1
|
Kim A, Lee DY, Sung JJ. Cdk5 inhibition in the SOD1 G93A transgenic mouse model of amyotrophic lateral sclerosis suppresses neurodegeneration and extends survival. J Neurochem 2024; 168:2908-2925. [PMID: 38934222 DOI: 10.1111/jnc.16160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Deregulated cyclin-dependent kinase 5 (Cdk5) activity closely correlates with hyperphosphorylated tau, a common pathology found in neurodegenerative diseases. Previous postmortem studies had revealed increased Cdk5 immunoreactivity in amyotrophic lateral sclerosis (ALS); hence, we investigated the effects of Cdk5 inhibition on ALS model mice and neurons in this study. For the in vitro study, motor neuron cell lines with wild-type superoxide dismutase 1 (SOD1) or SOD1G93A and primary neuronal cultures from SOD1G93A transgenic (TG) mice or non-TG mice were compared for the expression of proteins involved in tau pathology, neuroinflammation, apoptosis, and neuritic outgrowth by applying Cdk5-small interfering RNA or Cdk5-short hairpin RNA (shRNA). For the in vivo study, SOD1G93A mice and non-TG mice were intrathecally injected with adeno-associated virus 9 (AAV9)-scramble (SCR)-shRNA or AAV9-Cdk5-shRNA at the age of 5 weeks. Weight and motor function were measured three times per week from 60 days of age, longevity was evaluated, and the tissues were collected from 90-day-old or 120-day-old mice. Neurons with SOD1G93A showed increased phosphorylated tau, attenuated neuritic growth, mislocalization of SOD1, and enhanced neuroinflammation and apoptosis, all of which were reversed by Cdk5 inhibition. Weights did not show significant differences among non-TG and SOD1G93A mice with or without Cdk5 silencing. SOD1G93A mice treated with AAV9-Cdk5-shRNA showed significantly delayed disease onset, delayed rotarod failure, and prolonged survival compared with those treated with AAV9-SCR-shRNA. The brain and spinal cord of SOD1G93A mice intrathecally injected with AAV9-Cdk5-shRNA exhibited suppressed tau pathology, neuroinflammation, apoptosis, and an increased number of motor neurons compared to those of SOD1G93A mice injected with AAV9-SCR-shRNA. Cdk5 inhibition could be an important mechanism in the development of a new therapeutic strategy for ALS.
Collapse
Affiliation(s)
- Ahwon Kim
- Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Neurology, Chungbuk National University Hospital, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Do-Yeon Lee
- Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Neurology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung-Joon Sung
- Department of Neurology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Neurology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Abyadeh M, Gupta V, Paulo JA, Mahmoudabad AG, Shadfar S, Mirshahvaladi S, Gupta V, Nguyen CT, Finkelstein DI, You Y, Haynes PA, Salekdeh GH, Graham SL, Mirzaei M. Amyloid-beta and tau protein beyond Alzheimer's disease. Neural Regen Res 2024; 19:1262-1276. [PMID: 37905874 PMCID: PMC11467936 DOI: 10.4103/1673-5374.386406] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT The aggregation of amyloid-beta peptide and tau protein dysregulation are implicated to play key roles in Alzheimer's disease pathogenesis and are considered the main pathological hallmarks of this devastating disease. Physiologically, these two proteins are produced and expressed within the normal human body. However, under pathological conditions, abnormal expression, post-translational modifications, conformational changes, and truncation can make these proteins prone to aggregation, triggering specific disease-related cascades. Recent studies have indicated associations between aberrant behavior of amyloid-beta and tau proteins and various neurological diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as retinal neurodegenerative diseases like Glaucoma and age-related macular degeneration. Additionally, these proteins have been linked to cardiovascular disease, cancer, traumatic brain injury, and diabetes, which are all leading causes of morbidity and mortality. In this comprehensive review, we provide an overview of the connections between amyloid-beta and tau proteins and a spectrum of disorders.
Collapse
Affiliation(s)
| | - Vivek Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Sina Shadfar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Shahab Mirshahvaladi
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Christine T.O. Nguyen
- Department of Optometry and Vision Sciences, School of Health Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Yuyi You
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Paul A. Haynes
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Ghasem H. Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Stuart L. Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| |
Collapse
|
3
|
Tsuji K, Nakayama Y, Taruya J, Ito H. Persistence of Kii amyotrophic lateral sclerosis after the 2000s and its characteristic aging-related tau astrogliopathy. J Neuropathol Exp Neurol 2024; 83:79-93. [PMID: 38193356 DOI: 10.1093/jnen/nlad113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024] Open
Abstract
Kii amyotrophic lateral sclerosis (ALS) is a unique disease that occurs in the southern portion of the Kii Peninsula and exhibits a dual pathology of TAR DNA-binding protein of 43 kDa (TDP-43) proteinopathy and tauopathy. The incidence of ALS in this region was very high in the 1960s, briefly decreased through the 1980s, but began increasing again after 2000 with a change of high-concentration geographic foci. It is unclear, however, whether the unique pathological features have changed along with the incidence changes. This study analyzed postmortem specimens from neuropathologically confirmed Kii ALS cases from the 1970s (n = 4) and those after 1999 (n = 12) from the southern Kii Peninsula or outside of the area. Our results confirm the continued occurrence of Kii ALS after 2000 in the southern Kii Peninsula and the preservation of disease-specific neuronal tau pathology, including the widespread occurrence throughout the brain and spinal cord, sparse neuropil threads, and predominance in superficial layers. Furthermore, we assessed the glial tau pathology of Kii and non-Kii ALS in accordance with the aging-related tau astrogliopathy classification method for the first time and detected a unique brainstem predominant appearance of gray matter aging-related tau astrogliopathy in Kii ALS cases, which may provide clues to pathogenetic mechanisms.
Collapse
Affiliation(s)
- Kazumi Tsuji
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Yoshiaki Nakayama
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Junko Taruya
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Hidefumi Ito
- Department of Neurology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
4
|
Donison N, Hintermayer M, Subramaniam M, Santandrea E, Volkening K, Strong MJ. Upregulation of LRRK2 following traumatic brain injury does not directly phosphorylate Thr 175 tau. Front Cell Neurosci 2023; 17:1272899. [PMID: 38026695 PMCID: PMC10663351 DOI: 10.3389/fncel.2023.1272899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Phosphorylated microtubule-associated protein tau (tau) aggregates are a pathological hallmark of various neurodegenerative diseases, including chronic traumatic encephalopathy and amyotrophic lateral sclerosis with cognitive impairment. While there are many residues phosphorylated on tau, phosphorylation of threonine 175 (pThr175 tau) has been shown to initiate fibril formation in vitro and is present in pathological tau aggregates in vivo. Given this, preventing Thr175 tau phosphorylation presents a potential approach to reduce fibril formation; however, the kinase(s) acting on Thr175 are not yet fully defined. Using a single controlled cortical impact rodent model of traumatic brain injury (TBI), which rapidly induces Thr175 tau phosphorylation, we observed an upregulation and alteration in subcellular localization of leucine-rich repeat kinase 2 (LRRK2), a kinase that has been implicated in tau phosphorylation. LRRK2 upregulation was evident by one-day post-injury and persisted to day 10. The most notable changes were observed in microglia at the site of injury in the cortex. To determine if the appearance of pThr175 tau was causally related to the upregulation of LRRK2 expression, we examined the ability of LRRK2 to phosphorylate Thr175in vitro by co-transfecting 2N4R human WT-tau with either LRRK2-WT, constitutively-active LRRK2-G2019S or inactive LRRK2-3XKD. We found no significant difference in the level of pThr175 tau between the overexpression of LRRK2-WT, -G2019S or -3XKD, suggesting LRRK2 does not phosphorylate tau at Thr175. Further, downstream events known to follow Thr175 phosphorylation and known to be associated with pathological tau fibril formation (pSer9-GSK3β and pThr231 tau induction) also remained unchanged. We conclude that while LRRK2 expression is altered in TBI, it does not contribute directly to pThr175 tau generation.
Collapse
Affiliation(s)
- Neil Donison
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Matthew Hintermayer
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Maegha Subramaniam
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Erin Santandrea
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Kathryn Volkening
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Michael J. Strong
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
5
|
Jellinger KA. The Spectrum of Cognitive Dysfunction in Amyotrophic Lateral Sclerosis: An Update. Int J Mol Sci 2023; 24:14647. [PMID: 37834094 PMCID: PMC10572320 DOI: 10.3390/ijms241914647] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Cognitive dysfunction is an important non-motor symptom in amyotrophic lateral sclerosis (ALS) that has a negative impact on survival and caregiver burden. It shows a wide spectrum ranging from subjective cognitive decline to frontotemporal dementia (FTD) and covers various cognitive domains, mainly executive/attention, language and verbal memory deficits. The frequency of cognitive impairment across the different ALS phenotypes ranges from 30% to 75%, with up to 45% fulfilling the criteria of FTD. Significant genetic, clinical, and pathological heterogeneity reflects deficits in various cognitive domains. Modern neuroimaging studies revealed frontotemporal degeneration and widespread involvement of limbic and white matter systems, with hypometabolism of the relevant areas. Morphological substrates are frontotemporal and hippocampal atrophy with synaptic loss, associated with TDP-43 and other co-pathologies, including tau deposition. Widespread functional disruptions of motor and extramotor networks, as well as of frontoparietal, frontostriatal and other connectivities, are markers for cognitive deficits in ALS. Cognitive reserve may moderate the effect of brain damage but is not protective against cognitive decline. The natural history of cognitive dysfunction in ALS and its relationship to FTD are not fully understood, although there is an overlap between the ALS variants and ALS-related frontotemporal syndromes, suggesting a differential vulnerability of motor and non-motor networks. An assessment of risks or the early detection of brain connectivity signatures before structural changes may be helpful in investigating the pathophysiological mechanisms of cognitive impairment in ALS, which might even serve as novel targets for effective disease-modifying therapies.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, A-1150 Vienna, Austria
| |
Collapse
|
6
|
Mastrangelo A, Vacchiano V, Zenesini C, Ruggeri E, Baiardi S, Cherici A, Avoni P, Polischi B, Santoro F, Capellari S, Liguori R, Parchi P. Amyloid-Beta Co-Pathology Is a Major Determinant of the Elevated Plasma GFAP Values in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:13976. [PMID: 37762278 PMCID: PMC10531493 DOI: 10.3390/ijms241813976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/29/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Recent studies reported increased plasma glial acidic fibrillary protein (GFAP) levels in amyotrophic lateral sclerosis (ALS) patients compared to controls. We expanded these findings in a larger cohort, including 156 ALS patients and 48 controls, and investigated the associations of plasma GFAP with clinical variables and other biofluid biomarkers. Plasma GFAP and Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarkers were assessed by the single molecule array and the Lumipulse platforms, respectively. In ALS patients, plasma GFAP was higher than in controls (p < 0.001) and associated with measures of cognitive decline. Twenty ALS patients (12.8%) showed a positive amyloid status (A+), of which nine also exhibited tau pathology (A+T+, namely ALS-AD). ALS-AD patients showed higher plasma GFAP than A- ALS participants (p < 0.001) and controls (p < 0.001), whereas the comparison between A- ALS and controls missed statistical significance (p = 0.07). Plasma GFAP distinguished ALS-AD subjects more accurately (area under the curve (AUC) 0.932 ± 0.027) than plasma p-tau181 (AUC 0.692 ± 0.058, p < 0.0001) and plasma neurofilament light chain protein (AUC, 0.548 ± 0.088, p < 0.0001). Cognitive measures differed between ALS-AD and other ALS patients. AD co-pathology deeply affects plasma GFAP values in ALS patients. Plasma GFAP is an accurate biomarker for identifying AD co-pathology in ALS, which can influence the cognitive phenotype.
