101
|
Buckland ME, Affleck AJ, Pearce AJ, Suter CM. Chronic Traumatic Encephalopathy as a Preventable Environmental Disease. Front Neurol 2022; 13:880905. [PMID: 35769361 PMCID: PMC9234108 DOI: 10.3389/fneur.2022.880905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022] Open
Abstract
In this Perspective we explore the evolution of our understanding of chronic traumatic encephalopathy (CTE) and its relationship with repetitive head injury. As with many neurodegenerative conditions, there is an imperfect correspondence between neuropathology and clinical phenotype, but unlike other neurodegenerative diseases, CTE has a discrete and easily modifiable risk factor: exposure to repetitive head injury. Consequently, evaluation of the evidence regarding exposure to repetitive head injury and CTE risk should be undertaken using public or occupational health frameworks of medical knowledge. The current debate over the existence of CTE as a disease of concern is fuelled in part by immediate medico-legal considerations, and the involvement of high-profile athletes, with inevitable media interest. Moving beyond this debate has significant potential to address and reduce disease impact in the near future, and provide novel insights into mechanisms underlying abnormal protein accumulation in CTE and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Michael E. Buckland
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
- *Correspondence: Michael E. Buckland
| | - Andrew J. Affleck
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Alan J. Pearce
- College of Science, Health and Engineering, La Trobe University, Bundoora, VIC, Australia
| | - Catherine M. Suter
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
102
|
Capano LS, Sato C, Ficulle E, Yu A, Horie K, Kwon JS, Burbach KF, Barthélemy NR, Fox SG, Karch CM, Bateman RJ, Houlden H, Morimoto RI, Holtzman DM, Duff KE, Yoo AS. Recapitulation of endogenous 4R tau expression and formation of insoluble tau in directly reprogrammed human neurons. Cell Stem Cell 2022; 29:918-932.e8. [PMID: 35659876 PMCID: PMC9176216 DOI: 10.1016/j.stem.2022.04.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/21/2022] [Accepted: 04/27/2022] [Indexed: 01/07/2023]
Abstract
Tau is a microtubule-binding protein expressed in neurons, and the equal ratios between 4-repeat (4R) and 3-repeat (3R) isoforms are maintained in normal adult brain function. Dysregulation of 3R:4R ratio causes tauopathy, and human neurons that recapitulate tau isoforms in health and disease will provide a platform for elucidating pathogenic processes involving tau pathology. We carried out extensive characterizations of tau isoforms expressed in human neurons derived by microRNA-induced neuronal reprogramming of adult fibroblasts. Transcript and protein analyses showed that miR neurons expressed all six isoforms with the 3R:4R isoform ratio equivalent to that detected in human adult brains. Also, miR neurons derived from familial tauopathy patients with a 3R:4R ratio altering mutation showed increased 4R tau and the formation of insoluble tau with seeding activity. Our results collectively demonstrate the utility of miRNA-induced neuronal reprogramming to recapitulate endogenous tau regulation comparable with the adult brain in health and disease.
Collapse
Affiliation(s)
- Lucia S Capano
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular and Cell Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chihiro Sato
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Elena Ficulle
- UK Dementia Research Institute at University College London, London WC1E 6BT, UK
| | - Anan Yu
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208, USA
| | - Kanta Horie
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ji-Sun Kwon
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Computational and Systems Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kyle F Burbach
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan G Fox
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208, USA
| | - Celeste M Karch
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Knight ADRC, St. Louis, MO 63110, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Knight ADRC, St. Louis, MO 63110, USA
| | - Henry Houlden
- UK Dementia Research Institute at University College London, London WC1E 6BT, UK
| | - Richard I Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Knight ADRC, St. Louis, MO 63110, USA
| | - Karen E Duff
- UK Dementia Research Institute at University College London, London WC1E 6BT, UK.
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Knight ADRC, St. Louis, MO 63110, USA.
| |
Collapse
|
103
|
Li D, Liu C. Conformational strains of pathogenic amyloid proteins in neurodegenerative diseases. Nat Rev Neurosci 2022; 23:523-534. [DOI: 10.1038/s41583-022-00603-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2022] [Indexed: 12/11/2022]
|
104
|
Downregulation of glob1 mitigates human tau mediated neurotoxicity by restricting heterochromatin loss and elevating the autophagic response in drosophila. Mol Biol Rep 2022; 49:6581-6590. [PMID: 35633418 DOI: 10.1007/s11033-022-07498-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/21/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Human neuronal tauopathies are typically characterized by the accumulation of hyperphosphorylated tau in the forms of paired helical filaments and/or neurofibrillary tangles in the brain neurons. Tau-mediated heterochromatin loss and subsequent global transcriptional upsurge have been demonstrated as one of the key factors that promotes tau toxicity. We have reported earlier that expression of human tau-transgene in Drosophila induces the expression of glob1, and its restored level restricts tau etiology by regulating tau hyperphosphorylation and ROS generation via GSK-3β/p-Akt and Nrf2-keap1-ARE pathways, respectively. In view of this noted capability of glob1 in regulation of oxidative stress, and involvement of ROS in chromatin remodeling; we investigate if downregulation of glob1 restores tau-mediated heterochromatin loss in order to alleviate neurotoxicity. METHODS AND RESULTS The tauV337M transgene was expressed in Drosophila eye by utilizing GAL4/UAS system. Expression of glob1 was depleted in tauV337M expressing tissues by co-expressing an UAS-glob1RNAi transgene by GMR-Gal4 driver. Immunostaining and wstern blot analysis suggested that tissue-specific downregulation of glob1 restores the cellular level of CBP and minimizes tau-mediated heterochromatin loss. It also assists in mounting an improved protective autophagic response to alleviate the human tau-induced neurotoxicity in Drosophila tauopathy models. CONCLUSIONS Our study unfolds a novel aspect of the multitasking globin protein in restricting the pathogenesis of neuronal tauopathies. Interestingly, due to notable similarities between Drosophila glob1 and human globin gene(s), our findings may be helpful in developing novel therapeutic approaches against tauopathies.
Collapse
|
105
|
Sivanantharajah L, Mudher A. Curcumin as a Holistic Treatment for Tau Pathology. Front Pharmacol 2022; 13:903119. [PMID: 35662729 PMCID: PMC9160965 DOI: 10.3389/fphar.2022.903119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/02/2022] [Indexed: 12/12/2022] Open
Abstract
Global forecasts for prevalence of Alzheimer’s Disease (AD) estimate that 152.8 million people will have dementia in 2050, a sharp rise from 57.4 million in 2019 (GBD 2019). This rise can be attributable to increases in population growth and aging, but in the absence of disease-modifying therapies it poses a huge societal challenge that must be addressed urgently. One way to combat this challenge is to explore the utility of holistic treatments that may protect against AD, including traditional herbs, spices and other nutraceuticals that are pharmacologically safe, inexpensive and readily available. In this light, the spice turmeric, and its active ingredient curcumin, has been investigated as a potential holistic treatment for AD over the past 2 decades; however, promising results with animal studies have not translated to success in clinical trials. One issue is that most animal models examining the effects of curcumin and curcumin derivatives in AD have been done with a focus at ameliorating amyloid pathology. Due to the limited success of Amyloid-β-based drugs in recent clinical trials, tau-focused therapeutics provide a promising alternative. In this article, we aim to provide a clearer picture of what is currently known about the effectiveness of curcumin and curcumin derivatives to ameliorate tau pathology. Tau focused studies may help inform more successful clinical studies by placing greater emphasis on the development and optimised delivery of curcumin derivatives that more effectively target tau pathology.
Collapse
Affiliation(s)
- Lovesha Sivanantharajah
- School of Natural Sciences, Bangor University, Bangor Gwynedd, United Kingdom
- *Correspondence: Lovesha Sivanantharajah,
| | - Amritpal Mudher
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
106
|
Shao E, Chang CW, Li Z, Yu X, Ho K, Zhang M, Wang X, Simms J, Lo I, Speckart J, Holtzman J, Yu GQ, Roberson ED, Mucke L. TAU ablation in excitatory neurons and postnatal TAU knockdown reduce epilepsy, SUDEP, and autism behaviors in a Dravet syndrome model. Sci Transl Med 2022; 14:eabm5527. [PMID: 35476595 PMCID: PMC9102397 DOI: 10.1126/scitranslmed.abm5527] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intracellular accumulation of TAU aggregates is a hallmark of several neurodegenerative diseases. However, global genetic reduction of TAU is beneficial also in models of other brain disorders that lack such TAU pathology, suggesting a pathogenic role of nonaggregated TAU. Here, conditional ablation of TAU in excitatory, but not inhibitory, neurons reduced epilepsy, sudden unexpected death in epilepsy, overactivation of the phosphoinositide 3-kinase-AKT-mammalian target of rapamycin pathway, brain overgrowth (megalencephaly), and autism-like behaviors in a mouse model of Dravet syndrome, a severe epileptic encephalopathy of early childhood. Furthermore, treatment with a TAU-lowering antisense oligonucleotide, initiated on postnatal day 10, had similar therapeutic effects in this mouse model. Our findings suggest that excitatory neurons are the critical cell type in which TAU has to be reduced to counteract brain dysfunctions associated with Dravet syndrome and that overall cerebral TAU reduction could have similar benefits, even when initiated postnatally.
Collapse
Affiliation(s)
- Eric Shao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Che-Wei Chang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Zhiyong Li
- Alzheimer’s Disease Center, Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Xinxing Yu
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Kaitlyn Ho
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Michelle Zhang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Xin Wang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Jeffrey Simms
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Iris Lo
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Jessica Speckart
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Julia Holtzman
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
| | - Erik D. Roberson
- Alzheimer’s Disease Center, Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158 USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158 USA
| |
Collapse
|
107
|
Alzheimer’s Disease and Tau Self-Assembly: In the Search of the Missing Link. Int J Mol Sci 2022; 23:ijms23084192. [PMID: 35457009 PMCID: PMC9032712 DOI: 10.3390/ijms23084192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial neurodegenerative disease characterized by progressive cognitive impairment, apathy, and neuropsychiatric disorders. Two main pathological hallmarks have been described: neurofibrillary tangles, consisting of tau oligomers (hyperphosphorylated tau) and Aβ plaques. The influence of protein kinases and phosphatases on the hyperphosphorylation of tau is already known. Hyperphosphorylated tau undergoes conformational changes that promote its self-assembly. However, the process involving these mechanisms is yet to be elucidated. In vitro recombinant tau can be aggregated by the action of polyanions, such as heparin, arachidonic acid, and more recently, the action of polyphosphates. However, how that process occurs in vivo is yet to be understood. In this review, searching the most accurate and updated literature on the matter, we focus on the precise molecular events linking tau modifications, its misfolding and the initiation of its pathological self-assembly. Among these, we can identify challenges regarding tau phosphorylation, the link between tau heteroarylations and the onset of its self-assembly, as well as the possible metabolic pathways involving natural polyphosphates, that may play a role in tau self-assembly.
