1
|
Firdaus Z, Li X. Epigenetic Explorations of Neurological Disorders, the Identification Methods, and Therapeutic Avenues. Int J Mol Sci 2024; 25:11658. [PMID: 39519209 PMCID: PMC11546397 DOI: 10.3390/ijms252111658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Neurodegenerative disorders are major health concerns globally, especially in aging societies. The exploration of brain epigenomes, which consist of multiple forms of DNA methylation and covalent histone modifications, offers new and unanticipated perspective into the mechanisms of aging and neurodegenerative diseases. Initially, chromatin defects in the brain were thought to be static abnormalities from early development associated with rare genetic syndromes. However, it is now evident that mutations and the dysregulation of the epigenetic machinery extend across a broader spectrum, encompassing adult-onset neurodegenerative diseases. Hence, it is crucial to develop methodologies that can enhance epigenetic research. Several approaches have been created to investigate alterations in epigenetics on a spectrum of scales-ranging from low to high-with a particular focus on detecting DNA methylation and histone modifications. This article explores the burgeoning realm of neuroepigenetics, emphasizing its role in enhancing our mechanistic comprehension of neurodegenerative disorders and elucidating the predominant techniques employed for detecting modifications in the epigenome. Additionally, we ponder the potential influence of these advancements on shaping future therapeutic approaches.
Collapse
Affiliation(s)
- Zeba Firdaus
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
2
|
Sindi G, Ismael S, Uddin R, Slepchenko KG, Colvin RA, Lee D. Endogenous tau released from human ReNCell VM cultures by neuronal activity is phosphorylated at multiple sites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.597022. [PMID: 38854111 PMCID: PMC11160771 DOI: 10.1101/2024.06.02.597022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Tau is an intracellular protein but also known to be released into the extracellular fluid. Tau release mechanisms have drawn intense attention as these are known to play a key role in Alzheimer's disease (AD) pathology. However, tau can also be released under physiological conditions although its physiological function and release mechanisms have been poorly characterized, especially in human neuronal cells. We investigated endogenous tau release in ReNCell VM, a human neuroprogenitor cell line, under physiological conditions and found that tau is spontaneously released from cells. To study activity-dependent release of endogenous tau, human ReNCell VM culture was stimulated by 100μM AMPA or 50mM KCl for one-hour, tau was actively released to the culture medium. The released tau was highly phosphorylated at nine phosphorylation sites (pSites) detected by phospho-specific tau antibodies including AT270 (T175/T181), AT8 (S202/T205), AT100 (T212/S214), AT180 (T231), and PHF-1 (S396/S404), showing that these pSites are important for activity-dependent tau release from human ReNCell VM. Intracellular tau showed various phosphorylation status across these sites, with AT270 and PHF-1 highly phosphorylated while AT8 and AT180 were minimally phosphorylated, suggesting that AT8 and AT180 pSites exhibit a propensity for secretion rather than being retained intracellularly. This activity-dependent tau release was significantly decreased by inhibition of GSK-3β, demonstrating that GSK3β-dependent phosphorylation of tau plays an important role in its release by neuronal activity. In this study, we showed that ReNCell VM serves as a valuable model for studying endogenous physiological tau release. Further, ReNCell model can be also used to study pathological release of human tau that will contribute to our understanding of the progression of AD and related dementias.
Collapse
Affiliation(s)
| | - Sazan Ismael
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Reaz Uddin
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Kira G. Slepchenko
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Robert A. Colvin
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Daewoo Lee
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
3
|
Ellis MJ, Lekka C, Holden KL, Tulmin H, Seedat F, O'Brien DP, Dhayal S, Zeissler ML, Knudsen JG, Kessler BM, Morgan NG, Todd JA, Richardson SJ, Stefana MI. Identification of high-performing antibodies for the reliable detection of Tau proteoforms by Western blotting and immunohistochemistry. Acta Neuropathol 2024; 147:87. [PMID: 38761203 PMCID: PMC11102361 DOI: 10.1007/s00401-024-02729-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 05/20/2024]
Abstract
Antibodies are essential research tools whose performance directly impacts research conclusions and reproducibility. Owing to its central role in Alzheimer's disease and other dementias, hundreds of distinct antibody clones have been developed against the microtubule-associated protein Tau and its multiple proteoforms. Despite this breadth of offer, limited understanding of their performance and poor antibody selectivity have hindered research progress. Here, we validate a large panel of Tau antibodies by Western blot (79 reagents) and immunohistochemistry (35 reagents). We address the reagents' ability to detect the target proteoform, selectivity, the impact of protein phosphorylation on antibody binding and performance in human brain samples. While most antibodies detected Tau at high levels, many failed to detect it at lower, endogenous levels. By WB, non-selective binding to other proteins affected over half of the antibodies tested, with several cross-reacting with the related MAP2 protein, whereas the "oligomeric Tau" T22 antibody reacted with monomeric Tau by WB, thus calling into question its specificity to Tau oligomers. Despite the presumption that "total" Tau antibodies are agnostic to post-translational modifications, we found that phosphorylation partially inhibits binding for many such antibodies, including the popular Tau-5 clone. We further combine high-sensitivity reagents, mass-spectrometry proteomics and cDNA sequencing to demonstrate that presumptive Tau "knockout" human cells continue to express residual protein arising through exon skipping, providing evidence of previously unappreciated gene plasticity. Finally, probing of human brain samples with a large panel of antibodies revealed the presence of C-term-truncated versions of all main Tau brain isoforms in both control and tauopathy donors. Ultimately, we identify a validated panel of Tau antibodies that can be employed in Western blotting and/or immunohistochemistry to reliably detect even low levels of Tau expression with high selectivity. This work represents an extensive resource that will enable the re-interpretation of published data, improve reproducibility in Tau research, and overall accelerate scientific progress.