Collapse
Affiliation(s)
- Andrea Mastrangelo
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
| | - Veria Vacchiano
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Corrado Zenesini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Edoardo Ruggeri
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Simone Baiardi
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
| | - Arianna Cherici
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Patrizia Avoni
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Barbara Polischi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Francesca Santoro
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Sabina Capellari
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Rocco Liguori
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| | - Piero Parchi
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, 40139 Bologna, Italy; (A.M.); (S.B.); (P.A.); (S.C.); (R.L.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy; (V.V.); (C.Z.); (E.R.); (A.C.); (B.P.); (F.S.)
| |
Collapse
|
7
|
Vacchiano V, Mastrangelo A, Zenesini C, Baiardi S, Avoni P, Polischi B, Capellari S, Salvi F, Liguori R, Parchi P. Elevated plasma p-tau181 levels unrelated to Alzheimer's disease pathology in amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2023; 94:428-435. [PMID: 37012065 DOI: 10.1136/jnnp-2022-330709] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/15/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Phosphorylated-tau181 (p-tau181), a specific marker of Alzheimer's disease (AD) pathology, was found elevated in plasma but not in cerebrospinal fluid (CSF) of patients with amyotrophic lateral sclerosis (ALS). We expanded these findings in a larger patient cohort, exploring clinical/electrophysiological associations, prognostic value and longitudinal trajectories of the biomarker. METHODS We obtained baseline plasma samples from 148 ALS, 12 spinal muscular atrophy (SMA), and 88 AD patients, and 60 healthy controls. Baseline CSF and longitudinal plasma samples were from 130 and 39 patients with ALS. CSF AD markers were measured with the Lumipulse platform, and plasma p-tau181 with SiMoA. RESULTS Patients with ALS showed higher plasma p-tau181 levels than controls (p<0.001) and lower than AD participants (p=0.02). SMA patients had higher levels than controls (p=0.03). In patients with ALS, CSF p-tau and plasma p-tau181 did not correlate (p=0.37). Plasma p-tau181 significantly increased with the number of regions showing clinical/neurophysiological lower motor neurons (LMN) signs (p=0.007) and correlated with the degree of denervation in the lumbosacral area (r=0.51, p<0.0001). Plasma p-tau181 levels were higher in classic and LMN-predominant than in bulbar phenotype (p=0.004 and p=0.006). Multivariate Cox regression confirmed plasma p-tau181 as an independent prognostic factor in ALS (HR 1.90, 95% CI 1.25 to 2.90, p=0.003). Longitudinal analysis showed a significant rise in plasma p-tau181 values over time, especially in fast progressors. CONCLUSIONS Plasma p-tau181 is elevated in patients with ALS, independently from CSF levels, and is firmly associated with LMN dysfunction. The finding indicates that p-tau181 of putative peripheral origin might represent a confounding factor in using plasma p-tau181 for AD pathology screening, which deserves further investigation.
Collapse
Affiliation(s)
- Veria Vacchiano
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Andrea Mastrangelo
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Corrado Zenesini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Simone Baiardi
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Patrizia Avoni
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Barbara Polischi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Sabina Capellari
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Fabrizio Salvi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Rocco Liguori
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Piero Parchi
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
8
|
Molecular Investigations of Protein Aggregation in the Pathogenesis of Amyotrophic Lateral Sclerosis. Int J Mol Sci 2022; 24:ijms24010704. [PMID: 36614144 PMCID: PMC9820914 DOI: 10.3390/ijms24010704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating progressive neurodegenerative disorder characterized by selective loss of lower and upper motor neurons (MNs) in the brain and spinal cord, resulting in paralysis and eventually death due to respiratory insufficiency. Although the fundamental physiological mechanisms underlying ALS are not completely understood, the key neuropathological hallmarks of ALS pathology are the aggregation and accumulation of ubiquitinated protein inclusions within the cytoplasm of degenerating MNs. Herein, we discuss recent insights into the molecular mechanisms that lead to the accumulation of protein aggregates in ALS. This will contribute to a better understanding of the pathophysiology of the disease and may open novel avenues for the development of therapeutic strategies.
Collapse
|
9
|
Riku Y, Yoshida M, Iwasaki Y, Sobue G, Katsuno M, Ishigaki S. TDP-43 Proteinopathy and Tauopathy: Do They Have Pathomechanistic Links? Int J Mol Sci 2022; 23:ijms232415755. [PMID: 36555399 PMCID: PMC9779029 DOI: 10.3390/ijms232415755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Transactivation response DNA binding protein 43 kDa (TDP-43) and tau are major pathological proteins of neurodegenerative disorders, of which neuronal and glial aggregates are pathological hallmarks. Interestingly, accumulating evidence from neuropathological studies has shown that comorbid TDP-43 pathology is observed in a subset of patients with tauopathies, and vice versa. The concomitant pathology often spreads in a disease-specific manner and has morphological characteristics in each primary disorder. The findings from translational studies have suggested that comorbid TDP-43 or tau pathology has clinical impacts and that the comorbid pathology is not a bystander, but a part of the disease process. Shared genetic risk factors or molecular abnormalities between TDP-43 proteinopathies and tauopathies, and direct interactions between TDP-43 and tau aggregates, have been reported. Further investigations to clarify the pathogenetic factors that are shared by a broad spectrum of neurodegenerative disorders will establish key therapeutic targets.
Collapse
Affiliation(s)
- Yuichi Riku
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute 480-1195, Japan
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 744-8550, Japan
- Correspondence: or
| | - Mari Yoshida
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute 480-1195, Japan
| | - Yasushi Iwasaki
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute 480-1195, Japan
| | - Gen Sobue
- Graduate School of Medicine, Aichi Medical University, Nagakute 480-1195, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 744-8550, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya 744-8550, Japan
| | - Shinsuke Ishigaki
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan
| |
Collapse
|
10
|
Riku Y, Iwasaki Y, Ishigaki S, Akagi A, Hasegawa M, Nishioka K, Li Y, Riku M, Ikeuchi T, Fujioka Y, Miyahara H, Sone J, Hattori N, Yoshida M, Katsuno M, Sobue G. Motor neuron TDP-43 proteinopathy in progressive supranuclear palsy and corticobasal degeneration. Brain 2022; 145:2769-2784. [PMID: 35274674 DOI: 10.1093/brain/awac091] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/29/2022] [Accepted: 02/15/2022] [Indexed: 11/12/2022] Open
Abstract
Transactive response DNA-binding protein 43 kDa (TDP-43) is mislocalized from the nucleus and aggregates within the cytoplasm of affected neurons in amyotrophic lateral sclerosis (ALS) cases. TDP-43 pathology has also been found in brain tissues under non-ALS conditions, suggesting mechanistic links between TDP-43-related ALS (ALS-TDP) and various neurological disorders. This study aimed to assess TDP-43 pathology in the spinal cord motor neurons of tauopathies. We examined 106 spinal cords from consecutively autopsied cases with progressive supranuclear palsy (PSP, n = 26), corticobasal degeneration (CBD, n = 12), globular glial tauopathy (GGT, n = 5), Alzheimer's disease (AD, n = 21), or Pick disease (PiD, n = 6) and neurologically healthy controls (n = 36). Ten of the PSP cases (38%) and seven of the CBD cases (58%) showed mislocalization and cytoplasmic aggregation of TDP-43 in spinal cord motor neurons, which was prominent in the cervical cord. TDP-43-aggregates were found to be skein-like, round-shaped, granular, or dot-like and contained insoluble C-terminal fragments showing blotting pattern of ALS or frontotemporal lobar degeneration (FTLD). The lower motor neurons also showed cystatin-C aggregates, although Bunina bodies were absent in hematoxylin-eosin staining. The spinal cord TDP-43 pathology was often associated with TDP-43 pathology of the primary motor cortex. Positive correlations were shown between the severities of TDP-43 and 4-repeat (4R)-tau aggregates in the cervical cord. TDP-43 and 4R-tau aggregates burdens positively correlated with microglial burden in anterior horn. TDP-43 pathology of spinal cord motor neuron did not develop in an age-dependent manner and was not found in the AD, PiD, GGT, and control groups. Next, we assessed splicing factor proline/glutamine rich (SFPQ) expression in spinal cord motor neurons; SFPQ is a recently-identified regulator of ALS/FTLD pathogenesis, and it is also reported that interaction between SFPQ and fused-in-sarcoma (FUS) regulates splicing of microtubule-associated protein tau exon 10. Immunofluorescent and proximity-ligation assays revealed altered SFPQ/FUS-interactions in the neuronal nuclei of PSP, CBD, and ALS-TDP cases but not in AD, PiD, and GGT cases. Moreover, SFPQ expression was depleted in neurons containing TDP-43 or 4R-tau aggregates of PSP and CBD cases. Our results indicate that PSP and CBD may have properties of systematic motor neuron TDP-43 proteinopathy, suggesting mechanistic links with ALS-TDP. SFPQ dysfunction, arising from altered interaction with FUS, may be a candidate of the common pathway.
Collapse
Affiliation(s)
- Yuichi Riku
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan.,Department of Neurology, Graduate School of Nagoya University, Aichi, Japan
| | - Yasushi Iwasaki
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Shinsuke Ishigaki
- Department of Neurology, Graduate School of Nagoya University, Aichi, Japan
| | - Akio Akagi
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Masato Hasegawa
- Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kenya Nishioka
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yuanzhe Li
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Miho Riku
- Department of Pathology, Aichi Medical University, Aichi, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yusuke Fujioka
- Department of Neurology, Graduate School of Nagoya University, Aichi, Japan
| | - Hiroaki Miyahara
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Jun Sone
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Mari Yoshida
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Masahisa Katsuno
- Department of Neurology, Graduate School of Nagoya University, Aichi, Japan
| | - Gen Sobue
- Department of Neurology, Graduate School of Nagoya University, Aichi, Japan.,Aichi Medical University, Aichi, Japan
| |
Collapse
|
11
|
Strong MJ, Donison NS, Volkening K. Alterations in Tau Metabolism in ALS and ALS-FTSD. Front Neurol 2020; 11:598907. [PMID: 33329356 PMCID: PMC7719764 DOI: 10.3389/fneur.2020.598907] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022] Open
Abstract
There is increasing acceptance that amyotrophic lateral sclerosis (ALS), classically considered a neurodegenerative disease affecting almost exclusively motor neurons, is syndromic with both clinical and biological heterogeneity. This is most evident in its association with a broad range of neuropsychological, behavioral, speech and language deficits [collectively termed ALS frontotemporal spectrum disorder (ALS-FTSD)]. Although the most consistent pathology of ALS and ALS-FTSD is a disturbance in TAR DNA binding protein 43 kDa (TDP-43) metabolism, alterations in microtubule-associated tau protein (tau) metabolism can also be observed in ALS-FTSD, most prominently as pathological phosphorylation at Thr175 (pThr175tau). pThr175 has been shown to promote exposure of the phosphatase activating domain (PAD) in the tau N-terminus with the consequent activation of GSK3β mediated phosphorylation at Thr231 (pThr231tau) leading to pathological oligomer formation. This pathological cascade of tau phosphorylation has been observed in chronic traumatic encephalopathy with ALS (CTE-ALS) and in both in vivo and in vitro experimental paradigms, suggesting that it is of critical relevance to the pathobiology of ALS-FTSD. It is also evident that the co-existence of alterations in the metabolism of TDP-43 and tau acts synergistically in a rodent model to exacerbate the pathology of either.