Collapse
|
108
|
Zhang Y, Wu KM, Yang L, Dong Q, Yu JT. Tauopathies: new perspectives and challenges. Mol Neurodegener 2022; 17:28. [PMID: 35392986 PMCID: PMC8991707 DOI: 10.1186/s13024-022-00533-z] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/23/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tauopathies are a class of neurodegenerative disorders characterized by neuronal and/or glial tau-positive inclusions. MAIN BODY Clinically, tauopathies can present with a range of phenotypes that include cognitive/behavioral-disorders, movement disorders, language disorders and non-specific amnestic symptoms in advanced age. Pathologically, tauopathies can be classified based on the predominant tau isoforms that are present in the inclusion bodies (i.e., 3R, 4R or equal 3R:4R ratio). Imaging, cerebrospinal fluid (CSF) and blood-based tau biomarkers have the potential to be used as a routine diagnostic strategy and in the evaluation of patients with tauopathies. As tauopathies are strongly linked neuropathologically and genetically to tau protein abnormalities, there is a growing interest in pursuing of tau-directed therapeutics for the disorders. Here we synthesize emerging lessons on tauopathies from clinical, pathological, genetic, and experimental studies toward a unified concept of these disorders that may accelerate the therapeutics. CONCLUSIONS Since tauopathies are still untreatable diseases, efforts have been made to depict clinical and pathological characteristics, identify biomarkers, elucidate underlying pathogenesis to achieve early diagnosis and develop disease-modifying therapies.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Kai-Min Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Liu Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, National Center for Neurological Disorders, 12th Wulumuqi Zhong Road, Shanghai, 200040 China
| |
Collapse
|
109
|
Kow RL, Black AH, Saxton AD, Liachko NF, Kraemer BC. Loss of aly/ALYREF suppresses toxicity in both tau and TDP-43 models of neurodegeneration. GeroScience 2022; 44:747-761. [PMID: 35122183 PMCID: PMC9135935 DOI: 10.1007/s11357-022-00526-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/27/2022] [Indexed: 11/04/2022] Open
Abstract
Neurodegenerative diseases with tau pathology, or tauopathies, include Alzheimer's disease and related dementia disorders. Previous work has shown that loss of the poly(A) RNA-binding protein gene sut-2/MSUT2 strongly suppressed tauopathy in Caenorhabditis elegans, human cell culture, and mouse models of tauopathy. However, the mechanism of suppression is still unclear. Recent work has shown that MSUT2 protein interacts with the THO complex and ALYREF, which are components of the mRNA nuclear export complex. Additionally, previous work showed ALYREF homolog Ref1 modulates TDP-43 and G4C2 toxicity in Drosophila melanogaster models. We used transgenic C. elegans models of tau or TDP-43 toxicity to investigate the effects of loss of ALYREF function on tau and TDP-43 toxicity. In C. elegans, three genes are homologous to human ALYREF: aly-1, aly-2, and aly-3. We found that loss of C. elegans aly gene function, especially loss of both aly-2 and aly-3, suppressed tau-induced toxic phenotypes. Loss of aly-2 and aly-3 was also able to suppress TDP-43-induced locomotor behavior deficits. However, loss of aly-2 and aly-3 had divergent effects on mRNA and protein levels as total tau protein levels were reduced while mRNA levels were increased, but no significant effects were seen on total TDP-43 protein or mRNA levels. Our results suggest that although aly genes modulate both tau and TDP-43-induced toxicity phenotypes, the molecular mechanisms of suppression are different and separated from impacts on mRNA and protein levels. Altogether, this study highlights the importance of elucidating RNA-related mechanisms in both tau and TDP-43-induced toxicity.
Collapse
Affiliation(s)
- Rebecca L Kow
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, S182, 1660 South Columbian Way, Seattle, WA, 98108, USA.
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, 98104, USA.
| | - Aristide H Black
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, S182, 1660 South Columbian Way, Seattle, WA, 98108, USA
| | - Aleen D Saxton
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, S182, 1660 South Columbian Way, Seattle, WA, 98108, USA
| | - Nicole F Liachko
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, S182, 1660 South Columbian Way, Seattle, WA, 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, 98104, USA
| | - Brian C Kraemer
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, S182, 1660 South Columbian Way, Seattle, WA, 98108, USA.
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, 98104, USA.
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, 98195, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
110
|
Sengupta U, Kayed R. Amyloid β, Tau, and α-Synuclein aggregates in the pathogenesis, prognosis, and therapeutics for neurodegenerative diseases. Prog Neurobiol 2022; 214:102270. [DOI: 10.1016/j.pneurobio.2022.102270] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/28/2022] [Accepted: 04/13/2022] [Indexed: 12/11/2022]
|
111
|
Cellular Senescence and Aging in Myotonic Dystrophy. Int J Mol Sci 2022; 23:ijms23042339. [PMID: 35216455 PMCID: PMC8877951 DOI: 10.3390/ijms23042339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/06/2022] [Accepted: 02/12/2022] [Indexed: 01/10/2023] Open
Abstract
Myotonic dystrophy (DM) is a dominantly inherited multisystemic disorder affecting various organs, such as skeletal muscle, heart, the nervous system, and the eye. Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are caused by expanded CTG and CCTG repeats, respectively. In both forms, the mutant transcripts containing expanded repeats aggregate as nuclear foci and sequester several RNA-binding proteins, resulting in alternative splicing dysregulation. Although certain alternative splicing events are linked to the clinical DM phenotypes, the molecular mechanisms underlying multiple DM symptoms remain unclear. Interestingly, multi-systemic DM manifestations, including muscle weakness, cognitive impairment, cataract, and frontal baldness, resemble premature aging. Furthermore, cellular senescence, a critical contributor to aging, is suggested to play a key role in DM cellular pathophysiology. In particular, several senescence inducers including telomere shortening, mitochondrial dysfunction, and oxidative stress and senescence biomarkers such as cell cycle inhibitors, senescence-associated secretory phenotype, chromatin reorganization, and microRNA have been implicated in DM pathogenesis. In this review, we focus on the clinical similarities between DM and aging, and summarize the involvement of cellular senescence in DM and the potential application of anti-aging DM therapies.
Collapse
|
112
|
Pang K, Jiang R, Zhang W, Yang Z, Li LL, Shimozawa M, Tambaro S, Mayer J, Zhang B, Li M, Wang J, Liu H, Yang A, Chen X, Liu J, Winblad B, Han H, Jiang T, Wang W, Nilsson P, Guo W, Lu B. An App knock-in rat model for Alzheimer's disease exhibiting Aβ and tau pathologies, neuronal death and cognitive impairments. Cell Res 2022; 32:157-175. [PMID: 34789895 PMCID: PMC8807612 DOI: 10.1038/s41422-021-00582-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 10/04/2021] [Indexed: 12/21/2022] Open
Abstract
A major obstacle in Alzheimer's disease (AD) research is the lack of predictive and translatable animal models that reflect disease progression and drug efficacy. Transgenic mice overexpressing amyloid precursor protein (App) gene manifest non-physiological and ectopic expression of APP and its fragments in the brain, which is not observed in AD patients. The App knock-in mice circumvented some of these problems, but they do not exhibit tau pathology and neuronal death. We have generated a rat model, with three familiar App mutations and humanized Aβ sequence knocked into the rat App gene. Without altering the levels of full-length APP and other APP fragments, this model exhibits pathologies and disease progression resembling those in human patients: deposit of Aβ plaques in relevant brain regions, microglia activation and gliosis, progressive synaptic degeneration and AD-relevant cognitive deficits. Interestingly, we have observed tau pathology, neuronal apoptosis and necroptosis and brain atrophy, phenotypes rarely seen in other APP models. This App knock-in rat model may serve as a useful tool for AD research, identifying new drug targets and biomarkers, and testing therapeutics.
Collapse
Affiliation(s)
- Keliang Pang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Richeng Jiang
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology Head and Neck Surgery, The First Hospital of Jilin University, Changchun, China
| | - Wei Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Zhengyi Yang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Lin-Lin Li
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Makoto Shimozawa
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Johanna Mayer
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Baogui Zhang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Man Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Jiesi Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Hang Liu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Ailing Yang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xi Chen
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiazheng Liu
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Hua Han
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Tianzi Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Weiwen Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Wei Guo
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China.
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China.
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China.
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China.
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
113
|
Venkatramani A, Mukherjee S, Kumari A, Panda D. Shikonin impedes phase separation and aggregation of tau and protects SH-SY5Y cells from the toxic effects of tau oligomers. Int J Biol Macromol 2022; 204:19-33. [PMID: 35120943 DOI: 10.1016/j.ijbiomac.2022.01.172] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/20/2022] [Accepted: 01/28/2022] [Indexed: 12/22/2022]
Abstract
Tauopathies such as Alzheimer's and Parkinson's diseases involve the abnormal deposition of tau aggregates in the brain and neuronal tissues. We report that a natural naphthoquinone, shikonin, impeded the oligomerization and fibrillization of tau. The compound strongly inhibited heparin, arachidonic acid, and RNA-induced tau aggregation. Atomic force microscopy, dynamic light scattering, SDS-PAGE, and dot blot assays revealed that shikonin diminished tau oligomerization and decreased the mean size of tau oligomers. Transmission electron microscopy and atomic force microscopy analysis further showed that shikonin could suppress tau fibrillization and shorten the tau filaments. Shikonin inhibited tau droplet formation. The compound significantly reduced the aggregation rate of a tryptophan mutant (Y310W-tau) of tau. In addition, shikonin disaggregated preformed tau filaments with a half-maximal disaggregation concentration (DC50) of 6.3 ± 0.4 μM. Pre-treatment of neuroblastoma cells (SH-SY5Y) with shikonin protected the cells from the toxicity induced by tau oligomers and increased their viability. The findings imply that shikonin inhibited several steps in the tau aggregation pathways, especially the early stages, such as liquid-liquid phase separation. Therefore, shikonin is an attractive candidate for developing a therapy against tauopathy.
Collapse
Affiliation(s)
- Anuradha Venkatramani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Sandipan Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Anuradha Kumari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS. Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
114
|
Vallés-Saiz L, Peinado-Cahuchola R, Ávila J, Hernández F. Microtubule-associated protein tau in murine kidney: role in podocyte architecture. Cell Mol Life Sci 2022; 79:97. [PMID: 35084555 PMCID: PMC8794918 DOI: 10.1007/s00018-021-04106-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/29/2021] [Accepted: 12/19/2021] [Indexed: 12/25/2022]
Abstract
Tau is a cytoskeletal protein that is expressed mainly in neurons and is involved in several cellular processes, such as microtubule stabilization, axonal maintenance, and transport. Altered tau metabolism is related to different tauopathies being the most important Alzheimer's disease where aberrant hyperphosphorylated and aggregated tau is found in the central nervous system. Here, we have analyzed that function in kidney by using tau knockout mice generated by integrating GFP-encoding cDNA into exon 1 of MAPT (here referred to as TauGFP/GFP). IVIS Lumina from PerkinElmer demonstrated GFP expression in the kidney. We then demonstrated by qPCR that the main tau isoform in the kidney is Tau4R. The GFP reporter allowed us to demonstrate that tau is found in the glomeruli of the renal cortex, and specifically in podocytes. This was further confirmed by immunohistochemistry. TauGFP/GFP mice present a podocyte cytoskeleton more dynamic as they contain higher levels of detyrosinated tubulin than wild-type mice. In addition, transmission electron microscopy studies demonstrated glomerular damage with a decrease in urinary creatinine. Our results prove that tau has an important role in kidney metabolism under normal physiological conditions.
