Molecular Medicine (M.N.O., N.A.), University of Southern Denmark; Departments of Neurology (M.N.O.), Slagelse Hospital & Biochemistry & Immunology, Lillebaelt Hospital, Vejle, Denmark; Department of Pharmacology and Clinical Neuroscience (A.W., K.F., P.M.A.), Umeå University, Sweden; Department of Clinical Immunology (A.C.N., S.T.L.), Odense University Hospital, Denmark; Department of Pathology (M.W.), Odense University Hospital, Denmark; Biochemistry & Immunology (J.S.M., I.B.), Lillebaelt Hospital, Vejle, Denmark; Institute of Regional Health Research (J.S.M., I.B.), University of Southern Denmark, Odense; Department of Neurology (N.A.), Slagelse Hospital; and OPEN, Odense Patient Data Explorative Network (N.A.), Odense University Hospital, Denmark
| | - Nasrin Asgari
- From the Institutes of Regional Health Research and Molecular Medicine (M.N.O., N.A.), University of Southern Denmark; Departments of Neurology (M.N.O.), Slagelse Hospital & Biochemistry & Immunology, Lillebaelt Hospital, Vejle, Denmark; Department of Pharmacology and Clinical Neuroscience (A.W., K.F., P.M.A.), Umeå University, Sweden; Department of Clinical Immunology (A.C.N., S.T.L.), Odense University Hospital, Denmark; Department of Pathology (M.W.), Odense University Hospital, Denmark; Biochemistry & Immunology (J.S.M., I.B.), Lillebaelt Hospital, Vejle, Denmark; Institute of Regional Health Research (J.S.M., I.B.), University of Southern Denmark, Odense; Department of Neurology (N.A.), Slagelse Hospital; and OPEN, Odense Patient Data Explorative Network (N.A.), Odense University Hospital, Denmark.
| |
Collapse
|
99
|
Liu W, Venugopal S, Majid S, Ahn IS, Diamante G, Hong J, Yang X, Chandler SH. Single-cell RNA-seq analysis of the brainstem of mutant SOD1 mice reveals perturbed cell types and pathways of amyotrophic lateral sclerosis. Neurobiol Dis 2020; 141:104877. [PMID: 32360664 PMCID: PMC7519882 DOI: 10.1016/j.nbd.2020.104877] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/13/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease in which motor neurons throughout the brain and spinal cord progressively degenerate resulting in muscle atrophy, paralysis and death. Recent studies using animal models of ALS implicate multiple cell-types (e.g., astrocytes and microglia) in ALS pathogenesis in the spinal motor systems. To ascertain cellular vulnerability and cell-type specific mechanisms of ALS in the brainstem that orchestrates oral-motor functions, we conducted parallel single cell RNA sequencing (scRNA-seq) analysis using the high-throughput Drop-seq method. We isolated 1894 and 3199 cells from the brainstem of wildtype and mutant SOD1 symptomatic mice respectively, at postnatal day 100. We recovered major known cell types and neuronal subpopulations, such as interneurons and motor neurons, and trigeminal ganglion (TG) peripheral sensory neurons, as well as, previously uncharacterized interneuron subtypes. We found that the majority of the cell types displayed transcriptomic alterations in ALS mice. Differentially expressed genes (DEGs) of individual cell populations revealed cell-type specific alterations in numerous pathways, including previously known ALS pathways such as inflammation (in microglia), stress response (ependymal and an uncharacterized cell population), neurogenesis (astrocytes, oligodendrocytes, neurons), synapse organization and transmission (microglia, oligodendrocyte precursor cells, and neuronal subtypes), and mitochondrial function (uncharacterized cell populations). Other cell-type specific processes altered in SOD1 mutant brainstem include those from motor neurons (axon regeneration, voltage-gated sodium and potassium channels underlying excitability, potassium ion transport), trigeminal sensory neurons (detection of temperature stimulus involved in sensory perception), and cellular response to toxic substances (uncharacterized cell populations). DEGs consistently altered across cell types (e.g., Malat1), as well as cell-type specific DEGs, were identified. Importantly, DEGs from various cell types overlapped with known ALS genes from the literature and with top hits from an existing human ALS genome-wide association study (GWAS), implicating the potential cell types in which the ALS genes function with ALS pathogenesis. Our molecular investigation at single cell resolution provides comprehensive insights into the cell types, genes and pathways altered in the brainstem in a widely used ALS mouse model.
Collapse
Affiliation(s)
- Wenting Liu
- Department of Integrative Biology & Physiology, University of California, 2024 Terasaki Bld, 610 Charles E. Young Dr. East, Los Angeles, USA
| | - Sharmila Venugopal
- Department of Integrative Biology & Physiology, University of California, 2024 Terasaki Bld, 610 Charles E. Young Dr. East, Los Angeles, USA
| | - Sana Majid
- Department of Integrative Biology & Physiology, University of California, 2024 Terasaki Bld, 610 Charles E. Young Dr. East, Los Angeles, USA
| | - In Sook Ahn
- Department of Integrative Biology & Physiology, University of California, 2024 Terasaki Bld, 610 Charles E. Young Dr. East, Los Angeles, USA
| | - Graciel Diamante
- Department of Integrative Biology & Physiology, University of California, 2024 Terasaki Bld, 610 Charles E. Young Dr. East, Los Angeles, USA
| | - Jason Hong
- Department of Integrative Biology & Physiology, University of California, 2024 Terasaki Bld, 610 Charles E. Young Dr. East, Los Angeles, USA
| | - Xia Yang
- Department of Integrative Biology & Physiology, University of California, 2024 Terasaki Bld, 610 Charles E. Young Dr. East, Los Angeles, USA; Brain Research Institute, University of California, Los Angeles, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, USA.
| | - Scott H Chandler
- Department of Integrative Biology & Physiology, University of California, 2024 Terasaki Bld, 610 Charles E. Young Dr. East, Los Angeles, USA; Brain Research Institute, University of California, Los Angeles, USA.
| |
Collapse
|
100
|
Pandya VA, Patani R. Decoding the relationship between ageing and amyotrophic lateral sclerosis: a cellular perspective. Brain 2020; 143:1057-1072. [PMID: 31851317 PMCID: PMC7174045 DOI: 10.1093/brain/awz360] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/13/2019] [Accepted: 09/21/2019] [Indexed: 12/13/2022] Open
Abstract
With an ageing population comes an inevitable increase in the prevalence of age-associated neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), a relentlessly progressive and universally fatal disease characterized by the degeneration of upper and lower motor neurons within the brain and spinal cord. Indeed, the physiological process of ageing causes a variety of molecular and cellular phenotypes. With dysfunction at the neuromuscular junction implicated as a key pathological mechanism in ALS, and each lower motor unit cell type vulnerable to its own set of age-related phenotypes, the effects of ageing might in fact prove a prerequisite to ALS, rendering the cells susceptible to disease-specific mechanisms. Moreover, we discuss evidence for overlap between age and ALS-associated hallmarks, potentially implicating cell type-specific ageing as a key contributor to this multifactorial and complex disease. With a dearth of disease-modifying therapy currently available for ALS patients and a substantial failure in bench to bedside translation of other potential therapies, the unification of research in ageing and ALS requires high fidelity models to better recapitulate age-related human disease and will ultimately yield more reliable candidate therapeutics for patients, with the aim of enhancing healthspan and life expectancy.
Collapse
Affiliation(s)
- Virenkumar A Pandya
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, UK
- The Francis Crick Institute, London, UK
| | - Rickie Patani
- Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|