Collapse
Affiliation(s)
- Michael J Strong
- Molecular Medicine, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON, Canada.,Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Neil S Donison
- Molecular Medicine, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON, Canada.,Neuroscience Graduate Program, Western University, London, ON, Canada
| | - Kathryn Volkening
- Molecular Medicine, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON, Canada.,Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
12
|
Stevens CH, Guthrie NJ, van Roijen M, Halliday GM, Ooi L. Increased Tau Phosphorylation in Motor Neurons From Clinically Pure Sporadic Amyotrophic Lateral Sclerosis Patients. J Neuropathol Exp Neurol 2020; 78:605-614. [PMID: 31131395 DOI: 10.1093/jnen/nlz041] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the progressive degeneration of motor neurons. There is a pathological and genetic link between ALS and frontotemporal lobar degeneration (FTLD). Although FTLD is characterized by abnormal phosphorylated tau deposition, it is unknown whether tau is phosphorylated in ALS motor neurons. Therefore, this study assessed tau epitopes that are commonly phosphorylated in FTLD, including serine 396 (pS396), 214 (pS214), and 404 (pS404) in motor neurons from clinically pure sporadic ALS cases compared with controls. In ALS lower motor neurons, tau pS396 was observed in the nucleus or the nucleus and cytoplasm. In ALS upper motor neurons, tau pS396 was observed in the nucleus, cytoplasm, or both the nucleus and cytoplasm. Tau pS214 and pS404 was observed only in the cytoplasm of upper and lower motor neurons in ALS. The number of motor neurons (per mm2) positive for tau pS396 and pS214, but not pS404, was significantly increased in ALS. Furthermore, there was a significant loss of phosphorylated tau-negative motor neurons in ALS compared with controls. Together, our data identified a complex relationship between motor neurons positive for tau phosphorylated at specific residues and disease duration, suggesting that tau phosphorylation plays a role in ALS.
Collapse
Affiliation(s)
- Claire H Stevens
- School of Chemistry and Molecular Bioscience, University of Wollongong.,Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia
| | - Natalie J Guthrie
- School of Chemistry and Molecular Bioscience, University of Wollongong.,Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia
| | | | - Glenda M Halliday
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Lezanne Ooi
- School of Chemistry and Molecular Bioscience, University of Wollongong.,Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia
| |
Collapse
|
13
|
Angiopoietin-1 and ανβ3 integrin peptide promote the therapeutic effects of L-serine in an amyotrophic lateral sclerosis/Parkinsonism dementia complex model. Aging (Albany NY) 2019; 10:3507-3527. [PMID: 30476904 PMCID: PMC6286852 DOI: 10.18632/aging.101661] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/15/2018] [Indexed: 01/24/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult disorder of neurodegeneration that manifests as the destruction of upper and lower motor neurons. Beta-N-methylamino-L-alanine (L-BMAA), an amino acid not present in proteins, was found to cause intraneuronal protein misfolding and to induce ALS/Parkinsonism dementia complex (PDC), which presents symptoms analogous to those of Alzheimer’s-like dementia and Parkinsonism. L-serine suppresses the erroneous incorporation of L-BMAA into proteins in the human nervous system. In this study, angiopoietin-1, an endothelial growth factor crucial for vascular development and angiogenesis, and the integrin αvβ3 binding peptide C16, which inhibits inflammatory cell infiltration, were utilized to improve the local microenvironment within the central nervous system of an ALS/PDC rodent model by minimizing inflammation. Our results revealed that L-serine application yielded better effects than C16+ angiopoietin-1 treatment alone for alleviating apoptotic and autophagic changes and improving cognition and electrophysiological dysfunction, but not for improving the inflammatory micro-environment in the central nerve system, while further advances in attenuating the functional disability and pathological impairment induced by L-BMAA could be achieved by co-treatment with C16 and angiopoietin-1 in addition to L-serine. Therefore, C16+ angiopoietin-1 could be beneficial as a supplement to promote the effects of L-serine treatment.
Collapse
|
14
|
Moszczynski AJ, Harvey M, Fulcher N, de Oliveira C, McCunn P, Donison N, Bartha R, Schmid S, Strong MJ, Volkening K. Synergistic toxicity in an in vivo model of neurodegeneration through the co-expression of human TDP-43 M337V and tau T175D protein. Acta Neuropathol Commun 2019; 7:170. [PMID: 31703746 PMCID: PMC6839082 DOI: 10.1186/s40478-019-0816-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 09/22/2019] [Indexed: 02/08/2023] Open
Abstract
Although it has been suggested that the co-expression of multiple pathological proteins associated with neurodegeneration may act synergistically to induce more widespread neuropathology, experimental evidence of this is sparse. We have previously shown that the expression of Thr175Asp-tau (tauT175D) using somatic gene transfer with a stereotaxically-injected recombinant adeno-associated virus (rAAV9) vector induces tau pathology in rat hippocampus. In this study, we have examined whether the co-expression of human tauT175D with mutant human TDP-43 (TDP-43M337V) will act synergistically. Transgenic female Sprague-Dawley rats that inducibly express mutant human TDP-43M337V using the choline acetyltransferase (ChAT) tetracycline response element (TRE) driver with activity modulating tetracycline-controlled transactivator (tTA) were utilized in these studies. Adult rats were injected with GFP-tagged tau protein constructs in a rAAV9 vector through bilateral stereotaxic injection into the hippocampus. Injected tau constructs were: wild-type GFP-tagged 2N4R human tau (tauWT; n = 8), GFP-tagged tauT175D 2N4R human tau (tauT175D, pseudophosphorylated, toxic variant, n = 8), and GFP (control, n = 8). Six months post-injection, mutant TDP-43M337V expression was induced for 30 days. Behaviour testing identified motor deficits within 3 weeks after TDP-43 expression irrespective of tau expression, though social behaviour and sensorimotor gating remained unchanged. Increased tau pathology was observed in the hippocampus of both tauWT and tauT175D expressing rats and tauT175D pathology was increased in the presence of cholinergic neuronal expression of human TDP-43M337V. These data indicate that co-expression of pathological TDP-43 and tau protein exacerbate the pathology associated with either individual protein.
Collapse
|
15
|
Amir Mishan M, Rezaei Kanavi M, Shahpasand K, Ahmadieh H. Pathogenic Tau Protein Species: Promising Therapeutic Targets for Ocular Neurodegenerative Diseases. J Ophthalmic Vis Res 2019; 14:491-505. [PMID: 31875105 PMCID: PMC6825701 DOI: 10.18502/jovr.v14i4.5459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 06/11/2019] [Indexed: 12/13/2022] Open
Abstract
Tau is a microtubule-associated protein, which is highly expressed in the central nervous system as well as ocular neurons and stabilizes microtubule structure. It is a phospho-protein being moderately phosphorylated under physiological conditions but its abnormal hyperphosphorylation or some post-phosphorylation modifications would result in a pathogenic condition, microtubule dissociation, and aggregation. The aggregates can induce neuroinflammation and trigger some pathogenic cascades, leading to neurodegeneration. Taking these together, targeting pathogenic tau employing tau immunotherapy may be a promising therapeutic strategy in fighting with cerebral and ocular neurodegenerative disorders.
Collapse
Affiliation(s)
- Mohammad Amir Mishan
- Ocular Tissue Engineering Research Center, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamid Ahmadieh
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Wharton SB, Verber NS, Wagner BE, Highley JR, Fillingham DJ, Waller R, Strand K, Ince PG, Shaw PJ. Combined fused in sarcoma-positive (FUS+) basophilic inclusion body disease and atypical tauopathy presenting with an amyotrophic lateral sclerosis/motor neurone disease (ALS/MND)-plus phenotype. Neuropathol Appl Neurobiol 2019; 45:586-596. [PMID: 30659642 DOI: 10.1111/nan.12542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/14/2019] [Indexed: 12/11/2022]
Abstract
AIMS Amyotrophic lateral sclerosis/motor neurone disease (ALS/MND) is characterized by the presence of inclusions containing TDP-43 within motor neurones. In rare cases, ALS/MND may be associated with inclusions containing other proteins, such as fused in sarcoma (FUS), while motor system pathology may rarely be a feature of other neurodegenerative disorders. We here have investigated the association of FUS and tau pathology. METHODS We report a case with an ALS/MND-plus clinical syndrome which pathologically demonstrated both FUS pathology and an atypical tauopathy. RESULTS Clinical motor involvement was predominantly present in the upper motor neurone, and was accompanied by extrapyramidal features and sensory involvement, but with only minimal cognitive impairment. The presentation was sporadic and gene mutation screening was negative. Post mortem study demonstrated inclusions positive for FUS, including basophilic inclusion bodies. This was associated with 4R-tauopathy, largely as non-fibrillary diffuse phospho-tau in neurones, with granulovacuolar degeneration in a more restricted distribution. Double-staining revealed that neurones contained both types of protein pathology. CONCLUSION FUS-positive basophilic inclusion body disease is a rare cause of ALS/MND, but in this case was associated with an unusual atypical tauopathy. The coexistence of two such rare neuropathologies raises the question of a pathogenic interaction.
Collapse
Affiliation(s)
- S B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK.,Department of Histopathology, Sheffield Teaching Hospitals, Sheffield, UK
| | - N S Verber
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - B E Wagner
- Department of Histopathology, Sheffield Teaching Hospitals, Sheffield, UK
| | - J R Highley
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK.,Department of Histopathology, Sheffield Teaching Hospitals, Sheffield, UK
| | - D J Fillingham
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - R Waller
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - K Strand
- Queen Square Brain Bank for Neurological Disorders, University College London, London, UK
| | - P G Ince
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK.,Department of Histopathology, Sheffield Teaching Hospitals, Sheffield, UK
| | - P J Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| |
Collapse
|
17
|
Xue YC, Ruller CM, Fung G, Mohamud Y, Deng H, Liu H, Zhang J, Feuer R, Luo H. Enteroviral Infection Leads to Transactive Response DNA-Binding Protein 43 Pathology in Vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2853-2862. [DOI: 10.1016/j.ajpath.2018.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 08/17/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022]
|
18
|
Chardon JW, Jasmin BJ, Kothary R, Parks RJ. Report on the 4th Ottawa International Conference on Neuromuscular Disease and Biology - September 5-7, 2017, Ottawa, Canada. J Neuromuscul Dis 2018; 5:539-552. [PMID: 30373960 DOI: 10.3233/jnd-180353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jodi Warman Chardon
- Department of Medicine, The Ottawa Hospital and University of Ottawa, ON, Canada.,Department of Pediatrics (Genetics), Children's Hospital of Eastern Ontario, ON, Canada.,Neuroscience Program, Ottawa Hospital Research Institute, ON, Canada.,Centre for Neuromuscular Disease, University of Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, ON, Canada
| | - Bernard J Jasmin
- Centre for Neuromuscular Disease, University of Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, ON, Canada
| | - Rashmi Kothary
- Department of Medicine, The Ottawa Hospital and University of Ottawa, ON, Canada.,Centre for Neuromuscular Disease, University of Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, ON, Canada.,Regenerative Medicine Program, Ottawa Hospital Research Institute, ON, Canada
| | - Robin J Parks
- Department of Medicine, The Ottawa Hospital and University of Ottawa, ON, Canada.,Centre for Neuromuscular Disease, University of Ottawa, ON, Canada.,Regenerative Medicine Program, Ottawa Hospital Research Institute, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada
| |
Collapse
|
19
|
Moszczynski AJ, Gopaul J, McCunn P, Volkening K, Harvey M, Bartha R, Schmid S, Strong MJ. Somatic Gene Transfer Using a Recombinant Adenoviral Vector (rAAV9) Encoding Pseudophosphorylated Human Thr175 Tau in Adult Rat Hippocampus Induces Tau Pathology. J Neuropathol Exp Neurol 2018; 77:685-695. [DOI: 10.1093/jnen/nly044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Alexander J Moszczynski
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
| | | | | | - Kathryn Volkening
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
| | - Madeline Harvey
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
| | | | | | - Michael J Strong
- Department of Clinical Neurological Sciences, Molecular Medicine Group, Robarts Research Institute
- Department of Clinical Neurological Sciences, University Hospital, University of Western Ontario, Ontario, Canada
| |
Collapse
|
20
|
Revisiting the concept of amyotrophic lateral sclerosis as a multisystems disorder of limited phenotypic expression. Curr Opin Neurol 2018; 30:599-607. [PMID: 28914734 DOI: 10.1097/wco.0000000000000488] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW The current review will examine the contemporary evidence that amyotrophic lateral sclerosis (ALS) is a syndrome in which the unifying feature is a progressive loss of upper and lower motor neuron function. RECENT FINDINGS Although ALS is traditionally viewed as a neurodegenerative disorder affecting the motor neurons, there is considerable phenotypic heterogeneity and widespread involvement of the central nervous system. A broad range of both causative and disease modifying genetic variants are associated with both sporadic and familial forms of ALS. A significant proportion of ALS patients have an associated frontotemporal dysfunction which can be a harbinger of a significantly shorter survival and for which there is increasing evidence of a fundamental disruption of tau metabolism in those affected individuals. Although the traditional neuropathology of the degenerating motor neurons in ALS is that of neuronal cytoplasmic inclusions composed neuronal intermediate filaments, the presence of neuronal cytoplasmic inclusions composed of RNA binding proteins suggests a key role for RNA dysmetabolism in the pathogenesis of ALS. SUMMARY ALS is a complex multisystem neurodegenerative syndrome with marked heterogeneity at not only the level of clinical expression, but also etiologically.