Collapse
Affiliation(s)
- Laura Vallés-Saiz
- Centro de Biología Molecular "Severo Ochoa", CSIC/UAM, Universidad Autónoma de Madrid, Cantoblanco, 28049, Madrid, Spain
| | - Rocio Peinado-Cahuchola
- Centro de Biología Molecular "Severo Ochoa", CSIC/UAM, Universidad Autónoma de Madrid, Cantoblanco, 28049, Madrid, Spain
| | - Jesús Ávila
- Centro de Biología Molecular "Severo Ochoa", CSIC/UAM, Universidad Autónoma de Madrid, Cantoblanco, 28049, Madrid, Spain
| | - Félix Hernández
- Centro de Biología Molecular "Severo Ochoa", CSIC/UAM, Universidad Autónoma de Madrid, Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
115
|
Jiang XJ, Wu YQ, Ma R, Chang YM, Li LL, Zhu JH, Liu GP, Li G. PINK1 Alleviates Cognitive Impairments via Attenuating Pathological Tau Aggregation in a Mouse Model of Tauopathy. Front Cell Dev Biol 2022; 9:736267. [PMID: 35059394 PMCID: PMC8763800 DOI: 10.3389/fcell.2021.736267] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/25/2021] [Indexed: 01/18/2023] Open
Abstract
As a primary cause of dementia and death in older people, Alzheimer’s disease (AD) has become a common problem and challenge worldwide. Abnormal accumulation of tau proteins in the brain is a hallmark pathology of AD and is closely related to the clinical progression and severity of cognitive deficits. Here, we found that overexpression of phosphatase and tensin homolog (PTEN)-induced kinase 1 (PINK1) effectively promoted the degradation of tau, thereby rescuing neuron loss, synaptic damage, and cognitive impairments in a mouse model of tauopathy with AAV-full-length human Tau (hTau) injected into the hippocampal CA1 area (hTau mice). Overexpression of PINK1 activated autophagy, and chloroquine but not MG132 reversed the PINK1-induced decrease in human Tau levels and cognitive improvement in hTau mice. Furthermore, PINK1 also ameliorated mitochondrial dysfunction induced by hTau. Taken together, our data revealed that PINK1 overexpression promoted degradation of abnormal accumulated tau via the autophagy–lysosome pathway, indicating that PINK1 may be a potential target for AD treatment.
Collapse
Affiliation(s)
- Xing Jun Jiang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Qing Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Ma
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Min Chang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Lu Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Hui Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong Ping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Gang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
116
|
Kovacs GG, Ghetti B, Goedert M. Classification of Diseases with Accumulation of Tau Protein. Neuropathol Appl Neurobiol 2022; 48:e12792. [PMID: 35064600 PMCID: PMC9352145 DOI: 10.1111/nan.12792] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 01/07/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Laboratory Medicine Program & Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indiana, USA
| | - Michel Goedert
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, UK
| |
Collapse
|
117
|
Chen C, Lu J, Peng W, Mak MS, Yang Y, Zhu Z, Wang S, Hou J, Zhou X, Xin W, Hu Y, Tsim KWK, Han Y, Liu Q, Pi R. Acrolein, an endogenous aldehyde induces Alzheimer's disease-like pathologies in mice: A new sporadic AD animal model. Pharmacol Res 2022; 175:106003. [PMID: 34838693 DOI: 10.1016/j.phrs.2021.106003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/01/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that mainly affects elderly people. However, the translational research of AD is frustrating, suggesting that the development of new AD animal models is crucial. By gavage administration of acrolein, we constructed a simple sporadic AD animal model which showed classic pathologies of AD in 1 month. The AD-like phenotypes and pathological changes were as followed. 1) olfactory dysfunctions, cognitive impairments and psychological symptoms in C57BL/6 mice; 2) increased levels of Aβ1-42 and Tau phosphorylation (S396/T231) in cortex and hippocampus; 3) astrocytes and microglia proliferation; 4) reduced levels of postsynaptic density 95(PSD95) and Synapsin1, as well as the density of dendritic spines in the CA1 and DG neurons of the hippocampus; 5) high-frequency stimulation failed to induce the long-term potentiation (LTP) in the hippocampus after exposure to acrolein for 4 weeks; 6) decreased blood oxygen level-dependent (BOLD) signal in the olfactory bulb and induced high T2 signals in the hippocampus, which matched to the clinical observation in the brain of AD patients, and 7) activated RhoA/ROCK2/ p-cofilin-associated pathway in hippocampus of acrolein-treated mice, which may be the causes of synaptic damage and neuroinflammation in acrolein mice model. Taken together, the acrolein-induced sporadic AD mouse model closely reflects the pathological features of AD, which will be useful for the research on the mechanism of AD onset and the development of anti-AD drugs.
Collapse
Affiliation(s)
- Chen Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Junfeng Lu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Weijia Peng
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Marvin Sh Mak
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yang Yang
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zeyu Zhu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Shuyi Wang
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Jiawei Hou
- Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Xin Zhou
- Zhongshan school of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wenjun Xin
- Zhongshan school of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510014, China
| | - Karl Wah Keung Tsim
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hong Kong, China
| | - Qinyu Liu
- The seventh affiliated hospital, Sun Yat-Sen University, Shenzhen 518107, China.
| | - Rongbiao Pi
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangzhou, Guangzhou 510006, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China.
| |
Collapse
|
118
|
Petrozziello T, Amaral AC, Dujardin S, Farhan SMK, Chan J, Trombetta BA, Kivisäkk P, Mills AN, Bordt EA, Kim SE, Dooley PM, Commins C, Connors TR, Oakley DH, Ghosal A, Gomez-Isla T, Hyman BT, Arnold SE, Spires-Jones T, Cudkowicz ME, Berry JD, Sadri-Vakili G. Novel genetic variants in MAPT and alterations in tau phosphorylation in amyotrophic lateral sclerosis post-mortem motor cortex and cerebrospinal fluid. Brain Pathol 2021; 32:e13035. [PMID: 34779076 PMCID: PMC8877756 DOI: 10.1111/bpa.13035] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/22/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Although the molecular mechanisms underlying amyotrophic lateral sclerosis (ALS) are not yet fully understood, several studies report alterations in tau phosphorylation in both sporadic and familial ALS. Recently, we have demonstrated that phosphorylated tau at S396 (pTau‐S396) is mislocalized to synapses in ALS motor cortex (mCTX) and contributes to mitochondrial dysfunction. Here, we demonstrate that while there was no overall increase in total tau, pTau‐S396, and pTau‐S404 in ALS post‐mortem mCTX, total tau and pTau‐S396 were increased in C9ORF72‐ALS. Additionally, there was a significant decrease in pTau‐T181 in ALS mCTX compared controls. Furthermore, we leveraged the ALS Knowledge Portal and Project MinE data sets and identified ALS‐specific genetic variants across MAPT, the gene encoding tau. Lastly, assessment of cerebrospinal fluid (CSF) samples revealed a significant increase in total tau levels in bulbar‐onset ALS together with a decrease in CSF pTau‐T181:tau ratio in all ALS samples, as reported previously. While increases in CSF tau levels correlated with a faster disease progression as measured by the revised ALS functional rating scale (ALSFRS‐R), decreases in CSF pTau‐T181:tau ratio correlated with a slower disease progression, suggesting that CSF total tau and pTau‐T181 ratio may serve as biomarkers of disease in ALS. Our findings highlight the potential role of pTau‐T181 in ALS, as decreases in CSF pTau‐T181:tau ratio may reflect the significant decrease in pTau‐T181 in post‐mortem mCTX. Taken together, these results indicate that tau phosphorylation is altered in ALS post‐mortem mCTX as well as in CSF and, importantly, the newly described pathogenic or likely pathogenic variants identified in MAPT in this study are adjacent to T181 and S396 phosphorylation sites further highlighting the potential role of these tau functional domains in ALS. Although the molecular mechanisms underlying amyotrophic lateral sclerosis (ALS) are not yet fully understood, recent studies report alterations in tau phosphorylation in ALS. Our study builds on these findings and demonstrates that tau phosphorylation is altered in post‐mortem ALS motor cortex and highlights new and ALS‐specific variants in MAPT, the gene encoding tau. Lastly, we report alterations in phosphorylated tau in ALS cerebrospinal fluid that may function as a predictive biomarker for ALS.![]()
Collapse
Affiliation(s)
- Tiziana Petrozziello
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ana C Amaral
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Simon Dujardin
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sali M K Farhan
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - James Chan
- Biostatistics Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bianca A Trombetta
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pia Kivisäkk
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexandra N Mills
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evan A Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Spencer E Kim
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Patrick M Dooley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Caitlin Commins
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Theresa R Connors
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Derek H Oakley
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anubrata Ghosal
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Teresa Gomez-Isla
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Steven E Arnold
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tara Spires-Jones
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, University of Edinburgh, UK
| | - Merit E Cudkowicz
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James D Berry
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ghazaleh Sadri-Vakili
- Sean M. Healey & AMG Center for ALS at Mass General, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
119
|
Zimmer-Bensch G, Zempel H. DNA Methylation in Genetic and Sporadic Forms of Neurodegeneration: Lessons from Alzheimer's, Related Tauopathies and Genetic Tauopathies. Cells 2021; 10:3064. [PMID: 34831288 PMCID: PMC8624300 DOI: 10.3390/cells10113064] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/14/2022] Open
Abstract
Genetic and sporadic forms of tauopathies, the most prevalent of which is Alzheimer's Disease, are a scourge of the aging society, and in the case of genetic forms, can also affect children and young adults. All tauopathies share ectopic expression, mislocalization, or aggregation of the microtubule associated protein TAU, encoded by the MAPT gene. As TAU is a neuronal protein widely expressed in the CNS, the overwhelming majority of tauopathies are neurological disorders. They are characterized by cognitive dysfunction often leading to dementia, and are frequently accompanied by movement abnormalities such as parkinsonism. Tauopathies can lead to severe neurological deficits and premature death. For some tauopathies there is a clear genetic cause and/or an epigenetic contribution. However, for several others the disease etiology is unclear, with few tauopathies being environmentally triggered. Here, we review current knowledge of tauopathies listing known genetic and important sporadic forms of these disease. Further, we discuss how DNA methylation as a major epigenetic mechanism emerges to be involved in the disease pathophysiology of Alzheimer's, and related genetic and non-genetic tauopathies. Finally, we debate the application of epigenetic signatures in peripheral blood samples as diagnostic tools and usages of epigenetic therapy strategies for these diseases.
Collapse
Affiliation(s)
- Geraldine Zimmer-Bensch
- Functional Epigenetics in the Animal Model, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany
- Research Training Group 2416 MultiSenses-MultiScales, Institute for Biology II, RWTH Aachen University, 52074 Aachen, Germany
| | - Hans Zempel
- Institute of Human Genetics, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
120
|
Longitudinal Assessment of Tau-Associated Pathology by 18F-THK5351 PET Imaging: A Histological, Biochemical, and Behavioral Study. Diagnostics (Basel) 2021; 11:diagnostics11101874. [PMID: 34679572 PMCID: PMC8535097 DOI: 10.3390/diagnostics11101874] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/17/2022] Open
Abstract
Several common and debilitating neurodegenerative disorders are characterized by the intracellular accumulation of neurofibrillary tangles (NFTs), which are composed of hyperphosphorylated tau protein. In Alzheimer's disease (AD), NFTs are accompanied by extracellular amyloid-beta (Aβ), but primary tauopathy disorders are marked by the accumulation of tau protein alone, including forms of frontotemporal dementia (FTD), corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP), among others. 18F-THK5351 has been reported to bind pathological tau as well as associated reactive astrogliosis. The goal of this study was to validate the ability of the PET tracer 18F-THK5351 to detect early changes in tau-related pathology and its relation to other pathological hallmarks. We demonstrated elevated in vivo 18F-THK5351 PET signaling over time in transgenic P301S tau mice from 8 months that had a positive correlation with histological and biochemical tau changes, as well as motor, memory, and learning impairment. This study indicates that 18F-THK5351 may help fill a critical need to develop PET imaging tracers that detect aberrant tau aggregation and related neuropathology in order to diagnose the onset of tauopathies, gain insights into their underlying pathophysiologies, and to have a reliable biomarker to follow during treatment trials.
Collapse
|
121
|
Preclinical validation of a novel oral Edaravone formulation for treatment of frontotemporal dementia. Neurotox Res 2021; 39:1689-1707. [PMID: 34599751 DOI: 10.1007/s12640-021-00405-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 10/20/2022]
Abstract
Oxidative stress is a key factor in the pathogenesis of several neurodegenerative disorders and is involved in the accumulation of amyloid beta plaques and Tau inclusions. Edaravone (EDR) is a free radical scavenger that is approved for motor neuron disease and acute ischemic stroke. EDR alleviates pathologies and cognitive impairment of AD via targeting multiple key pathways in transgenic mice. Herein, we aimed to study the effect of EDR on Tau pathology in P301L mice; an animal model of frontotemporal dementia (FTD), at two age time points representing the early and late stages of the disease. A novel EDR formulation was utilized in the study and the drug was delivered orally in drinking water for 3 months. Then, behavioral tests were conducted followed by animal sacrifice and brain dissection. Treatment with EDR improved the reference memory and accuracy in the probe trial as evaluated in Morris water maze, as well as novel object recognition and significantly alleviated motor deficits in these mice. EDR also reduced the levels of 4-hydroxy-2-nonenal and 3-nitrotyrosine adducts. In addition, immunohistochemistry showed that EDR reduced tau phosphorylation and neuroinflammation and partially rescued neurons against oxidative neurotoxicity. Moreover, EDR attenuated downstream pathologies involved in Tau hyperphosphorylation. These results suggest that EDR may be a potential therapeutic agent for the treatment of FTD.
Collapse
|
122
|
Soliman AS, Umstead A, Grabinski T, Kanaan NM, Lee A, Ryan J, Lamp J, Vega IE. EFhd2 brain interactome reveals its association with different cellular and molecular processes. J Neurochem 2021; 159:992-1007. [PMID: 34543436 PMCID: PMC9552186 DOI: 10.1111/jnc.15517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 08/11/2021] [Accepted: 09/09/2021] [Indexed: 01/06/2023]
Abstract
EFhd2 is a conserved calcium-binding protein that is highly expressed in the central nervous system. We have shown that EFhd2 interacts with tau protein, a key pathological hallmark in Alzheimer's disease and related dementias. However, EFhd2's physiological and pathological functions in the brain are still poorly understood. To gain insights into its physiological function, we identified proteins that co-immunoprecipitated with EFhd2 from mouse forebrain and hindbrain, using tandem mass spectrometry (MS). In addition, quantitative mass spectrometry was used to detect protein abundance changes due to the deletion of the Efhd2 gene in mouse forebrain and hindbrain regions. Our data show that mouse EFhd2 is associated with cytoskeleton components, vesicle trafficking modulators, cellular stress response-regulating proteins, and metabolic proteins. Moreover, proteins associated with the cytoskeleton, vesicular transport, calcium signaling, stress response, and metabolic pathways showed differential abundance in Efhd2(-/-) mice. This study presents, for the first time, an EFhd2 brain interactome that it is associated with different cellular and molecular processes. These findings will help prioritize further studies to investigate the mechanisms by which EFhd2 modulates these processes in physiological and pathological conditions of the nervous system.
Collapse
Affiliation(s)
- Ahlam S Soliman
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Neuroscience Program, Michigan State University, Grand Rapids, Michigan, USA.,Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Andrew Umstead
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Integrated Mass Spectrometry Unit, College of Human Medicine, Grand Rapids, Michigan, USA
| | - Tessa Grabinski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Nicholas M Kanaan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Neuroscience Program, Michigan State University, Grand Rapids, Michigan, USA.,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, Michigan, USA
| | - Andy Lee
- NeuroInitiatives LLC, Jacksonville, Florida, USA
| | - John Ryan
- NeuroInitiatives LLC, Jacksonville, Florida, USA
| | - Jared Lamp
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Integrated Mass Spectrometry Unit, College of Human Medicine, Grand Rapids, Michigan, USA
| | - Irving E Vega
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Neuroscience Program, Michigan State University, Grand Rapids, Michigan, USA.,Integrated Mass Spectrometry Unit, College of Human Medicine, Grand Rapids, Michigan, USA.,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, Michigan, USA.,Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
123
|
"Parkinson's disease" on the way to progressive supranuclear palsy: a review on PSP-parkinsonism. Neurol Sci 2021; 42:4927-4936. [PMID: 34532773 DOI: 10.1007/s10072-021-05601-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022]
Abstract
Progressive supranuclear palsy (PSP) is a progressive atypical parkinsonian syndrome characterised by postural instability, supranuclear ophthalmoplegia, dysarthria, dysphagia, executive dysfunction and other features. This clinical presentation represents the classic PSP-Richardson syndrome (PSP-RS). However, several other clinical subtypes have been recognised, including PSP-parkinsonism (PSP-P), probably the second most common PSP variant. Unlike PSP-RS, PSP-P often presents with an asymmetric onset, tremor and a moderate initial response to levodopa, especially during the first years of the disease, thus resembling Parkinson's disease (PD). It runs a more favourable course, but over time, PSP-P may evolve clinically into PSP-RS. Therefore, it may seem that PSP-P stands clinically between PD and PSP. There are several peculiarities that can distinguish PSP-P from these entities. As there is lack of systematic reviews on PSP-P in the literature, we decided to summarise all the necessary data about the epidemiology, clinical picture, neuroimaging, genetics and other aspects of this PSP variant in order to provide complete information for the reader.
Collapse
|
124
|
Kang SG, Han ZZ, Daude N, McNamara E, Wohlgemuth S, Molina-Porcel L, Safar JG, Mok SA, Westaway D. Pathologic tau conformer ensembles induce dynamic, liquid-liquid phase separation events at the nuclear envelope. BMC Biol 2021; 19:199. [PMID: 34503506 PMCID: PMC8428099 DOI: 10.1186/s12915-021-01132-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 08/26/2021] [Indexed: 02/07/2023] Open
Abstract
Background The microtubule-associated protein tau forms aggregates in different neurodegenerative diseases called tauopathies. Prior work has shown that a single P301L mutation in tau gene, MAPT, can promote alternative tau folding pathways that correlate with divergent clinical diagnoses. Using progressive chemical denaturation, some tau preparations from the brain featured complex transitions starting at low concentrations of guanidine hydrochloride (GdnHCl) denaturant, indicating an ensemble of differently folded tau species called conformers. On the other hand, brain samples with abundant, tangle-like pathology had simple GdnHCl unfolding profile resembling the profile of fibrillized recombinant tau and suggesting a unitary conformer composition. In studies here we sought to understand tau conformer progression and potential relationships with condensed liquid states, as well as associated perturbations in cell biological processes. Results As starting material, we used brain samples from P301L transgenic mice containing tau conformer ensembles that unfolded at low GdnHCl concentrations and with signatures resembling brain material from P301L subjects presenting with language or memory problems. We seeded reporter cells expressing a soluble form of 4 microtubule-binding repeat tau fused to GFP or YFP reporter moieties, resulting in redistribution of dispersed fluorescence signals into focal assemblies that could fuse together and move within processes between adjacent cells. Nuclear envelope fluorescent tau signals and small fluorescent inclusions behaved as a demixed liquid phase, indicative of liquid-liquid phase separation (LLPS); these droplets exhibited spherical morphology, fusion events and could recover from photobleaching. Moreover, juxtanuclear tau assemblies were associated with disrupted nuclear transport and reduced cell viability in a stable cell line. Staining for thioflavin S (ThS) became more prevalent as tau-derived inclusions attained cross-sectional area greater than 3 μm2, indicating (i) a bipartite composition, (ii) in vivo progression of tau conformers, and (iii) that a mass threshold applying to demixed condensates may drive liquid-solid transitions. Conclusions Tau conformer ensembles characterized by denaturation at low GdnHCl concentration templated the production of condensed droplets in living cells. These species exhibit dynamic changes and develop in vivo, and the larger ThS-positive assemblies may represent a waystation to arrive at intracellular fibrillar tau inclusions seen in end-stage genetic tauopathies. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01132-y.
Collapse
Affiliation(s)
- Sang-Gyun Kang
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada
| | - Zhuang Zhuang Han
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Nathalie Daude
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada
| | - Emily McNamara
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Serene Wohlgemuth
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada
| | | | - Jiri G Safar
- Department of Neurology and Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Sue-Ann Mok
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, 204 Brain and Aging Research Building, Edmonton, AB, T6G 2 M8, Canada. .,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada. .,Division of Neurology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
125
|
Sex-Related Motor Deficits in the Tau-P301L Mouse Model. Biomedicines 2021; 9:biomedicines9091160. [PMID: 34572348 PMCID: PMC8471835 DOI: 10.3390/biomedicines9091160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 01/22/2023] Open
Abstract
The contribution of mouse models for basic and translational research at different levels is important to understand neurodegenerative diseases, including tauopathies, by studying the alterations in the corresponding mouse models in detail. Moreover, several studies demonstrated that pathological as well as behavioral changes are influenced by the sex. For this purpose, we performed an in-depth characterization of the behavioral alterations in the transgenic Tau-P301L mouse model. Sex-matched wild type and homozygous Tau-P301L mice were tested in a battery of behavioral tests at different ages. Tau-P301L male mice showed olfactory and motor deficits as well as increased Tau pathology, which was not observed in Tau-P301L female mice. Both Tau-P301L male and female mice had phenotypic alterations in the SHIRPA test battery and cognitive deficits in the novel object recognition test. This study demonstrated that Tau-P301L mice have phenotypic alterations, which are in line with the histological changes and with a sex-dependent performance in those tests. Summarized, the Tau-P301L mouse model shows phenotypic alterations due to the presence of neurofibrillary tangles in the brain.