Collapse
|
21
|
Moszczynski AJ, Hintermayer MA, Strong MJ. Phosphorylation of Threonine 175 Tau in the Induction of Tau Pathology in Amyotrophic Lateral Sclerosis-Frontotemporal Spectrum Disorder (ALS-FTSD). A Review. Front Neurosci 2018; 12:259. [PMID: 29731706 PMCID: PMC5919950 DOI: 10.3389/fnins.2018.00259] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 04/04/2018] [Indexed: 11/17/2022] Open
Abstract
Approximately 50–60% of all patients with amyotrophic lateral sclerosis (ALS) will develop a deficit of frontotemporal function, ranging from frontotemporal dementia (FTD) to one or more deficits of neuropsychological, speech or language function which are collectively known as the frontotemporal spectrum disorders of ALS (ALS-FTSD). While the neuropathology underlying these disorders is most consistent with a widespread alteration in the metabolism of transactive response DNA-binding protein 43 (TDP-43), in both ALS with cognitive impairment (ALSci) and ALS with FTD (ALS-FTD; also known as MND-FTD) there is evidence for alterations in the metabolism of the microtubule associated protein tau. This alteration in tau metabolism is characterized by pathological phosphorylation at residue Thr175 (pThr175 tau) which in vitro is associated with activation of GSK3β (pTyr216GSK3β), phosphorylation of Thr231tau, and the formation of cytoplasmic inclusions with increased rates of cell death. This putative pathway of pThr175 induction of pThr231 and the formation of pathogenic tau inclusions has been recently shown to span a broad range of tauopathies, including chronic traumatic encephalopathy (CTE) and CTE in association with ALS (CTE-ALS). This pathway can be experimentally triggered through a moderate traumatic brain injury, suggesting that it is a primary neuropathological event and not secondary to a more widespread neuronal dysfunction. In this review, we discuss the neuropathological underpinnings of the postulate that ALS is associated with a tauopathy which manifests as a FTSD, and examine possible mechanisms by which phosphorylation at Thr175tau is induced. We hypothesize that this might lead to an unfolding of the hairpin structure of tau, activation of GSK3β and pathological tau fibril formation through the induction of cis-Thr231 tau conformers. A potential role of TDP-43 acting synergistically with pathological tau metabolism is proposed.
Collapse
Affiliation(s)
- Alexander J Moszczynski
- Molecular Medicine Research Group, Schulich School of Medicine & Dentistry, Robarts Research Institute, Western University, London, ON, Canada
| | - Matthew A Hintermayer
- Molecular Medicine Research Group, Schulich School of Medicine & Dentistry, Robarts Research Institute, Western University, London, ON, Canada
| | - Michael J Strong
- Molecular Medicine Research Group, Schulich School of Medicine & Dentistry, Robarts Research Institute, Western University, London, ON, Canada.,Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| |
Collapse
|
22
|
Moszczynski AJ, Strong W, Xu K, McKee A, Brown A, Strong MJ. Pathologic Thr 175 tau phosphorylation in CTE and CTE with ALS. Neurology 2018; 90:e380-e387. [PMID: 29298849 PMCID: PMC5791789 DOI: 10.1212/wnl.0000000000004899] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 10/22/2017] [Indexed: 12/14/2022] Open
Abstract
Objective To investigate whether chronic traumatic encephalopathy (CTE) and CTE with amyotrophic lateral sclerosis (CTE-ALS) exhibit features previously observed in other tauopathies of pathologic phosphorylation of microtubule-associated protein tau at Thr175 (pThr175 tau) and Thr231 (pThr231 tau), and glycogen synthase kinase–3β (GSK3β) activation, and whether these pathologic features are a consequence of traumatic brain injury (TBI). Methods Tau isoform expression was assayed by western blot in 6 stage III CTE cases. We also used immunohistochemistry to analyze 5 cases each of CTE, CTE-ALS, and 5 controls for expression of activated GSK3β, pThr175 tau, pThr231 tau, and oligomerized tau within spinal cord tissue and hippocampus. Using a rat model of moderate TBI, we assessed tau pathology and phospho-GSK3β expression at 3 months postinjury. Results CTE and CTE-ALS are characterized by the presence of all 6 tau isoforms in both soluble and insoluble tau isolates. Activated GSK3β, pThr175 tau, pThr231 tau, and oligomerized tau protein expression was observed in hippocampal neurons and spinal motor neurons. We observed tau neuronal pathology (fibrillar inclusions and axonal damage) and increased levels of pThr175 tau and activated GSK3β in moderate TBI rats. Conclusions Pathologic phosphorylation of tau at Thr175 and Thr231 and activation of GSK3β are features of the tauopathy of CTE and CTE-ALS. These features can be replicated in an animal model of moderate TBI.
Collapse
Affiliation(s)
- Alexander J Moszczynski
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA
| | - Wendy Strong
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA
| | - Kathy Xu
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA
| | - Ann McKee
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA
| | - Arthur Brown
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA
| | - Michael J Strong
- From the Molecular Medicine Research Group, Robarts Research Institute (A.J.M., W.S., K.X., A.B., M.J.S.), and Department of Clinical Neurological Sciences (M.J.S.), Schulich School of Medicine & Dentistry, Western University, Canada; and VA Boston Healthcare System, Boston University Alzheimer's Disease and CTE Center (A.M.), Boston University School of Medicine, MA.
| |
Collapse
|
23
|
Ma L, Zhao Y, Chen Y, Cheng B, Peng A, Huang K. Caenorhabditis elegans as a model system for target identification and drug screening against neurodegenerative diseases. Eur J Pharmacol 2017; 819:169-180. [PMID: 29208474 DOI: 10.1016/j.ejphar.2017.11.051] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/30/2017] [Indexed: 12/12/2022]
Abstract
Over the past decades, Caenorhabditis elegans (C. elegans) has been widely used as a model system because of its small size, transparent body, short generation time and lifespan (~3 days and 3 weeks, respectively), completely sequenced genome and tractability to genetic manipulation. Protein misfolding and aggregation are key pathological features in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease and Amyotrophic lateral sclerosis. Animal models, including C. elegans, have been extensively used to discover and validate new drugs against neurodegenerative diseases. The well-defined and genetically tractable nervous system of C. elegans offers an effective model to explore basic mechanistic pathways of neurodegenerative diseases. Recent progress in high-throughput drug screening also provides a powerful approach for identifying chemical modulators of biological processes. Here, we summarize the latest progress of using C. elegans as a model system for target identification and drug screening in neurodegenerative diseases.
Collapse
Affiliation(s)
- Liang Ma
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yudan Zhao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuchen Chen
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Biao Cheng
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Anlin Peng
- Department of Pharmacy, The Third Hospital of Wuhan, Wuhan 430060, China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; Center for Biomedicine Research, Wuhan Institute of Biotechnology, Wuhan 430075, China.
| |
Collapse
|
24
|
Alonso R, Pisa D, Fernández-Fernández AM, Rábano A, Carrasco L. Fungal infection in neural tissue of patients with amyotrophic lateral sclerosis. Neurobiol Dis 2017; 108:249-260. [DOI: 10.1016/j.nbd.2017.09.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/24/2017] [Accepted: 09/03/2017] [Indexed: 12/11/2022] Open
|
25
|
Gu J, Wu F, Xu W, Shi J, Hu W, Jin N, Qian W, Wang X, Iqbal K, Gong CX, Liu F. TDP-43 suppresses tau expression via promoting its mRNA instability. Nucleic Acids Res 2017; 45:6177-6193. [PMID: 28335005 PMCID: PMC5449590 DOI: 10.1093/nar/gkx175] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 03/06/2017] [Indexed: 12/12/2022] Open
Abstract
In the brains of individuals with Alzheimer's disease (AD) and chronic traumatic encephalopathy, tau pathology is accompanied usually by intracellular aggregation of transactive response DNA-binding protein 43 (TDP-43). However, the role of TDP-43 in tau pathogenesis is not understood. Here, we investigated the role of TDP-43 in tau expression in vitro and in vivo. We found that TDP-43 suppressed tau expression by promoting its mRNA instability through the UG repeats of its 3΄-untranslated region (3΄-UTR). The C-terminal region of TDP-43 was required for this function. Neurodegenerative diseases-causing TDP-43 mutations affected tau mRNA instability differentially, in that some promoted and others did not significantly affect tau mRNA instability. The expression levels of tau and TDP-43 were inverse in the frontal cortex and the cerebellum. Accompanied with cytoplasmic accumulation of TDP-43, tau expression was elevated in TDP-43M337V transgenic mouse brains. The level of TDP-43, which is decreased in AD brains, was found to correlate negatively with the tau level in human brain. Our findings indicate that TDP-43 suppresses tau expression by promoting the instability of its mRNA. Down-regulation of TDP-43 may be involved in the tau pathology in AD and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Jianlan Gu
- Key Laboratory of Neuroregeneration of Jiangsu and ministry of education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
- Department of Biochemistry and molecular biology, School of Medicine, Nantong University, Nantong, Jiangsu 226001, China
| | - Feng Wu
- Key Laboratory of Neuroregeneration of Jiangsu and ministry of education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Wen Xu
- Key Laboratory of Neuroregeneration of Jiangsu and ministry of education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Jianhua Shi
- Key Laboratory of Neuroregeneration of Jiangsu and ministry of education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Wen Hu
- Key Laboratory of Neuroregeneration of Jiangsu and ministry of education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Nana Jin
- Key Laboratory of Neuroregeneration of Jiangsu and ministry of education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Wei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and ministry of education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Department of Biochemistry and molecular biology, School of Medicine, Nantong University, Nantong, Jiangsu 226001, China
| | - Xinglong Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Cheng-Xin Gong
- Key Laboratory of Neuroregeneration of Jiangsu and ministry of education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Fei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and ministry of education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
- To whom correspondence should be addressed. Tel: +1 718 494 5263; Fax: +1 718 494 1080;
| |
Collapse
|
26
|
Takeda T. Possible concurrence of TDP-43, tau and other proteins in amyotrophic lateral sclerosis/frontotemporal lobar degeneration. Neuropathology 2017; 38:72-81. [PMID: 28960544 DOI: 10.1111/neup.12428] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/15/2017] [Accepted: 08/15/2017] [Indexed: 12/11/2022]
Abstract
Transactivation response DNA-binding protein 43 kDa (TDP-43) has been regarded as a major component of ubiquitin-positive/tau-negative inclusions of motor neurons and the frontotemporal cortices in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Neurofibrillary tangles (NFT), an example of tau-positive inclusions, are biochemically and morphologically distinguished from TDP-43-positive inclusions, and are one of the pathological core features of Alzheimer disease (AD). Although ALS/FTLD and AD are distinct clinical entities, they can coexist in an individual patient. Whether concurrence of ALS/FTLD-TDP-43 and AD-tau is incidental is still controversial, because aging is a common risk factor for ALS/FTLD and AD development. Indeed, it remains unclear whether the pathogenesis of ALS/FTLD is a direct causal link to tau accumulation. Recent studies suggested that AD pathogenesis could cause the accumulation of TDP-43, while abnormal TDP-43 accumulation could also lead to abnormal tau expression. Overlapping presence of TDP-43 and tau, when observed in a brain during autopsy, should attract attention, and should initiate the search for the pathological substrate for this abnormal protein accumulation. In addition to tau, other proteins including α-synuclein and amyloid β should be also taken into account as candidates for an interaction with TDP-43. Awareness of a possible comorbidity between TDP-43, tau and other proteins in patients with ALS/FTLD will be useful for our understanding of the influence of these proteins on the disease development and its clinical manifestation.