Collapse
|
126
|
Moloney CM, Lowe VJ, Murray ME. Visualization of neurofibrillary tangle maturity in Alzheimer's disease: A clinicopathologic perspective for biomarker research. Alzheimers Dement 2021; 17:1554-1574. [PMID: 33797838 PMCID: PMC8478697 DOI: 10.1002/alz.12321] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/11/2021] [Accepted: 02/03/2021] [Indexed: 12/29/2022]
Abstract
Neurofibrillary tangles, one of the neuropathologic hallmarks of Alzheimer's disease, have a dynamic lifespan of maturity that associates with progressive neuronal dysfunction and cognitive deficits. As neurofibrillary tangles mature, the biology of the neuron undergoes extensive changes that may impact biomarker recognition and therapeutic targeting. Neurofibrillary tangle maturity encompasses three levels: pretangles, mature tangles, and ghost tangles. In this review, we detail distinct and overlapping characteristics observed in the human brain regarding morphologic changes the neuron undergoes, conversion from intracellular to extracellular space, tau immunostaining patterns, and tau isoform expression changes across the lifespan of the neurofibrillary tangle. Post-translational modifications of tau such as phosphorylation, ubiquitination, conformational events, and truncations are discussed to contextualize tau immunostaining patterns. We summarize accumulated and emerging knowledge of neurofibrillary tangle maturity, discuss the current tools used to interpret the dynamic nature in the postmortem brain, and consider implications for cognitive dysfunction and tau biomarkers.
Collapse
Affiliation(s)
| | - Val J. Lowe
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
| | | |
Collapse
|
127
|
Weismiller HA, Holub TJ, Krzesinski BJ, Margittai M. A thiol-based intramolecular redox switch in four-repeat tau controls fibril assembly and disassembly. J Biol Chem 2021; 297:101021. [PMID: 34339733 PMCID: PMC8387771 DOI: 10.1016/j.jbc.2021.101021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/17/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress has been implicated in the pathogenesis and progression of several tauopathies, including Alzheimer's disease. The deposition of fibrillar inclusions made of tau protein is one of the pathological hallmarks of these disorders. Although it is becoming increasingly evident that the specific fibril structure may vary from one tauopathy to another and it is recognized that different types of isoforms (three-repeat and four-repeat tau) can be selectively deposited, little is known about the role oxidation may play in aggregation. Four-repeat tau contains two cysteines that can form an intramolecular disulfide bond, resulting in a structurally restrained compact monomer. There is discrepancy as to whether this monomer can aggregate or not. Using isolated four-repeat tau monomers (htau40) with intramolecular disulfide bonds, we demonstrate that these proteins form fibrils. The fibrils are less stable than fibrils formed under reducing conditions but are highly effective in seeding oxidized tau monomers. Conversely, a strong seeding barrier prevents incorporation of reduced tau monomers, tau mimics in which the cysteines have been replaced by alanines or serines, and three-repeat tau (htau23), a single-cysteine isoform. The barrier also holds true when seed and monomer types are reversed, indicating that oxidized and reduced tau are incompatible with each other. Surprisingly, fibrils composed of compact tau disaggregate upon reduction, highlighting the importance of the intramolecular disulfide bond for fibril stability. The findings uncover a novel binary redox switch that controls the aggregation and disaggregation of these fibrils and extend the conformational spectrum of tau aggregates.
Collapse
Affiliation(s)
- Hilary A Weismiller
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado, USA
| | - Tyler J Holub
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado, USA
| | - Brad J Krzesinski
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado, USA
| | - Martin Margittai
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado, USA.
| |
Collapse
|
128
|
Rasmussen J, Ewing AD, Bodea LG, Bodea GO, Gearing M, Faulkner GJ. An early proinflammatory transcriptional response to tau pathology is age-specific and foreshadows reduced tau burden. Brain Pathol 2021; 32:e13018. [PMID: 34463402 PMCID: PMC9048516 DOI: 10.1111/bpa.13018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/11/2021] [Accepted: 08/13/2021] [Indexed: 11/29/2022] Open
Abstract
Age is one of the strongest risk factors for the development of neurodegenerative diseases, the majority of which involve misfolded protein aggregates in the brain. These protein aggregates are thought to drive pathology and are attractive targets for the development of new therapies. However, it is unclear how age influences the onset of pathology and the accompanying molecular response. To address this knowledge gap, we used a model of seeded tau pathology to profile the transcriptomic changes in 3 and 12 month old mice in response to developing tau hyperphosphorylation and aggregation. First, we found the burden of hyperphosphorylated tau pathology in mice injected at 12 months of age was moderately reduced compared to animals injected at 3 months. On a molecular level, we found an inflammation-related subset of genes, including C3 and the disease-associated microglia genes Ctsd, Cst7, and Clec7a, were more expressed early in disease in 12 but not 3 month old mice. These findings provide evidence of an early, age-specific response to tau pathology, which could serve as a marker for the severity of downstream pathology.
Collapse
Affiliation(s)
- Jay Rasmussen
- Queensland Brain Institute, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Adam D Ewing
- Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Liviu-Gabriel Bodea
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Gabriela O Bodea
- Queensland Brain Institute, University of Queensland, St Lucia, Brisbane, Queensland, Australia.,Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Marla Gearing
- Departments of Pathology and Laboratory Medicine and Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Geoffrey J Faulkner
- Queensland Brain Institute, University of Queensland, St Lucia, Brisbane, Queensland, Australia.,Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| |
Collapse
|
129
|
Tau K321/K353 pseudoacetylation within KXGS motifs regulates tau-microtubule interactions and inhibits aggregation. Sci Rep 2021; 11:17069. [PMID: 34426645 PMCID: PMC8382713 DOI: 10.1038/s41598-021-96627-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 08/10/2021] [Indexed: 11/09/2022] Open
Abstract
Alzheimer's disease is the leading cause of dementia and a defining hallmark is the progressive brain deposition of tau aggregates. The insidious accumulation of brain tau inclusions is also involved in a group of neurodegenerative diseases termed frontotemporal dementias. In all of these disorders, tau aggregates are enriched in post-translational modifications including acetylation, which has recently been identified at multiple sites. While most evidence suggest that tau acetylation is detrimental and promotes tau aggregation, a few studies support that tau acetylation within the KXGS motif can be protective and inhibit tau aggregation. To model site-specific acetylation at K259, K290, K321, and K353, acetylmimetics were created by mutating lysine to glutamine residues, which approximates size and charge of acetylation. HEK293T cells were transfected to express wild type tau, tau pathogenic mutations (P301L and P301L/S320F) or tau acetylmimetics and assessed by cell-based assays for microtubule binding and tau aggregation. Acetylmimetics within the KXGS motif (K259Q, K290Q, K321Q, K353Q) leads to significant decreased tau-microtubule interactions. Acetylmimetics K321Q and K353Q within the context of the pathogenic P301L tau mutation strongly inhibited prion-like seeded aggregation. This protective effect was confirmed to decrease intrinsic aggregation of P301L/S320F tau double mutation. Surprisingly, K321Q and K353Q acetylmimetics altered the conformational structure of P301L/S320F tau to extensively impair Thioflavin S binding. Site-specific acetylation of tau at K321 and K353 could represent a natural protective mechanism against tau aggregation and could be a potential therapeutic target.
Collapse
|
130
|
Giong HK, Subramanian M, Yu K, Lee JS. Non-Rodent Genetic Animal Models for Studying Tauopathy: Review of Drosophila, Zebrafish, and C. elegans Models. Int J Mol Sci 2021; 22:8465. [PMID: 34445171 PMCID: PMC8395099 DOI: 10.3390/ijms22168465] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Tauopathy refers to a group of progressive neurodegenerative diseases, including frontotemporal lobar degeneration and Alzheimer's disease, which correlate with the malfunction of microtubule-associated protein Tau (MAPT) due to abnormal hyperphosphorylation, leading to the formation of intracellular aggregates in the brain. Despite extensive efforts to understand tauopathy and develop an efficient therapy, our knowledge is still far from complete. To find a solution for this group of devastating diseases, several animal models that mimic diverse disease phenotypes of tauopathy have been developed. Rodents are the dominating tauopathy models because of their similarity to humans and established disease lines, as well as experimental approaches. However, powerful genetic animal models using Drosophila, zebrafish, and C. elegans have also been developed for modeling tauopathy and have contributed to understanding the pathophysiology of tauopathy. The success of these models stems from the short lifespans, versatile genetic tools, real-time in-vivo imaging, low maintenance costs, and the capability for high-throughput screening. In this review, we summarize the main findings on mechanisms of tauopathy and discuss the current tauopathy models of these non-rodent genetic animals, highlighting their key advantages and limitations in tauopathy research.
Collapse
Affiliation(s)
- Hoi-Khoanh Giong
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- KRIBB School, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Manivannan Subramanian
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Kweon Yu
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- KRIBB School, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Jeong-Soo Lee
- Disease Target Structure Research Center, KRIBB, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.-K.G.); (M.S.)
- KRIBB School, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Dementia DTC R&D Convergence Program, KIST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Korea
| |
Collapse
|
131
|
Li Q, Xiong C, Liu H, Ge H, Yao X, Liu H. Computational Insights Into the Inhibition Mechanism of Proanthocyanidin B2 on Tau Hexapeptide (PHF6) Oligomer. Front Chem 2021; 9:666043. [PMID: 34336783 PMCID: PMC8316602 DOI: 10.3389/fchem.2021.666043] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/14/2021] [Indexed: 11/13/2022] Open
Abstract
The formation of amyloid fibrils from Tau is a key pathogenic feature of Alzheimer's disease (AD). To disturb the formation of Tau aggregates is considered as a promising therapeutic strategy for AD. Recently, a natural product proanthocyanidin B2 (PB2) was confirmed to not only inhibit Tau aggregation, but also disaggregate Tau fibrils. Herein, to explore the inhibition mechanism of PB2 against Tau fibril and to provide the useful information for drug design and discovery, all-atom molecular dynamics simulations were carried out for the ordered Tau hexapeptide PHF6 oligomer in the presence and absence of PB2. The obtained result shows that PB2 can transform PHF6 oligomer from the ordered β-sheet structure into disordered one. Moreover, the clustering analysis and binding free energy calculations identify that S3 site is the most potential binding site. At S3 site, by hydrophobic and hydrogen bond interactions, the residues V309, Y310 and K311 are essential for binding with PB2, especially K311. In a word, our study reveals the molecular mechanism of PB2 inhibiting PHF6 aggregation and it will provide some valuable information for the development of Tau aggregation inhibitors.