Collapse
Affiliation(s)
- Takahiro Takeda
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
27
|
Gleixner AM, Hutchison DF, Sannino S, Bhatia TN, Leak LC, Flaherty PT, Wipf P, Brodsky JL, Leak RK. N-Acetyl-l-Cysteine Protects Astrocytes against Proteotoxicity without Recourse to Glutathione. Mol Pharmacol 2017; 92:564-575. [PMID: 28830914 DOI: 10.1124/mol.117.109926] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
N-acetyl-l-cysteine (NAC) exhibits protective properties in brain injury models and has undergone a number of clinical trials. Most studies of NAC have focused on neurons. However, neuroprotection may be complemented by the protection of astrocytes because healthier astrocytes can better support the viability of neurons. Here, we show that NAC can protect astrocytes against protein misfolding stress (proteotoxicity), the hallmark of neurodegenerative disorders. Although NAC is thought to be a glutathione precursor, NAC protected primary astrocytes from the toxicity of the proteasome inhibitor MG132 without eliciting any increase in glutathione. Furthermore, glutathione depletion failed to attenuate the protective effects of NAC. MG132 elicited a robust increase in the folding chaperone heat shock protein 70 (Hsp70), and NAC mitigated this effect. Nevertheless, three independent inhibitors of Hsp70 function ablated the protective effects of NAC, suggesting that NAC may help preserve Hsp70 chaperone activity and improve protein quality control without need for Hsp70 induction. Consistent with this view, NAC abolished an increase in ubiquitinated proteins in MG132-treated astrocytes. However, NAC did not affect the loss of proteasome activity in response to MG132, demonstrating that it boosted protein homeostasis and cell viability without directly interfering with the efficacy of this proteasome inhibitor. The thiol-containing molecules l-cysteine and d-cysteine both mimicked the protective effects of NAC, whereas the thiol-lacking molecule N-acetyl-S-methyl-l-cysteine failed to exert protection or blunt the rise in ubiquitinated proteins. Collectively, these findings suggest that the thiol group in NAC is required for its effects on glial viability and protein quality control.
Collapse
Affiliation(s)
- Amanda M Gleixner
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Daniel F Hutchison
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Sara Sannino
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Tarun N Bhatia
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Lillian C Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Patrick T Flaherty
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Peter Wipf
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Jeffrey L Brodsky
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| |
Collapse
|
28
|
Spires-Jones TL, Attems J, Thal DR. Interactions of pathological proteins in neurodegenerative diseases. Acta Neuropathol 2017; 134:187-205. [PMID: 28401333 PMCID: PMC5508034 DOI: 10.1007/s00401-017-1709-7] [Citation(s) in RCA: 270] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/30/2017] [Accepted: 04/01/2017] [Indexed: 02/06/2023]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), frontotemporal lobar degeneration (FTD), Lewy body disease (LBD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) have in common that protein aggregates represent pathological hallmark lesions. Amyloid β-protein, τ-protein, α-synuclein, and TDP-43 are the most frequently aggregated proteins in these disorders. Although they are assumed to form disease-characteristic aggregates, such as amyloid plaques and neurofibrillary tangles in AD or Lewy bodies in LBD/PD, they are not restricted to these clinical presentations. They also occur in non-diseased individuals and can co-exist in the same brain without or with a clinical picture of a distinct dementing or movement disorder. In this review, we discuss the co-existence of these pathologies and potential additive effects in the human brain as well as related functional findings on cross-seeding and molecular interactions between these aggregates/proteins. We conclude that there is evidence for interactions at the molecular level as well as for additive effects on brain damage by multiple pathologies occurring in different functionally important neurons. Based upon this information, we hypothesize a cascade of events that may explain general mechanisms in the development of neurodegenerative disorders: (1) distinct lesions are a prerequisite for the development of a distinct disease (e.g., primary age-related tauopathy for AD), (2) disease-specific pathogenic events further trigger the development of a specific disease (e.g., Aβ aggregation in AD that exaggerate further Aβ and AD-related τ pathology), (3) the symptomatic disease manifests, and (4) neurodegenerative co-pathologies may be either purely coincidental or (more likely) have influence on the disease development and/or its clinical presentation.
Collapse
Affiliation(s)
- Tara L Spires-Jones
- Centre for Dementia Prevention, and Euan MacDonald Centre for Motor Neurone Disease, The University of Edinburgh Centre for Cognitive and Neural Systems, 1 George Square, Edinburgh, EH8 9JZ, UK.
| | - Johannes Attems
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Dietmar Rudolf Thal
- Departement Neurowetenschappen, Katholieke Universiteit Leuven, Herestraat 49, 3000, Leuven, Belgium
- Departement Pathologische Ontleedkunde, UZ Leuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
29
|
Volkening K, Strong WL, Seaton S, Yang W, Strong MJ. C9orf72 mutations do not influence the tau signature of amyotrophic lateral sclerosis with cognitive impairment (ALSci). Amyotroph Lateral Scler Frontotemporal Degener 2017; 18:549-554. [PMID: 28562075 DOI: 10.1080/21678421.2017.1332075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE C9orf72 mutations are associated with amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD) and ALS-FTD. In addition to ALS-FTD, ALS patients may develop a spectrum of neuropsychological and neuropsychiatric deficits including ALS with cognitive impairment (ALSci). Here we examine the extent to which C9orf72 mutations are associated with ALSci and whether this alters the tau molecular signature. METHODS We identified 16 ALSci cases within a post-mortem archive of 94 fully genotyped ALS cases, eight of which harboured a C9orf72 mutation, in addition to three cognitively-intact ALS cases with a C9orf72 mutation. Tau was fractionated into soluble and insoluble fractions, with or without dephosphorylation, and immunoblots for tau phospho-isoforms performed. RESULTS Regardless of cognitive state or the presence of C9orf72 mutation, all ALS cases demonstrated six tau isoforms in both soluble and insoluble tau isolates. This pattern was unaffected by dephosphorylation. pThr175tau isoforms, a molecular signature of ALSci, were present regardless of C9orf72 genetic status. The pathognomic paired helical triplet in the insoluble tau fraction of Alzheimer's disease was not observed, regardless of cognitive or C9orf72 status. CONCLUSIONS These findings suggest that the presence of a C9orf72 mutation does not influence the tau signature of ALS or ALSci.
Collapse
Affiliation(s)
- Kathryn Volkening
- a Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine & Dentistry , University of Western Ontario , London , Ontario , Canada and.,b Clinical Neurological Sciences, Schulich School of Medicine & Dentistry , University of Western Ontario , London , Ontario , Canada
| | - Wendy L Strong
- a Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine & Dentistry , University of Western Ontario , London , Ontario , Canada and
| | - Shauntel Seaton
- a Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine & Dentistry , University of Western Ontario , London , Ontario , Canada and
| | - Wencheng Yang
- a Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine & Dentistry , University of Western Ontario , London , Ontario , Canada and
| | - Michael J Strong
- a Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine & Dentistry , University of Western Ontario , London , Ontario , Canada and.,b Clinical Neurological Sciences, Schulich School of Medicine & Dentistry , University of Western Ontario , London , Ontario , Canada
| |
Collapse
|
30
|
Strong MJ, Abrahams S, Goldstein LH, Woolley S, Mclaughlin P, Snowden J, Mioshi E, Roberts-South A, Benatar M, HortobáGyi T, Rosenfeld J, Silani V, Ince PG, Turner MR. Amyotrophic lateral sclerosis - frontotemporal spectrum disorder (ALS-FTSD): Revised diagnostic criteria. Amyotroph Lateral Scler Frontotemporal Degener 2017; 18:153-174. [PMID: 28054827 PMCID: PMC7409990 DOI: 10.1080/21678421.2016.1267768] [Citation(s) in RCA: 585] [Impact Index Per Article: 83.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 10/20/2022]
Abstract
This article presents the revised consensus criteria for the diagnosis of frontotemporal dysfunction in amyotrophic lateral sclerosis (ALS) based on an international research workshop on frontotemporal dementia (FTD) and ALS held in London, Canada in June 2015. Since the publication of the Strong criteria, there have been considerable advances in the understanding of the neuropsychological profile of patients with ALS. Not only is the breadth and depth of neuropsychological findings broader than previously recognised - - including deficits in social cognition and language - but mixed deficits may also occur. Evidence now shows that the neuropsychological deficits in ALS are extremely heterogeneous, affecting over 50% of persons with ALS. When present, these deficits significantly and adversely impact patient survival. It is the recognition of this clinical heterogeneity in association with neuroimaging, genetic and neuropathological advances that has led to the current re-conceptualisation that neuropsychological deficits in ALS fall along a spectrum. These revised consensus criteria expand upon those of 2009 and embrace the concept of the frontotemporal spectrum disorder of ALS (ALS-FTSD).
Collapse
Affiliation(s)
- Michael J Strong
- a Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry , London , Ontario , Canada
| | - Sharon Abrahams
- b Department of Psychology, School of Philosophy, Psychology & Language Sciences , Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh , Edinburgh , UK
| | - Laura H Goldstein
- c King's College London, Department of Psychology, Institute of Psychiatry, Psychology and Neuroscience , London , UK
| | - Susan Woolley
- d Forbes Norris MDA/ALS Research Centre, California Pacific Medical Centre , San Francisco , CA , USA
| | - Paula Mclaughlin
- e Western University , Schulich School of Medicine & Dentistry , London , ON , Canada
| | - Julie Snowden
- f Greater Manchester Neuroscience Centre , Salford Royal NHS Trust and University of Manchester , Manchester , UK
| | - Eneida Mioshi
- g Faculty of Medicine and Health Sciences , University of East Anglia , Norwich , UK
| | - Angie Roberts-South
- h Northwestern University , Roxelyn and Richard Pepper Department of Communication Sciences and Disorders , Evanston , IL , USA
| | - Michael Benatar
- i Department of Neurology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Tibor HortobáGyi
- j Department of Neuropathology , Institute of Pathology, University of Debrecen , Debrecen , Hungary
| | - Jeffrey Rosenfeld
- k Department of Neurology , Loma Linda University School of Medicine , Loma Linda , CA , USA
| | - Vincenzo Silani
- l Department of Neurology and Laboratory Neuroscience - IRCCS Istituto Auxologico Italiano, Department of Pathophysiology and Transplantation , 'Dino Ferrari' Centre, Università degli Studi di Milano , Milan , Italy
| | - Paul G Ince
- m Sheffield Institute for Translational Neuroscience, Department of Neuroscience , The University of Sheffield , Sheffield , UK , and
| | - Martin R Turner
- n Nuffield Department of Clinical Neurosciences , University of Oxford , Oxford , UK
| |
Collapse
|
31
|
Moszczynski AJ, Yang W, Hammond R, Ang LC, Strong MJ. Threonine 175, a novel pathological phosphorylation site on tau protein linked to multiple tauopathies. Acta Neuropathol Commun 2017; 5:6. [PMID: 28077166 PMCID: PMC5225652 DOI: 10.1186/s40478-016-0406-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/15/2016] [Indexed: 12/13/2022] Open
Abstract
Microtubule associated protein tau (tau) deposition is associated with a spectrum of neurodegenerative diseases collectively termed tauopathies. We have previously shown that amyotrophic lateral sclerosis (ALS) with cognitive impairment (ALSci) is associated with tau phosphorylation at Thr175 and that this leads to activation of GSK3β which then induces phosphorylation at tau Thr231. This latter step leads to dissociation of tau from microtubules and pathological tau fibril formation. To determine the extent to which this pathway is unique to ALS, we have investigated the expression of pThr175 tau and pThr231 tau across a range of frontotemporal degenerations. Representative sections from the superior frontal cortex, anterior cingulate cortex (ACC), amygdala, hippocampal formation, basal ganglia, and substantia nigra were selected from neuropathologically confirmed cases of Alzheimer’s disease (AD; n = 3), vascular dementia (n = 2), frontotemporal lobar degeneration (FTLD; n = 4), ALS (n = 5), ALSci (n = 6), Parkinson’s disease (PD; n = 5), corticobasal degeneration (CBD; n = 2), diffuse Lewy body dementia (DLBD; n = 2), mixed DLBD (n = 3), multisystem atrophy (MSA; n = 6) and Pick’s disease (n = 1) and three neuropathologically-normal control groups aged 50–60 (n = 6), 60–70 (n = 6) and 70–80 (n = 8). Sections were examined using a panel of phospho-tau antibodies (pSer208,210, pThr217, pThr175, pThr231, pSer202 and T22 (oligomeric tau)). Across diseases, phospho-tau load was most prominent in layers II/III of the entorhinal cortex, amygdala and hippocampus. This is in contrast to the preferential deposition of phospho-tau in the ACC and frontal cortex in ALSci. Controls showed pThr175 tau expression only in the 7th decade of life and only in the presence of tau pathology and tau oligomers. With the exception of DLBD, we observed pThr175 co-localizing with pThr231 in the same cell populations as T22 positivity. This suggests that this pathway may be a common mechanism of toxicity across the tauopathies.
Collapse
|
32
|
Vintilescu CR, Afreen S, Rubino AE, Ferreira A. The Neurotoxic TAU 45-230 Fragment Accumulates in Upper and Lower Motor Neurons in Amyotrophic Lateral Sclerosis Subjects. Mol Med 2016; 22:477-486. [PMID: 27496042 PMCID: PMC5072411 DOI: 10.2119/molmed.2016.00095] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/23/2016] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and lethal neurodegenerative disease characterized by the loss of upper and lower motor neurons leading to muscle paralysis in affected individuals. Numerous mechanisms have been implicated in the death of these neurons. However, the pathobiology of this disease has not been completely elucidated. In the present study, we investigated to what extent tau cleavage and the generation of the neurotoxic tau45-230 fragment is associated with ALS. Quantitative Western blot analysis indicated that high levels of tau45-230 accumulated in lumbar and cervical spinal cord specimens obtained from ALS subjects. This neurotoxic tau fragment was also detected in ALS upper motor neurons located in the precentral gyrus. Our results also showed that tau45-230 aggregates were present in the spinal cord of ALS patients. On the other hand, this neurotoxic fragment was not generated in a mouse model of a familial form of this disease. Together, these results suggest a potential role for this neurotoxic tau fragment in the mechanisms leading to the degeneration of motor neurons in the context of sporadic ALS.
Collapse
Affiliation(s)
- Claudia R Vintilescu
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, IL 60611
| | - Sana Afreen
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, IL 60611
| | - Ashlee E Rubino
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, IL 60611
| | - Adriana Ferreira
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, IL 60611
| |
Collapse
|
33
|
Gleixner AM, Posimo JM, Pant DB, Henderson MP, Leak RK. Astrocytes Surviving Severe Stress Can Still Protect Neighboring Neurons from Proteotoxic Injury. Mol Neurobiol 2016; 53:4939-60. [PMID: 26374549 PMCID: PMC4792804 DOI: 10.1007/s12035-015-9427-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 09/03/2015] [Indexed: 12/18/2022]
Abstract
Astrocytes are one of the major cell types to combat cellular stress and protect neighboring neurons from injury. In order to fulfill this important role, astrocytes must sense and respond to toxic stimuli, perhaps including stimuli that are severely stressful and kill some of the astrocytes. The present study demonstrates that primary astrocytes that managed to survive severe proteotoxic stress were protected against subsequent challenges. These findings suggest that the phenomenon of preconditioning or tolerance can be extended from mild to severe stress for this cell type. Astrocytic stress adaptation lasted at least 96 h, the longest interval tested. Heat shock protein 70 (Hsp70) was raised in stressed astrocytes, but inhibition of neither Hsp70 nor Hsp32 activity abolished their resistance against a second proteotoxic challenge. Only inhibition of glutathione synthesis abolished astrocytic stress adaptation, consistent with our previous report. Primary neurons were plated upon previously stressed astrocytes, and the cocultures were then exposed to another proteotoxic challenge. Severely stressed astrocytes were still able to protect neighboring neurons against this injury, and the protection was unexpectedly independent of glutathione synthesis. Stressed astrocytes were even able to protect neurons after simultaneous application of proteasome and Hsp70 inhibitors, which otherwise elicited synergistic, severe loss of neurons when applied together. Astrocyte-induced neuroprotection against proteotoxicity was not elicited with astrocyte-conditioned media, suggesting that physical cell-to-cell contacts may be essential. These findings suggest that astrocytes may adapt to severe stress so that they can continue to protect neighboring cell types from profound injury.
Collapse
Affiliation(s)
- Amanda M Gleixner
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 407 Mellon Hall, 600 Forbes Avenue, Pittsburgh, PA, 15282, USA
| | - Jessica M Posimo
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 407 Mellon Hall, 600 Forbes Avenue, Pittsburgh, PA, 15282, USA
| | - Deepti B Pant
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 407 Mellon Hall, 600 Forbes Avenue, Pittsburgh, PA, 15282, USA
| | - Matthew P Henderson
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 407 Mellon Hall, 600 Forbes Avenue, Pittsburgh, PA, 15282, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 407 Mellon Hall, 600 Forbes Avenue, Pittsburgh, PA, 15282, USA.
| |
Collapse
|
34
|
Przybyla M, Stevens CH, van der Hoven J, Harasta A, Bi M, Ittner A, van Hummel A, Hodges JR, Piguet O, Karl T, Kassiou M, Housley GD, Ke YD, Ittner LM, van Eersel J. Disinhibition-like behavior in a P301S mutant tau transgenic mouse model of frontotemporal dementia. Neurosci Lett 2016; 631:24-29. [DOI: 10.1016/j.neulet.2016.08.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/19/2016] [Accepted: 08/04/2016] [Indexed: 11/28/2022]
|
35
|
Pathological tau deposition in Motor Neurone Disease and frontotemporal lobar degeneration associated with TDP-43 proteinopathy. Acta Neuropathol Commun 2016; 4:33. [PMID: 27036121 PMCID: PMC4818389 DOI: 10.1186/s40478-016-0301-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/16/2016] [Indexed: 12/12/2022] Open
Abstract
It has been suggested that patients with motor neurone disease (MND) and those with MND combined with behavioural variant frontotemporal dementia (bvFTD) (ie FTD + MND) or with FTD alone might exist on a continuum based on commonalities of neuropathology and/or genetic risk. Moreover, it has been reported that both a neuronal and a glial cell tauopathy can accompany the TDP-43 proteinopathy in patients with motor neurone disease (MND) with cognitive changes, and that the tauopathy may be fundamental to disease pathogenesis and clinical phenotype. In the present study, we sought to substantiate these latter findings, and test this concept of a pathological continuum, in a consecutive series of 41 patients with MND, 16 with FTD + MND and 23 with FTD without MND. Paraffin sections of frontal, entorhinal, temporal and occipital cortex and hippocampus were immunostained for tau pathology using anti-tau antibodies, AT8, pThr175 and pThr217, and for amyloid β protein (Aβ) using 4G8 antibody. Twenty four (59 %) patients with MND, 7 (44 %) patients with FTD + MND and 10 (43 %) patients with FTD showed ‘significant’ tau pathology (ie more than just an isolated neurofibrillary tangle or a few neuropil threads in one or more brain regions examined). In most instances, this bore the histological characteristics of an Alzheimer’s disease process involving entorhinal cortex, hippocampus, temporal cortex, frontal cortex and occipital cortex in decreasing frequency, accompanied by a deposition of Aβ up to Thal phase 3, though 2 patients with MND, and 1 with FTD did show tau pathology beyond Braak stage III. Four other patients with MND showed novel neuronal tau pathology, within the frontal cortex alone, specifically detected by pThr175 antibody, which was characterised by a fine granular or more clumped aggregation of tau without neurofibrillary tangles or neuropil threads. However, none of these 4 patients had clinically evident cognitive disorder, and this type of tau pathology was not seen in any of the FTD + MND or FTD patients. Finally, two patients, one with MND and one with FTD, showed a tau pathology consistent with Argyrophilic Grain Disease (AGD). Western blotting and use of 3- and 4-repeat tau antibodies confirmed the histological interpretation of Alzheimer’s disease type pathology in all instances except for those patients with accompanying AGD where a banding pattern on western blot, and immunohistochemistry, confirmed 4-repeat tauopathy. In all 3 patient groups, amyloid pathology was more likely to be present in patients dying after 65 years of age, and in the presence of APOE ε4 allele. We conclude that tau pathological changes are equally common amongst patients with MND, FTD + MND and FTD though, in most instances, these are limited in extent. In patients with MND, when cognitive impairment is present this is most likely due to an accompanying/evolving (coincidental) Alzheimer’s disease process or, as in a single case, Dementia with Lewy bodies, within the cerebral cortex rather than as a result of TDP-43 proteinopathy. Conversely, in FTD and FTD + MND dementia is more likely to be associated with TDP-43 proteinopathy than tau. Hence, present study shows no progression in severity of (tau) pathology from MND through FTD + MND to FTD, and does not support the concept of these conditions forming a continuum of clinical or pathological change.
Collapse
|
36
|
Kahlson MA, Colodner KJ. Glial Tau Pathology in Tauopathies: Functional Consequences. J Exp Neurosci 2016; 9:43-50. [PMID: 26884683 PMCID: PMC4750898 DOI: 10.4137/jen.s25515] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/21/2015] [Accepted: 12/29/2015] [Indexed: 12/22/2022] Open
Abstract
Tauopathies are a class of neurodegenerative diseases characterized by the presence of hyperphosphorylated and aggregated tau pathology in neuronal and glial cells. Though the ratio of neuronal and glial tau aggregates varies across diseases, glial tau aggregates can populate the same degenerating brain regions as neuronal tau aggregates. While much is known about the deleterious consequences of tau pathology in neurons, the relative contribution of glial tau pathology to these diseases is less clear. Recent studies using a number of model systems implicate glial tau pathology in contributing to tauopathy pathogenesis. This review aims to highlight the functional consequences of tau overexpression in glial cells and explore the potential contribution of glial tau pathology in the pathogenesis of neurodegenerative tauopathies.
Collapse
Affiliation(s)
- Martha A Kahlson
- Department of Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, USA
| | - Kenneth J Colodner
- Department of Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, USA
| |
Collapse
|
37
|
Woolley SC, Strong MJ. Frontotemporal Dysfunction and Dementia in Amyotrophic Lateral Sclerosis. Neurol Clin 2015; 33:787-805. [PMID: 26515622 DOI: 10.1016/j.ncl.2015.07.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although amyotrophic lateral sclerosis (ALS) is classically considered a disorder exclusively affecting motor neurons, there is substantial clinical, neuroimaging, and neuropathologic evidence that more than half of patients have an associated syndrome of frontotemporal dysfunction. These syndromes range from frontotemporal dementia to behavioral or cognitive syndromes. Neuroimaging and neuropathologic findings are consistent with frontotemporal lobar degeneration that underpins alterations in network connectivity. Future clinical trials need to be stratified based on the presence or absence of frontotemporal dysfunction on the disease course of ALS.