Collapse
Affiliation(s)
- Qin Li
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Chunmei Xiong
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hongli Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Huizhen Ge
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, China
| | - Huanxiang Liu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
132
|
Abstract
Tauopathies consist of over 25 different neurodegenerative diseases that include argyrophilic grain disease (AGD), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Pick’s disease (PiD). Tauopathies are defined by brain accumulation of microtubule-associated protein tau in fibrillar aggregates, whose prevalence strongly correlates with dementia. Dominant mutations in tau cause neurodegenerative diseases, and most increase its aggregation propensity. Pathogenesis of tauopathies may involve pathological tau conformers that serve as templates to recruit native protein into growing assemblies and also move between brain cells to cause disease progression, similar to prions. Prions adopt pathological conformations, termed “strains,” that stably propagate in living systems, and create unique patterns of neuropathology. Data from multiple laboratories now suggest that tau acts as a prion. It propagates unique strains indefinitely in cultured cells, and when these are inoculated into mouse models, they create defined neuropathological patterns, which establish a direct link between conformation and disease. In humans, distinct fibril structures are associated with different diseases, but causality has not been established as in mice. Cryo-EM structures of tau fibrils isolated from tauopathy brains reveal distinct fibril cores across disease. Interestingly, the conformation of the tau monomer unit within different fibril subtypes from the same patient appears relatively preserved. This is consistent with data that the tau monomer samples an ensemble of conformations that act as distinct pathologic templates in the formation of restricted numbers of strains. The propensity of a tau monomer to adopt distinct conformations appears to be linked to defined local motifs that expose different patterns of amyloidogenic amino acid sequences. The prion hypothesis, which predicts that protein structure dictates resultant disease, has proved particularly useful to understand the diversity of human tauopathies. The challenge now is to develop methods to rapidly classify patients according to the structure of the underlying pathological protein assemblies to achieve more accurate diagnosis and effective therapy.
Collapse
|
133
|
Schwerin SC, Chatterjee M, Hutchinson EB, Djankpa FT, Armstrong RC, McCabe JT, Perl DP, Juliano SL. Expression of GFAP and Tau Following Blast Exposure in the Cerebral Cortex of Ferrets. J Neuropathol Exp Neurol 2021; 80:112-128. [PMID: 33421075 DOI: 10.1093/jnen/nlaa157] [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
Blast exposures are a hallmark of contemporary military conflicts. We need improved preclinical models of blast traumatic brain injury for translation of pharmaceutical and therapeutic protocols. Compared with rodents, the ferret brain is larger, has substantial sulci, gyri, a higher white to gray matter ratio, and the hippocampus in a ventral position; these attributes facilitate comparison with the human brain. In this study, ferrets received compressed air shock waves and subsequent evaluation of glia and forms of tau following survival of up to 12 weeks. Immunohistochemistry and Western blot demonstrated altered distributions of astrogliosis and tau expression after blast exposure. Many aspects of the astrogliosis corresponded to human pathology: increased subpial reactivity, gliosis at gray-white matter interfaces, and extensive outlining of blood vessels. MRI analysis showed numerous hypointensities occurring in the 12-week survival animals, appearing to correspond to luminal expansions of blood vessels. Changes in forms of tau, including phosphorylated tau, and the isoforms 3R and 4R were noted using immunohistochemistry and Western blot in specific regions of the cerebral cortex. Of particular interest were the 3R and 4R isoforms, which modified their ratio after blast. Our data strongly support the ferret as an animal model with highly translational features to study blast injury.
Collapse
Affiliation(s)
- Susan C Schwerin
- From the Department of Anatomy Physiology and Genetics, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | | | - Elizabeth B Hutchinson
- Quantitative Medical Imaging Section, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Francis T Djankpa
- From the Department of Anatomy Physiology and Genetics, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Program in Neuroscience, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Department of Physiology, School of Medical Sciences, University of Cape Coast, Ghana
| | - Regina C Armstrong
- From the Department of Anatomy Physiology and Genetics, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Program in Neuroscience, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Joseph T McCabe
- From the Department of Anatomy Physiology and Genetics, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Program in Neuroscience, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Daniel P Perl
- Program in Neuroscience, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Sharon L Juliano
- From the Department of Anatomy Physiology and Genetics, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Program in Neuroscience, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
134
|
Saito T, Chiku T, Oka M, Wada-Kakuda S, Nobuhara M, Oba T, Shinno K, Abe S, Asada A, Sumioka A, Takashima A, Miyasaka T, Ando K. Disulfide bond formation in microtubule-associated tau protein promotes tau accumulation and toxicity in vivo. Hum Mol Genet 2021; 30:1955-1967. [PMID: 34137825 PMCID: PMC8522637 DOI: 10.1093/hmg/ddab162] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 11/12/2022] Open
Abstract
Accumulation of microtubule-associated tau protein is thought to cause neuron loss in a group of neurodegenerative diseases called tauopathies. In diseased brains, tau molecules adopt pathological structures that propagate into insoluble forms with disease-specific patterns. Several types of posttranslational modifications in tau are known to modulate its aggregation propensity in vitro, but their influence on tau accumulation and toxicity at the whole-organism level has not been fully elucidated. Herein, we utilized a series of transgenic Drosophila models to compare systematically the toxicity induced by five tau constructs with mutations or deletions associated with aggregation, including substitutions at seven disease-associated phosphorylation sites (S7A and S7E), deletions of PHF6 and PHF6* sequences (ΔPHF6 and ΔPHF6*), and substitutions of cysteine residues in the microtubule binding repeats (C291/322A). We found that substitutions and deletions resulted in different patterns of neurodegeneration and accumulation, with C291/322A having a dramatic effect on both tau accumulation and neurodegeneration. These cysteines formed disulfide bonds in mouse primary cultured neurons and in the fly retina, and stabilized tau proteins. Additionally, they contributed to tau accumulation under oxidative stress. We also found that each of these cysteine residues contributes to the microtubule polymerization rate and microtubule levels at equilibrium, but none of them affected tau binding to polymerized microtubules. Since tau proteins expressed in the Drosophila retina are mostly present in the early stages of tau filaments self-assembly, our results suggest that disulfide bond formation by these cysteine residues could be attractive therapeutic targets.
Collapse
Affiliation(s)
- Taro Saito
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan.,Department of Biological Sciences, Faculty of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Tomoki Chiku
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Mikiko Oka
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Satoko Wada-Kakuda
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Mika Nobuhara
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Toshiya Oba
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Kanako Shinno
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Saori Abe
- Department of Biological Sciences, Faculty of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Akiko Asada
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan.,Department of Biological Sciences, Faculty of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Akio Sumioka
- Faculty of Science, Department of Life Science, Gakushuin University, Tokyo, Japan
| | - Akihiko Takashima
- Faculty of Science, Department of Life Science, Gakushuin University, Tokyo, Japan
| | - Tomohiro Miyasaka
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Kanae Ando
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan.,Department of Biological Sciences, Faculty of Science, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
135
|
Winder Z, Wilcock D, Jicha GA. Diagnostic and Prognostic Laboratory Testing for Alzheimer Disease. Clin Lab Med 2021; 40:289-303. [PMID: 32718500 DOI: 10.1016/j.cll.2020.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This article focuses on current clinical laboratory testing to diagnose Alzheimer disease and monitor its progression throughout its disease course. Several clinically available tests focus on analysis of amyloid and tau levels in cerebrospinal fluid as well as autosomal dominant and risk factor genes. Although the current armament of clinical laboratory testing is limited by invasiveness of cerebrospinal fluid collection, rarity of autosomal dominant genetic mutations, and uncertainties of risk inherent in nonpenetrant genes, the field is poised to advance the clinical repertoire of laboratory diagnostic testing.
Collapse
Affiliation(s)
- Zachary Winder
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA
| | - Donna Wilcock
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA
| | - Gregory A Jicha
- Department of Neurology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA.
| |
Collapse
|
136
|
Xia Y, Prokop S, Giasson BI. "Don't Phos Over Tau": recent developments in clinical biomarkers and therapies targeting tau phosphorylation in Alzheimer's disease and other tauopathies. Mol Neurodegener 2021; 16:37. [PMID: 34090488 PMCID: PMC8180161 DOI: 10.1186/s13024-021-00460-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Phosphorylation is one of the most prevalent post-translational modifications found in aggregated tau isolated from Alzheimer’s disease (AD) patient brains. In tauopathies like AD, increased phosphorylation or hyperphosphorylation can contribute to microtubule dysfunction and is associated with tau aggregation. In this review, we provide an overview of the structure and functions of tau protein as well as the physiologic roles of tau phosphorylation. We also extensively survey tau phosphorylation sites identified in brain tissue and cerebrospinal fluid from AD patients compared to age-matched healthy controls, which may serve as disease-specific biomarkers. Recently, new assays have been developed to measure minute amounts of specific forms of phosphorylated tau in both cerebrospinal fluid and plasma, which could potentially be useful for aiding clinical diagnosis and monitoring disease progression. Additionally, multiple therapies targeting phosphorylated tau are in various stages of clinical trials including kinase inhibitors, phosphatase activators, and tau immunotherapy. With promising early results, therapies that target phosphorylated tau could be useful at slowing tau hyperphosphorylation and aggregation in AD and other tauopathies.
Collapse
Affiliation(s)
- Yuxing Xia
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA.,Department of Pathology, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA.,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, BMS J483/CTRND, 1275 Center Drive, Gainesville, FL, 32610, USA. .,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA. .,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, 32610, USA.
| |
Collapse
|
137
|
Creekmore BC, Chang YW, Lee EB. The Cryo-EM Effect: Structural Biology of Neurodegenerative Disease Aggregates. J Neuropathol Exp Neurol 2021; 80:514-529. [PMID: 33970243 PMCID: PMC8177849 DOI: 10.1093/jnen/nlab039] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neurogenerative diseases are characterized by diverse protein aggregates with a variety of microscopic morphologic features. Although ultrastructural studies of human neurodegenerative disease tissues have been conducted since the 1960s, only recently have near-atomic resolution structures of neurodegenerative disease aggregates been described. Solid-state nuclear magnetic resonance spectroscopy and X-ray crystallography have provided near-atomic resolution information about in vitro aggregates but pose logistical challenges to resolving the structure of aggregates derived from human tissues. Recent advances in cryo-electron microscopy (cryo-EM) have provided the means for near-atomic resolution structures of tau, amyloid-β (Aβ), α-synuclein (α-syn), and transactive response element DNA-binding protein of 43 kDa (TDP-43) aggregates from a variety of diseases. Importantly, in vitro aggregate structures do not recapitulate ex vivo aggregate structures. Ex vivo tau aggregate structures indicate individual tauopathies have a consistent aggregate structure unique from other tauopathies. α-syn structures show that even within a disease, aggregate heterogeneity may correlate to disease course. Ex vivo structures have also provided insight into how posttranslational modifications may relate to aggregate structure. Though there is less cryo-EM data for human tissue-derived TDP-43 and Aβ, initial structural studies provide a basis for future endeavors. This review highlights structural variations across neurodegenerative diseases and reveals fundamental differences between experimental systems and human tissue derived protein inclusions.