Collapse
Affiliation(s)
- Susan C Woolley
- Forbes Norris MDA/ALS Research Center, California Pacific Medical Center, 2324 Sacramento Street, Suite 111, San Francisco, CA 94115, USA
| | - Michael J Strong
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
38
|
Coan G, Mitchell CS. An Assessment of Possible Neuropathology and Clinical Relationships in 46 Sporadic Amyotrophic Lateral Sclerosis Patient Autopsies. NEURODEGENER DIS 2015; 15:301-12. [PMID: 26183171 DOI: 10.1159/000433581] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 05/26/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Recent studies have suggested overlapping pathological features among motor neuron, cognitive and neurodegenerative diseases. AIMS/METHODS Secondary analysis of 46 amyotrophic lateral sclerosis (ALS) patient autopsies was performed to independently assess pathological feature prevalence (e.g. percent of patients with any positive finding), degree of severity (e.g. mild, moderate, severe), and 2,200+ potential clinical/neuropathological correlations. The possible impact of gender, onset age, onset type (limb vs. bulbar), riluzole treatment, and severe TDP-43 pathology was assessed within patient subgroups. RESULTS Assessed features (prevalence, severity) include: lateral corticospinal tract degeneration (89%, moderate); Purkinje cell loss (85%, mild); localized neuronal loss (83%, mild to moderate); TDP-43 inclusions (80%, moderate); Betz cell loss (76%, mild); neurofibrillary tangles (78%, severe); anterior corticospinal tract degeneration (72%, moderate); spinal ventral root atrophy (65%, moderate); atherosclerosis (35%, mild); β-amyloid (35%, mild); tauopathy/tau inclusions (17%, mild); ventricular dilation (13%, mild); Lewy body formation (11%, mild); microinfarcts (7%, mild); and α-synuclein (4%, mild). Twenty-two percent of patients met criteria for Alzheimer's disease (AD) and 26% for frontotemporal lobar degeneration. Substantial differences were identified in the AD group and in the different onset age groups. CONCLUSION Our findings support the hypothesis that ALS and its variants could comprise a larger neuropathological continuum.
Collapse
Affiliation(s)
- Grant Coan
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Ga., USA
| | | |
Collapse
|
39
|
Alonso R, Pisa D, Marina AI, Morato E, Rábano A, Rodal I, Carrasco L. Evidence for fungal infection in cerebrospinal fluid and brain tissue from patients with amyotrophic lateral sclerosis. Int J Biol Sci 2015; 11:546-58. [PMID: 25892962 PMCID: PMC4400386 DOI: 10.7150/ijbs.11084] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/10/2015] [Indexed: 12/14/2022] Open
Abstract
Among neurogenerative diseases, amyotrophic lateral sclerosis (ALS) is a fatal illness characterized by a progressive motor neuron dysfunction in the motor cortex, brainstem and spinal cord. ALS is the most common form of motor neuron disease; yet, to date, the exact etiology of ALS remains unknown. In the present work, we have explored the possibility of fungal infection in cerebrospinal fluid (CSF) and in brain tissue from ALS patients. Fungal antigens, as well as DNA from several fungi, were detected in CSF from ALS patients. Additionally, examination of brain sections from the frontal cortex of ALS patients revealed the existence of immunopositive fungal antigens comprising punctate bodies in the cytoplasm of some neurons. Fungal DNA was also detected in brain tissue using PCR analysis, uncovering the presence of several fungal species. Finally, proteomic analyses of brain tissue demonstrated the occurrence of several fungal peptides. Collectively, our observations provide compelling evidence of fungal infection in the ALS patients analyzed, suggesting that this infection may play a part in the etiology of the disease or may constitute a risk factor for these patients.
Collapse
Affiliation(s)
- Ruth Alonso
- 1. Centro de Biología Molecular "Severo Ochoa". c/Nicolás Cabrera, 1. Universidad Autónoma de Madrid. Cantoblanco. 28049 Madrid. Spain
| | - Diana Pisa
- 1. Centro de Biología Molecular "Severo Ochoa". c/Nicolás Cabrera, 1. Universidad Autónoma de Madrid. Cantoblanco. 28049 Madrid. Spain
| | - Ana Isabel Marina
- 1. Centro de Biología Molecular "Severo Ochoa". c/Nicolás Cabrera, 1. Universidad Autónoma de Madrid. Cantoblanco. 28049 Madrid. Spain
| | - Esperanza Morato
- 1. Centro de Biología Molecular "Severo Ochoa". c/Nicolás Cabrera, 1. Universidad Autónoma de Madrid. Cantoblanco. 28049 Madrid. Spain
| | - Alberto Rábano
- 2. Department of Neuropathology and Tissue Bank, Unidad de Investigación Proyecto Alzheimer, Fundación CIEN, Instituto de Salud Carlos III, Madrid. Spain
| | - Izaskun Rodal
- 2. Department of Neuropathology and Tissue Bank, Unidad de Investigación Proyecto Alzheimer, Fundación CIEN, Instituto de Salud Carlos III, Madrid. Spain
| | - Luis Carrasco
- 1. Centro de Biología Molecular "Severo Ochoa". c/Nicolás Cabrera, 1. Universidad Autónoma de Madrid. Cantoblanco. 28049 Madrid. Spain
| |
Collapse
|
40
|
Moszczynski AJ, Gohar M, Volkening K, Leystra-Lantz C, Strong W, Strong MJ. Thr175-phosphorylated tau induces pathologic fibril formation via GSK3β-mediated phosphorylation of Thr231 in vitro. Neurobiol Aging 2014; 36:1590-9. [PMID: 25573097 DOI: 10.1016/j.neurobiolaging.2014.12.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/11/2014] [Accepted: 12/03/2014] [Indexed: 12/13/2022]
Abstract
We have previously shown that amyotrophic lateral sclerosis with cognitive impairment can be characterized by pathologic inclusions of microtubule-associated protein tau (tau) phosphorylated at Thr(175) (pThr(175)) in association with GSK3β activation. We have now examined whether pThr(175) induces GSK3β activation and whether this leads to pathologic fibril formation through Thr(231) phosphorylation. Seventy-two hours after transfection of Neuro2A cells with pseudophosphorylated green fluorescent protein-tagged 2N4R tau (Thr(175)Asp), phosphorylated kinase glycogen synthase kinase 3 beta (active GSK3β) levels were significantly increased as was pathologic fibril formation and cell death. Treatment with each of 4 GSK3β inhibitors or small hairpin RNA knockdown of GSK3β abolished fibril formation and prevented cell death. Inhibition of Thr(231) phosphorylation (Thr(231)Ala) prevented pathologic tau fibril formation, regardless of Thr(175) state, whereas Thr(231)Asp (pseudophosphorylated at Thr(231)) developed pathologic tau fibrils. Ser(235) mutations did not affect fibril formation, indicating an unprimed mechanism of Thr(231) phosphorylation. These findings suggest a mechanism of tau pathology by which pThr(175) induces GSK3β phosphorylation of Thr(231) leading to fibril formation, indicating a potential therapeutic avenue for amyotrophic lateral sclerosis with cognitive impairment.
Collapse
Affiliation(s)
- Alexander J Moszczynski
- Graduate Program in Neuroscience, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - May Gohar
- Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Kathryn Volkening
- Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Cheryl Leystra-Lantz
- Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Wendy Strong
- Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Michael J Strong
- Graduate Program in Neuroscience, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
41
|
Fang P, Xu W, Wu C, Zhu M, Li X, Hong D. MAPT as a predisposing gene for sporadic amyotrophic lateral sclerosis in the Chinese Han population. Neural Regen Res 2014; 8:3116-23. [PMID: 25206632 PMCID: PMC4158701 DOI: 10.3969/j.issn.1673-5374.2013.33.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/15/2013] [Indexed: 01/23/2023] Open
Abstract
A previous study of European Caucasian patients with sporadic amyotrophic lateral sclerosis demonstrated that a polymorphism in the microtubule-associated protein Tau (MAPT) gene was significantly associated with sporadic amyotrophic lateral sclerosis pathogenesis. Here, we tested this association in 107 sporadic amyotrophic lateral sclerosis patients and 100 healthy controls from the Chinese Han population. We screened the mutation-susceptible regions of MAPT - the 3' and 5' untranslated regions as well as introns 9, 10, 11, and 12 - by direct sequencing, and identified 33 genetic variations. Two of these, 105788 A > G in intron 9 and 123972 T > A in intron 11, were not present in the control group. The age of onset in patients with the 105788 A > G and/or the 123972 T > A variant was younger than that in patients without either genetic variation. Moreover, the pa-tients with a genetic variation were more prone to bulbar palsy and breathing difficulties than those with the wild-type genotype. This led to a shorter survival period in patients with a MAPT genetic variant. Our study suggests that the MAPT gene is a potential risk gene for sporadic amyotrophic lateral sclerosis in the Chinese Han population.
Collapse
Affiliation(s)
- Pu Fang
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Wenyuan Xu
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Chengsi Wu
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Min Zhu
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Xiaobing Li
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Daojun Hong
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
42
|
Faravelli I, Riboldi G, Nizzardo M, Simone C, Zanetta C, Bresolin N, Comi GP, Corti S. Stem cell transplantation for amyotrophic lateral sclerosis: therapeutic potential and perspectives on clinical translation. Cell Mol Life Sci 2014; 71:3257-68. [PMID: 24699704 PMCID: PMC11113626 DOI: 10.1007/s00018-014-1613-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/26/2014] [Accepted: 03/17/2014] [Indexed: 12/14/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease characterized by degeneration of upper and lower motor neurons. There are currently no clinically impactful treatments for this disorder. Death occurs 3-5 years after diagnosis, usually due to respiratory failure. ALS pathogenesis seems to involve several pathological mechanisms (i.e., oxidative stress, inflammation, and loss of the glial neurotrophic support, glutamate toxicity) with different contributions from environmental and genetic factors. This multifaceted combination highlights the concept that an effective therapeutic approach should counteract simultaneously different aspects: stem cell therapies are able to maintain or rescue motor neuron function and modulate toxicity in the central nervous system (CNS) at the same time, eventually representing the most comprehensive therapeutic approach for ALS. To achieve an effective cell-mediated therapy suitable for clinical applications, several issues must be addressed, including the identification of the most performing cell source, a feasible administration protocol, and the definition of therapeutic mechanisms. The method of cell delivery represents a major issue in developing cell-mediated approaches since the cells, to be effective, need to be spread across the CNS, targeting both lower and upper motor neurons. On the other hand, there is the need to define a strategy that could provide a whole distribution without being too invasive or burdened by side effects. Here, we review the recent advances regarding the therapeutic potential of stem cells for ALS with a focus on the minimally invasive strategies that could facilitate an extensive translation to their clinical application.
Collapse
Affiliation(s)
- Irene Faravelli
- Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, Dino Ferrari Centre, Neurology Unit, University of Milan, IRCCS Foundation Ca’Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Giulietta Riboldi
- Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, Dino Ferrari Centre, Neurology Unit, University of Milan, IRCCS Foundation Ca’Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Monica Nizzardo
- Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, Dino Ferrari Centre, Neurology Unit, University of Milan, IRCCS Foundation Ca’Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Chiara Simone
- Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, Dino Ferrari Centre, Neurology Unit, University of Milan, IRCCS Foundation Ca’Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Chiara Zanetta
- Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, Dino Ferrari Centre, Neurology Unit, University of Milan, IRCCS Foundation Ca’Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Nereo Bresolin
- Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, Dino Ferrari Centre, Neurology Unit, University of Milan, IRCCS Foundation Ca’Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Giacomo P. Comi
- Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, Dino Ferrari Centre, Neurology Unit, University of Milan, IRCCS Foundation Ca’Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation (DEPT), Neuroscience Section, Dino Ferrari Centre, Neurology Unit, University of Milan, IRCCS Foundation Ca’Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy
| |
Collapse
|
43
|
Ravits J, Appel S, Baloh RH, Barohn R, Brooks BR, Elman L, Floeter MK, Henderson C, Lomen-Hoerth C, Macklis JD, McCluskey L, Mitsumoto H, Przedborski S, Rothstein J, Trojanowski JQ, van den Berg LH, Ringel S. Deciphering amyotrophic lateral sclerosis: what phenotype, neuropathology and genetics are telling us about pathogenesis. Amyotroph Lateral Scler Frontotemporal Degener 2013; 14 Suppl 1:5-18. [PMID: 23678876 DOI: 10.3109/21678421.2013.778548] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized phenotypically by progressive weakness and neuropathologically by loss of motor neurons. Phenotypically, there is marked heterogeneity. Typical ALS has mixed upper motor neuron (UMN) and lower motor neuron (LMN) involvement. Primary lateral sclerosis has predominant UMN involvement. Progressive muscular atrophy has predominant LMN involvement. Bulbar and limb ALS have predominant regional involvement. Frontotemporal dementia has significant cognitive and behavioral involvement. These phenotypes can be so distinctive that they would seem to have differing biology. However, they cannot be distinguished, at least neuropathologically or genetically. In sporadic ALS (SALS), they are mostly characterized by ubiquitinated cytoplasmic inclusions of TDP-43. In familial ALS (FALS), where phenotypes are indistinguishable from SALS and similarly heterogeneous, each mutated gene has its own genetic and molecular signature. Overall, since the same phenotypes can have multiple causes including different gene mutations, there must be multiple molecular mechanisms causing ALS - and ALS is a syndrome. Since, however, multiple phenotypes can be caused by one single gene mutation, a single molecular mechanism can cause heterogeneity. What the mechanisms are remain unknown, but active propagation of the pathology neuroanatomically seems to be a principal component. Leading candidate mechanisms include RNA processing, cell-cell interactions between neurons and non-neuronal neighbors, focal seeding from a misfolded protein that has prion-like propagation, and fatal errors introduced during neurodevelopment of the motor system. If fundamental mechanisms could be identified and understood, ALS therapy could rationally target progression and stop the disease - a goal that seems increasingly achievable.
Collapse
Affiliation(s)
- John Ravits
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
D’Amico E, Factor-Litvak P, Santella RM, Mitsumoto H. Clinical perspective on oxidative stress in sporadic amyotrophic lateral sclerosis. Free Radic Biol Med 2013; 65:509-527. [PMID: 23797033 PMCID: PMC3859834 DOI: 10.1016/j.freeradbiomed.2013.06.029] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 06/14/2013] [Accepted: 06/14/2013] [Indexed: 12/12/2022]
Abstract
Sporadic amyotrophic lateral sclerosis (ALS) is one of the most devastating neurological diseases; most patients die within 3 to 4 years after symptom onset. Oxidative stress is a disturbance in the pro-oxidative/antioxidative balance favoring the pro-oxidative state. Autopsy and laboratory studies in ALS indicate that oxidative stress plays a major role in motor neuron degeneration and astrocyte dysfunction. Oxidative stress biomarkers in cerebrospinal fluid, plasma, and urine are elevated, suggesting that abnormal oxidative stress is generated outside of the central nervous system. Our review indicates that agricultural chemicals, heavy metals, military service, professional sports, excessive physical exertion, chronic head trauma, and certain foods might be modestly associated with ALS risk, with a stronger association between risk and smoking. At the cellular level, these factors are all involved in generating oxidative stress. Experimental studies indicate that a combination of insults that induce modest oxidative stress can exert additive deleterious effects on motor neurons, suggesting that multiple exposures in real-world environments are important. As the disease progresses, nutritional deficiency, cachexia, psychological stress, and impending respiratory failure may further increase oxidative stress. Moreover, accumulating evidence suggests that ALS is possibly a systemic disease. Laboratory, pathologic, and epidemiologic evidence clearly supports the hypothesis that oxidative stress is central in the pathogenic process, particularly in genetically susceptive individuals. If we are to improve ALS treatment, well-designed biochemical and genetic epidemiological studies, combined with a multidisciplinary research approach, are needed and will provide knowledge crucial to our understanding of ALS etiology, pathophysiology, and prognosis.
Collapse
Affiliation(s)
- Emanuele D’Amico
- Eleanor and Lou Gehrig MDA/ALS Research Center, The Neurological Institute of New York, Columbia University Medical Center, 710 West 168th Street (NI-9), New York, NY 10032, ;
| | - Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032,
| | - Regina M. Santella
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University Medical Center, 722 West 168th Street, New York, NY 10032,
| | - Hiroshi Mitsumoto
- Eleanor and Lou Gehrig MDA/ALS Research Center, The Neurological Institute of New York, Columbia University Medical Center, 710 West 168th Street (NI-9), New York, NY 10032
| |
Collapse
|
45
|
Jouroukhin Y, Ostritsky R, Assaf Y, Pelled G, Giladi E, Gozes I. NAP (davunetide) modifies disease progression in a mouse model of severe neurodegeneration: protection against impairments in axonal transport. Neurobiol Dis 2013; 56:79-94. [PMID: 23631872 DOI: 10.1016/j.nbd.2013.04.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 04/04/2013] [Accepted: 04/09/2013] [Indexed: 12/12/2022] Open
Abstract
NAP (davunetide) is a novel neuroprotective compound with mechanism of action that appears to involve microtubule (MT) stabilization and repair. To evaluate, for the first time, the impact of NAP on axonal transport in vivo and to translate it to neuroprotection in a severe neurodegeneration, the SOD1-G93A mouse model for amyotrophic lateral sclerosis (ALS) was used. Manganese-enhanced magnetic resonance imaging (MRI), estimating axonal transport rates, revealed a significant reduction of the anterograde axonal transport in the ALS mice compared to healthy control mice. Acute NAP treatment normalized axonal transport rates in these ALS mice. Tau hyperphosphorylation, associated with MT dysfunction and defective axonal transport, was discovered in the brains of the ALS mice and was significantly reduced by chronic NAP treatment. Furthermore, in healthy wild type (WT) mice, NAP reversed axonal transport disruption by colchicine, suggesting drug-dependent protection against axonal transport impairment through stabilization of the neuronal MT network. Histochemical analysis showed that chronic NAP treatment significantly protected spinal cord motor neurons against ALS-like pathology. Sequential MRI measurements, correlating brain structure with ALS disease progression, revealed a significant damage to the ventral tegmental area (VTA), indicative of impairments to the dopaminergic pathways relative to healthy controls. Chronic daily NAP treatment of the SOD1-G93A mice, initiated close to disease onset, delayed degeneration of the trigeminal, facial and hypoglossal motor nuclei as was significantly apparent at days 90-100 and further protected the VTA throughout life. Importantly, protection of the VTA was significantly correlated with longevity and overall, NAP treatment significantly prolonged life span in the ALS mice.
Collapse
Affiliation(s)
- Yan Jouroukhin
- Department of Human Molecular Genetics and Biochemistry, Sagol School of Neuroscience, Adams Super Center for Brain Studies, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | | | | | |
Collapse
|
46
|
Dobson-Stone C, Luty AA, Thompson EM, Blumbergs P, Brooks WS, Short CL, Field CD, Panegyres PK, Hecker J, Solski JA, Blair IP, Fullerton JM, Halliday GM, Schofield PR, Kwok JBJ. Frontotemporal dementia-amyotrophic lateral sclerosis syndrome locus on chromosome 16p12.1-q12.2: genetic, clinical and neuropathological analysis. Acta Neuropathol 2013; 125:523-33. [PMID: 23338750 PMCID: PMC3611035 DOI: 10.1007/s00401-013-1078-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 12/13/2022]
Abstract
Numerous families exhibiting both frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) have been described, and although many of these have been shown to harbour a repeat expansion in C9ORF72, several C9ORF72-negative FTD-ALS families remain. We performed neuropathological and genetic analysis of a large European Australian kindred (Aus-12) with autosomal dominant inheritance of dementia and/or ALS. Affected Aus-12 members developed either ALS or dementia; some of those with dementia also had ALS and/or extrapyramidal features. Neuropathology was most consistent with frontotemporal lobar degeneration with type B TDP pathology, but with additional phosphorylated tau pathology consistent with corticobasal degeneration. Aus-12 DNA samples were negative for mutations in all known dementia and ALS genes, including C9ORF72 and FUS. Genome-wide linkage analysis provided highly suggestive evidence (maximum multipoint LOD score of 2.9) of a locus on chromosome 16p12.1-16q12.2. Affected individuals shared a chromosome 16 haplotype flanked by D16S3103 and D16S489, spanning 37.9 Mb, with a smaller suggestive disease haplotype spanning 24.4 Mb defined by recombination in an elderly unaffected individual. Importantly, this smaller region does not overlap with FUS. Whole-exome sequencing identified four variants present in the maximal critical region that segregate with disease. Linkage analysis incorporating these variants generated a maximum multipoint LOD score of 3.0. These results support the identification of a locus on chromosome 16p12.1-16q12.2 responsible for an unusual cluster of neurodegenerative phenotypes. This region overlaps with a separate locus on 16q12.1-q12.2 reported in an independent ALS family, indicating that this region may harbour a second major locus for FTD-ALS.
Collapse
Affiliation(s)
- Carol Dobson-Stone
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW 2031 Australia
- University of New South Wales, Sydney, Australia
| | - Agnes A. Luty
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW 2031 Australia
- University of New South Wales, Sydney, Australia
| | - Elizabeth M. Thompson
- SA Clinical Genetics Service, SA Pathology, Women’s and Children’s Hospital, Adelaide, Australia
- Department of Paediatrics, University of Adelaide, North Terrace, Adelaide, Australia
| | - Peter Blumbergs
- Institute of Medical and Veterinary Science, Adelaide, Australia
| | - William S. Brooks
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW 2031 Australia
- University of New South Wales, Sydney, Australia
| | - Cathy L. Short
- Department of Neurology, The Queen Elizabeth Hospital, Woodville, Adelaide, Australia
| | - Colin D. Field
- Division of Rehabilitation and Aged Care, Memory Clinic, Repatriation General Hospital, Daw Park, Adelaide, Australia
- Present Address: Adelaide Dementia Driving Clinic, North Adelaide, Australia
| | | | - Jane Hecker
- Department of General Medicine, Royal Adelaide Hospital, Adelaide, Australia
| | - Jennifer A. Solski
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord Hospital, Sydney, Australia
| | - Ian P. Blair
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord Hospital, Sydney, Australia
| | - Janice M. Fullerton
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW 2031 Australia
- University of New South Wales, Sydney, Australia
| | - Glenda M. Halliday
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW 2031 Australia
- University of New South Wales, Sydney, Australia
| | - Peter R. Schofield
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW 2031 Australia
- University of New South Wales, Sydney, Australia
| | - John B. J. Kwok
- Neuroscience Research Australia, Barker St, Randwick, Sydney, NSW 2031 Australia
- University of New South Wales, Sydney, Australia
| |
Collapse
|
47
|
Titler AM, Posimo JM, Leak RK. Astrocyte plasticity revealed by adaptations to severe proteotoxic stress. Cell Tissue Res 2013; 352:427-43. [PMID: 23420451 DOI: 10.1007/s00441-013-1571-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 01/17/2013] [Indexed: 12/21/2022]
Abstract
Neurodegeneration is characterized by an accumulation of misfolded proteins in neurons. It is less well appreciated that glia often also accumulate misfolded proteins. However, glia are highly plastic and may adapt to stress readily. Endogenous adaptations to stress can be measured by challenging stressed cells with a second hit and then measuring viability. For example, subtoxic stress can elicit preconditioning or tolerance against second hits. However, it is not known if severe stress that kills half the population can elicit endogenous adaptations in the remaining survivors. Glia, with their resilient nature, offer an ideal model in which to test this new hypothesis. The present study is the first demonstration that astrocytes surviving one LC50 hit of the proteasome inhibitor MG132 were protected against a second MG132 hit. ATP loss in response to the second hit was also prevented. MG132 caused compensatory rises in stress-sensitive heat shock proteins. However, stressed astrocytes exhibited an even greater rise in ubiquitin-conjugated proteins upon the second hit, illustrating the severity of the proteotoxicity and verifying the continued impact of MG132. Despite this stress, MG132-pretreated astrocytes were completely prevented from losing glutathione with the second hit. Furthermore, inhibiting glutathione synthesis rendered astrocytes sensitive to the second hit, unmasking the cumulative impact of two hits by removal of an endogenous adaptation. These findings suggest that stressed astrocytes become progressively harder to kill by virtue of antioxidant defenses. Such plasticity may permit astrocytes under severe stress to better support neurons and help explain the protracted nature of neurodegeneration.
Collapse
Affiliation(s)
- Amanda M Titler
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA 15282, USA
| | | | | |
Collapse
|