Collapse
Affiliation(s)
- Benjamin C Creekmore
- From the Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Graduate Program in Biochemistry and Molecular Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yi-Wei Chang
- From the Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward B Lee
- Send correspondence to: Edward B. Lee, MD, PhD, Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 613A Stellar Chance Laboratories, 422 Curie Blvd., Philadelphia, PA 19104, USA; E-mail:
| |
Collapse
|
138
|
Cho JD, Kim YA, Rafikian EE, Yang M, Santa-Maria I. Marked Mild Cognitive Deficits in Humanized Mouse Model of Alzheimer's-Type Tau Pathology. Front Behav Neurosci 2021; 15:634157. [PMID: 34093145 PMCID: PMC8175658 DOI: 10.3389/fnbeh.2021.634157] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/21/2021] [Indexed: 01/23/2023] Open
Abstract
Hyperphosphorylation and the subsequent aggregation of tau protein into neurofibrillary tangles (NFTs) are well-established neuropathological hallmarks of Alzheimer’s disease (AD) and associated tauopathies. To further examine the impact and progression of human tau pathology in neurodegenerative contexts, the humanized tau (htau) mouse model was originally created. Despite AD-like tau pathological features recapitulated in the htau mouse model, robustness of behavioral phenotypes has not been fully established. With the ultimate goal of evaluating the htau mouse model as a candidate for testing AD therapeutics, we set out to verify, in-house, the presence of robust, replicable cognitive deficits in the htau mice. The present study shows behavioral data collected from a carefully curated battery of learning and memory tests. Here we report a significant short-term spatial memory deficit in aged htau mice, representing a novel finding in this model. However, we did not find salient impairments in long-term learning and memory previously reported in this mouse model. Here, we attempted to understand the discrepancies in the literature by highlighting the necessity of scrutinizing key procedural differences across studies. Reported cognitive deficits in the htau model may depend on task difficulty and other procedural details. While the htau mouse remains a unique and valuable animal model for replicating late onset AD-like human tau pathology, its cognitive deficits are modest under standard testing conditions. The overarching message is that before using any AD mouse model to evaluate treatment efficacies, it is imperative to first characterize and verify the presence of behavioral deficits in-house.
Collapse
Affiliation(s)
- Joshua D Cho
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States.,Department of Pathology & Cell Biology, Columbia University, New York, NY, United States
| | - Yoon A Kim
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States.,Department of Pathology & Cell Biology, Columbia University, New York, NY, United States
| | - Elizabeth E Rafikian
- The Mouse NeuroBehavior Core, Institute for Genomic Medicine, Columbia University, New York, NY, United States
| | - Mu Yang
- The Mouse NeuroBehavior Core, Institute for Genomic Medicine, Columbia University, New York, NY, United States
| | - Ismael Santa-Maria
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States.,Department of Pathology & Cell Biology, Columbia University, New York, NY, United States
| |
Collapse
|
139
|
Sarkar S. Shaggy functions downstream of dMyc and their concurrent downregulation confers additive rescue against tau toxicity in Drosophila. Biofactors 2021; 47:461-477. [PMID: 33651466 DOI: 10.1002/biof.1721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022]
Abstract
Neurodegenerative tauopathies such as Alzheimer's and Parkinson's diseases are characterized by hyperphosphorylation of tau protein and their subsequent aggregation in the forms of paired helical filaments and/or neurofibrillary tangles in specific areas of the brain. Despite several attempts, it remains a challenge to develop reliable biomarkers or effective drugs against tauopathies. It is increasingly evident now that due to the involvement of multiple cellular cascades affected by the pathogenic tau molecules, a single genetic modifier or a molecule is unlikely to be efficient enough to provide an inclusive rescue. Hence, multitargets based combinatorial approach(s) have been suggested to provide an efficient rescue against tauopathies. We have reported earlier that targeted downregulation of dmyc (a Drosophila homolog of human cmyc proto-oncogene) restricts tau etiology by limiting tau hyperphosphorylation and heterochromatin loss. Although, dmyc generates a significant rescue; however, it is not proficient enough to provide a complete alleviation against tauopathies. Here, we report that tissue-specific concurrent downregulation of dmyc and gsk3β conveys a near-complete rescue against tau toxicity in Drosophila. We noted that combinatorial downregulation of dmyc and gsk3β reduces tau hyperphosphorylation, restricts the formation of neurofibrillary tangles, and restores heterochromatin loss to the physiological level. Our subsequent investigations revealed that dmyc regulates gsk3β via protein phosphatase 2A (dPP2A) in a dose-dependent manner to regulate tau pathogenesis. We propose that dmyc and gsk3β candidates can be utilized in a synergistic manner for the development of an efficient combinatorial therapeutic approach against the devastating human tauopathies.
Collapse
Affiliation(s)
- Surajit Sarkar
- Department of Genetics, University of Delhi South Campus, New Delhi, India
| |
Collapse
|
140
|
Nisha, Sarkar S. Downregulation of glob1 suppresses pathogenesis of human neuronal tauopathies in Drosophila by regulating tau phosphorylation and ROS generation. Neurochem Int 2021; 146:105040. [PMID: 33865914 DOI: 10.1016/j.neuint.2021.105040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/30/2022]
Abstract
Human tauopathies represent a group of neurodegenerative disorders, characterized by abnormal hyperphosphorylation and aggregation of tau protein, which ultimately cause neurodegeneration. The aberrant tau hyperphosphorylation is mostly attributed to the kinases/phosphatases imbalance, which is majorly contributed by the generation of reactive oxygen species (ROS). Globin(s) represent a well-conserved group of proteins which are involved in O2 management, regulation of cellular ROS in different cell types. Similarly, Drosophila globin1 (a homologue of human globin) with its known roles in oxygen management and development of nervous system exhibits striking similarities with the mammalian neuroglobin. Several recent evidences support the hypothesis that neuroglobins are associated with Alzheimer's disease pathogenesis. We herein noted that targeted expression of human-tau induces the cellular level of Glob1 protein in Drosophila tauopathy models. Subsequently, RNAi mediated restored level of Glob1 restricts the pathogenic effect of human-tau by minimizing its hyperphosphorylation via GSK-3β/p-Akt and p-JNK pathways. In addition, it also activates the Nrf2-keap1-ARE cascade to stabilize the tau-mediated increased level of ROS. These two parallel cellular events provide a significant rescue against human tau-mediated neurotoxicity in the fly models. For the first time we report a direct involvement of an oxygen sensing globin gene in tau etiology. In view of the fact that human genome encodes for the multiple Globin proteins including a nervous system specific neuroglobin; and therefore, our findings may pave the way to investigate if the conserved oxygen sensing globin gene(s) can be exploited in devising novel therapeutic strategies against tauopathies.
Collapse
Affiliation(s)
- Nisha
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Surajit Sarkar
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India.
| |
Collapse
|
141
|
Peet BT, Spina S, Mundada N, La Joie R. Neuroimaging in Frontotemporal Dementia: Heterogeneity and Relationships with Underlying Neuropathology. Neurotherapeutics 2021; 18:728-752. [PMID: 34389969 PMCID: PMC8423978 DOI: 10.1007/s13311-021-01101-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2021] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia encompasses a group of clinical syndromes defined pathologically by degeneration of the frontal and temporal lobes. Historically, these syndromes have been challenging to diagnose, with an average of about three years between the time of symptom onset and the initial evaluation and diagnosis. Research in the field of neuroimaging has revealed numerous biomarkers of the various frontotemporal dementia syndromes, which has provided clinicians with a method of narrowing the differential diagnosis and improving diagnostic accuracy. As such, neuroimaging is considered a core investigative tool in the evaluation of neurodegenerative disorders. Furthermore, patterns of neurodegeneration correlate with the underlying neuropathological substrates of the frontotemporal dementia syndromes, which can aid clinicians in determining the underlying etiology and improve prognostication. This review explores the advancements in neuroimaging and discusses the phenotypic and pathologic features of behavioral variant frontotemporal dementia, semantic variant primary progressive aphasia, and nonfluent variant primary progressive aphasia, as seen on structural magnetic resonance imaging and positron emission tomography.
Collapse
Affiliation(s)
- Bradley T Peet
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA.
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Nidhi Mundada
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| |
Collapse
|
142
|
Croce KR, Yamamoto A. Dissolving the Complex Role Aggregation Plays in Neurodegenerative Disease. Mov Disord 2021; 36:1061-1069. [PMID: 33755257 DOI: 10.1002/mds.28522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/11/2021] [Accepted: 01/18/2021] [Indexed: 11/10/2022] Open
Abstract
Prominent neuropathological hallmarks of many adult-onset neurodegenerative diseases include the deposition and accumulation of misfolded proteins or conformers; however, their role in pathogenesis has remained unclear. This is in part due to the deceptive simplicity of the question and our limited understanding of how protein homeostasis is maintained in the compartmentalized cells of the central nervous system, especially in the context of the adult brain. Building on studies from simple cell-based systems and invertebrate animals, we recently identified a protein central to the specific and selective turnover of aggregated proteins in the adult brain, the autophagy-linked FYVE protein (Alfy)/Wdfy3. Depletion of Alfy levels in a mouse model of Huntington's disease showed that it accelerated the accumulation of the aggregated mutant huntingtin protein, as well as the onset of behavioral deficits. Although the motor dysfunction was accelerated in the model, this was in the absence of increasing overt cell loss, implicating protein aggregates as a modifier of circuit dysfunction rather than driving degeneration per se. We discuss these findings in the context of what is known about protein accumulation and how we can use proteins such as Alfy to determine if protein accumulation is a shared pathogenic event across different adult-onset diseases. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Katherine R Croce
- Doctoral Program in Pathobiology, Columbia University, New York, New York, USA
| | - Ai Yamamoto
- Departments of Neurology, Pathology and Cell Biology, Columbia University, New York, New York, USA
| |
Collapse
|
143
|
Ahmed TF, Ahmed A, Imtiaz F. History in perspective: How Alzheimer's Disease came to be where it is? Brain Res 2021; 1758:147342. [PMID: 33548268 DOI: 10.1016/j.brainres.2021.147342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/18/2021] [Accepted: 01/28/2021] [Indexed: 01/03/2023]
Abstract
Treatment of Alzheimer's Disease (AD) remains an unsolved issue despite the pronounced global attention it has received from researchers over the last four decades. Determining the primary cause of the disease is challenging due to its long prodromal phase and multifactorial etiology. Regardless, academic disagreements amongst the scientific community have helped in making significant advancements in underpinning the molecular basis of disease pathogenesis. Substantial development in fluid and imaging biomarkers for AD led to a sharp turn in defining the disease as a molecular construct, dispensing its clinical definition. With conceptual progress, revisions in the diagnostic criteria of AD were made, culminating into the research framework proposed by National Institute on Aging and Alzheimer's Association in 2018 which unified different stages of the disease continuum, giving a common language of AT(N)1 classification to researchers. With realization that dementia is the final stage of AD spectrum, its early diagnosis by means of cerebrospinal fluid biomarkers, Positron Emission Tomography and Magnetic Resonance Imaging of the brain holds crucial importance in discovering ways of halting the disease progression. This article maps the insights into the pathogenesis as well as the diagnostic criteria and tests for AD as these have evolved over time. A contextualized timeline of how the understanding of AD has matured with advancing knowledge allows future research to be directed and unexplored avenues to be prioritized.
Collapse
Affiliation(s)
- Tehniat F Ahmed
- Department of Biochemistry, Institute of Biomedical Sciences, Dow University of Health Sciences, Karachi, Pakistan.
| | - Affan Ahmed
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Fauzia Imtiaz
- Department of Biochemistry, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
144
|
Lindstedt PR, Taylor RJ, Bernardes GJL, Vendruscolo M. Facile Installation of Post-translational Modifications on the Tau Protein via Chemical Mutagenesis. ACS Chem Neurosci 2021; 12:557-561. [PMID: 33464820 DOI: 10.1021/acschemneuro.0c00761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Post-translational modifications of proteins are ubiquitous in living organisms, as they enable an accurate control of the interactions of these macromolecules. For mechanistic studies, it would be highly advantageous to be able to produce in vitro post-translationally modified proteins with site-specificity. Here, we demonstrate one facile way to achieve this goal through the use of post-translational chemical mutagenesis. We illustrate this approach by performing site-specific phosphorylation and methylation of tau, a protein that stabilizes microtubules and whose aggregation is closely linked with Alzheimer's disease. We then verify the effects of the post-translational modifications on the ability of tau to control microtubule polymerization, revealing in particular an unexpected role for phosphorylation at S199, which is outside the microtubule-binding region of tau. These results show how the chemical mutagenesis approach that we present enables the systematic analysis of site-specific post-translational modifications of a key protein involved in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Philip R. Lindstedt
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Ross J. Taylor
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Gonçalo J. L. Bernardes
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| |
Collapse
|
145
|
Alausa A, Ogundepo S, Olaleke B, Adeyemi R, Olatinwo M, Ismail A. Chinese nutraceuticals and physical activity; their role in neurodegenerative tauopathies. Chin Med 2021; 16:1. [PMID: 33407732 PMCID: PMC7789572 DOI: 10.1186/s13020-020-00418-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The onset of neurodegenerative disease has not only been a major cause of scientific worry, but of economic burden to the health system. This condition has been further attributed to mis-stability, deletion or mutation of tau protein, causing the onset of Corticobasal degeneration, Pick's diseases, Progressive supranuclear palsy, Argyrophilic grains disease, Alzheimer's diseases etc. as scientifically renowned. This is mainly related to dysregulation of translational machinery, upregulation of proinflammatory cytokines and inhibition of several essential cascades such as ERK signaling cascade, GSK3β, CREB, and PKA/PKB (Akt) signaling cascades that enhances protein processing, normal protein folding, cognitive function, and microtubule associated tau stability. Administration of some nutrients and/or bioactive compounds has a high tendency to impede tau mediated inflammation at neuronal level. Furthermore, prevention and neutralization of protein misfolding through modulation of microtubule tau stability and prevention of protein misfolding is by virtue few of the numerous beneficial effects of physical activity. Of utmost important in this study is the exploration of promising bioactivities of nutraceuticals found in china and the ameliorating potential of physical activity on tauopathies, while highlighting animal and in vitro studies that have been investigated for comprehensive understanding of its potential and an insight into the effects on human highly probable to tau mediated neurodegeneration.
Collapse
Affiliation(s)
- Abdullahi Alausa
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | - Sunday Ogundepo
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | - Barakat Olaleke
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | - Rofiat Adeyemi
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria.
| | - Mercy Olatinwo
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | - Aminat Ismail
- Department of Science Laboratory Technology, Faculty of Pure & Applied Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| |
Collapse
|
146
|
Kim YC, Jeong BH. Identification of Somatic Mutations in Dementia-related Genes in Cancer Patients. Curr Alzheimer Res 2020; 17:835-844. [PMID: 33272183 DOI: 10.2174/1567205017666201203124341] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 09/10/2020] [Accepted: 10/01/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Dementia is an overall term of brain diseases, including Alzheimer's disease (AD), tauopathies and synucleinopathies. To date, somatic mutations in dementia-related genes, including the amyloid precursor protein (APP) gene, presenilin 1 (PSEN1) gene, PSEN2 gene, microtubule- associated protein tau (MAPT) gene, alpha-synuclein (SNCA) gene and leucine-rich repeat kinase 2 (LRRK2) gene, have been considered one cause of dementia. We have questioned the impact of somatic mutations in dementia-related genes on cancer. METHODS In the present study, we investigated somatic mutations in the APP, PSEN1, PSEN2, MAPT, SNCA and LRRK2 genes and the impact of these somatic mutations. RESULTS From The Cancer Genome Atlas (TCGA) database, we found 1,643 somatic mutations in the APP, PSEN1, PSEN2, MAPT, SNCA and LRRK2 genes in cancer patients. Strikingly, compared to the distributions of cancer types in total cancer patients, somatic mutations in the dementia-related genes showed an extremely low distribution in glioblastoma patients. CONCLUSION To the best of our knowledge, this is the first investigation of dementia-related genes in cancer patients.
Collapse
Affiliation(s)
- Yong-Chan Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeonbuk 54531, Korea
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeonbuk 54531, Korea
| |
Collapse
|
147
|
Boiarska Z, Passarella D. Microtubule-targeting agents and neurodegeneration. Drug Discov Today 2020; 26:604-615. [PMID: 33279455 DOI: 10.1016/j.drudis.2020.11.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/17/2020] [Accepted: 11/28/2020] [Indexed: 11/25/2022]
Abstract
The association of microtubule (MT) breakdown with neurodegeneration and neurotoxicity has provided an emerging therapeutic approach for neurodegenerative diseases. Tubulin binders are able to modulate MT dynamics and, as a result, are of particular interest both as potential therapeutics and experimental tools used to validate this strategy. Here, we provide a comprehensive overview of current knowledge and recent advancements regarding MT-targeting approaches for neurodegeneration and evaluate the potential application of MT-targeting agents (MTAs) based on available preclinical and clinical data.
Collapse
Affiliation(s)
- Zlata Boiarska
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | - Daniele Passarella
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy.
| |
Collapse
|
148
|
Kang SG, Eskandari-Sedighi G, Hromadkova L, Safar JG, Westaway D. Cellular Biology of Tau Diversity and Pathogenic Conformers. Front Neurol 2020; 11:590199. [PMID: 33304310 PMCID: PMC7693435 DOI: 10.3389/fneur.2020.590199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
Tau accumulation is a prominent feature in a variety of neurodegenerative disorders and remarkable effort has been expended working out the biochemistry and cell biology of this cytoplasmic protein. Tau's wayward properties may derive from germline mutations in the case of frontotemporal lobar degeneration (FTLD-MAPT) but may also be prompted by less understood cues—perhaps environmental or from molecular damage as a consequence of chronological aging—in the case of idiopathic tauopathies. Tau properties are undoubtedly affected by its covalent structure and in this respect tau protein is not only subject to changes in length produced by alternative splicing and endoproteolysis, but different types of posttranslational modifications that affect different amino acid residues. Another layer of complexity concerns alternate conformations—“conformers”—of the same covalent structures; in vivo conformers can encompass soluble oligomeric species, ramified fibrillar structures evident by light and electron microscopy and other forms of the protein that have undergone liquid-liquid phase separation to make demixed liquid droplets. Biological concepts based upon conformers have been charted previously for templated replication mechanisms for prion proteins built of the PrP polypeptide; these are now providing useful explanations to feature tau pathobiology, including how this protein accumulates within cells and how it can exhibit predictable patterns of spread across different neuroanatomical regions of an affected brain. In sum, the documented, intrinsic heterogeneity of tau forms and conformers now begins to speak to a fundamental basis for diversity in clinical presentation of tauopathy sub-types. In terms of interventions, emphasis upon subclinical events may be worthwhile, noting that irrevocable cell loss and ramified protein assemblies feature at end-stage tauopathy, whereas earlier events may offer better opportunities for diverting pathogenic processes. Nonetheless, the complexity of tau sub-types, which may be present even within intermediate disease stages, likely mitigates against one-size-fits-all therapeutic strategies and may require a suite of interventions. We consider the extent to which animal models of tauopathy can be reasonably enrolled in the campaign to produce such interventions and to slow the otherwise inexorable march of disease progression.
Collapse
Affiliation(s)
- Sang-Gyun Kang
- Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | | | - Lenka Hromadkova
- Department of Neurology and Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Jiri G Safar
- Department of Neurology and Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - David Westaway
- Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
149
|
Alyenbaawi H, Allison WT, Mok SA. Prion-Like Propagation Mechanisms in Tauopathies and Traumatic Brain Injury: Challenges and Prospects. Biomolecules 2020; 10:E1487. [PMID: 33121065 PMCID: PMC7692808 DOI: 10.3390/biom10111487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/23/2022] Open
Abstract
The accumulation of tau protein in the form of filamentous aggregates is a hallmark of many neurodegenerative diseases such as Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). These dementias share traumatic brain injury (TBI) as a prominent risk factor. Tau aggregates can transfer between cells and tissues in a "prion-like" manner, where they initiate the templated misfolding of normal tau molecules. This enables the spread of tau pathology to distinct parts of the brain. The evidence that tauopathies spread via prion-like mechanisms is considerable, but work detailing the mechanisms of spread has mostly used in vitro platforms that cannot fully reveal the tissue-level vectors or etiology of progression. We review these issues and then briefly use TBI and CTE as a case study to illustrate aspects of tauopathy that warrant further attention in vivo. These include seizures and sleep/wake disturbances, emphasizing the urgent need for improved animal models. Dissecting these mechanisms of tauopathy progression continues to provide fresh inspiration for the design of diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hadeel Alyenbaawi
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Medical Laboratories, Majmaah University, Majmaah 11952, Saudi Arabia
| | - W. Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Sue-Ann Mok
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
150
|
Jiang S, Bhaskar K. Degradation and Transmission of Tau by Autophagic-Endolysosomal Networks and Potential Therapeutic Targets for Tauopathy. Front Mol Neurosci 2020; 13:586731. [PMID: 33177989 PMCID: PMC7596180 DOI: 10.3389/fnmol.2020.586731] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/24/2020] [Indexed: 01/21/2023] Open
Abstract
Tauopathies are a class of neurodegenerative diseases, including Alzheimer’s disease (AD), Frontotemporal Dementia (FTD), Progressive Supranuclear Palsy (PSP), Corticobasal Degeneration (CBD), and many others where microtubule-associated protein tau (MAPT or tau) is hyperphosphorylated and aggregated to form insoluble paired helical filaments (PHFs) and ultimately neurofibrillary tangles (NFTs). Autophagic-endolysosomal networks (AELN) play important roles in tau clearance. Excessive soluble neurotoxic forms of tau and tau hyperphosphorylated at specific sites are cleared through the ubiquitin-proteasome system (UPS), Chaperon-mediated Autophagy (CMA), and endosomal microautophagy (e-MI). On the other hand, intra-neuronal insoluble tau aggregates are often degraded within lysosomes by macroautophagy. AELN defects have been observed in AD, FTD, CBD, and PSP, and lysosomal dysfunction was shown to promote the cleavage and neurotoxicity of tau. Moreover, several AD risk genes (e.g., PICALM, GRN, and BIN1) have been associated with dysregulation of AELN in the late-onset sporadic AD. Conversely, tau dissociation from microtubules interferes with retrograde transport of autophagosomes to lysosomes, and that tau fragments can also lead to lysosomal dysfunction. Recent studies suggest that tau is not merely an intra-neuronal protein, but it can be released to brain parenchyma via extracellular vesicles, like exosomes and ectosomes, and thus spread between neurons. Extracellular tau can also be taken up by microglial cells and astrocytes, either being degraded through AELN or propagated via exosomes. This article reviews the complex roles of AELN in the degradation and transmission of tau, potential diagnostic/therapeutic targets and strategies based on AELN-mediated tau clearance and propagation, and the current state of drug development targeting AELN and tau against tauopathies.
Collapse
Affiliation(s)
- Shanya Jiang
- Department of Molecular Genetics and Microbiology, The University of New Mexico, Albuquerque, NM, United States
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, The University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|