Collapse
Affiliation(s)
- Michael J Ellis
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Christiana Lekka
- Islet Biology Group, Department of Clinical & Biomedical Sciences, Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter, RILD Building, Exeter, UK
| | - Katie L Holden
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Hanna Tulmin
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Faheem Seedat
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Women's Centre, University of Oxford, John Radcliffe Hospital, Level 3, Oxford, UK
| | - Darragh P O'Brien
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Shalinee Dhayal
- Islet Biology Group, Department of Clinical & Biomedical Sciences, Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter, RILD Building, Exeter, UK
| | - Marie-Louise Zeissler
- Islet Biology Group, Department of Clinical & Biomedical Sciences, Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter, RILD Building, Exeter, UK
| | - Jakob G Knudsen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Oxford, Radcliffe, UK
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Benedikt M Kessler
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Noel G Morgan
- Islet Biology Group, Department of Clinical & Biomedical Sciences, Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter, RILD Building, Exeter, UK
| | - John A Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Sarah J Richardson
- Islet Biology Group, Department of Clinical & Biomedical Sciences, Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter, RILD Building, Exeter, UK
| | - M Irina Stefana
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK.
| |
Collapse
|
4
|
Hivare P, Mujmer K, Swarup G, Gupta S, Bhatia D. Endocytic pathways of pathogenic protein aggregates in neurodegenerative diseases. Traffic 2023; 24:434-452. [PMID: 37392160 DOI: 10.1111/tra.12906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 05/14/2023] [Accepted: 06/11/2023] [Indexed: 07/03/2023]
Abstract
Endocytosis is the fundamental uptake process through which cells internalize extracellular materials and species. Neurodegenerative diseases (NDs) are characterized by a progressive accumulation of intrinsically disordered protein species, leading to neuronal death. Misfolding in many proteins leads to various NDs such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS) and other disorders. Despite the significance of disordered protein species in neurodegeneration, their spread between cells and the cellular uptake of extracellular species is not entirely understood. This review discusses the major internalization mechanisms of the different conformer species of these proteins and their endocytic mechanisms. We briefly introduce the broad types of endocytic mechanisms found in cells and then summarize what is known about the endocytosis of monomeric, oligomeric and aggregated conformations of tau, Aβ, α-Syn, Huntingtin, Prions, SOD1, TDP-43 and other proteins associated with neurodegeneration. We also highlight the key players involved in internalizing these disordered proteins and the several techniques and approaches to identify their endocytic mechanisms. Finally, we discuss the obstacles involved in studying the endocytosis of these protein species and the need to develop better techniques to elucidate the uptake mechanisms of a particular disordered protein species.
Collapse
Affiliation(s)
- Pravin Hivare
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| | - Kratika Mujmer
- Center for Brain and Cognitive Sciences, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| | - Gitanjali Swarup
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| | - Sharad Gupta
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
- Center for Biomedical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| | - Dhiraj Bhatia
- Biological Engineering Discipline, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
- Center for Biomedical Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| |
Collapse
|
5
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
6
|
Jafarzadeh J, Payahoo L, Yousefi M, Barzegar A. The comprehensive mechanistic insight into the effects of vitamin D on dementia – a review. NUTRITION & FOOD SCIENCE 2022; 52:698-721. [DOI: 10.1108/nfs-08-2021-0256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
PurposeThis paper aims to depict the mechanistic role of vitamin D on dementia prevention, relief of the severity and the complication of the disease. All papers indexed in scientific databases, including Scopus, Elsevier, PubMed, Embase and Google Scholar between 2000 and 2021 were extracted and discussed. To present the mechanistic role of vitamin D in declining the severity of dementia, keywords including dementia, vitamin D, oxidative stress, inflammation, amyloid beta-Peptides were used.Design/methodology/approachDementia is a prevalent cognitive disorder worldwide, especially in elderly people, which is accompanied by serious disabilities. Besides genetic, biological and lifestyle factors are involved in the incidence of dementia. An unhealthy diet along with micronutrient deficiencies are among modifiable factors. Vitamin D is one of the important micronutrients in brain health. Besides the involvement in gene expression, bone mineralization, apoptosis, inflammation, skeletal maturation, neurotropic action and hemostasis of phosphate and calcium, vitamin D also exerts neuroprotective effects via genomic and non-genomic pathways.FindingsVitamin D up-regulates the expression of various genes involved in dementia incidence via various mechanisms. Decreasing oxidative stress and the neuro-inflammatory cytokines levels, regulation of the expression of alternated Proteins including Tau and Amyloid-ß, calcium homeostasis in the central nervous system and also vascular are considered main mechanisms.Originality/valueConsidering the importance of diet in preventing dementia, adherence to a healthy diet that provides essential nutrients to brain function seems to be urgent. Controlling serum levels of vitamin D periodically and providing vitamin D by related sources or supplements, if there is a deficiency, is recommended. Future studies are needed to clarify other related mechanisms.
Collapse
|
7
|
Caamaño-Moreno M, Gargini R. Tauopathies: the role of tau in cellular crosstalk and synaptic dysfunctions. Neuroscience 2022; 518:38-53. [PMID: 35272005 DOI: 10.1016/j.neuroscience.2022.02.034] [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: 12/30/2021] [Revised: 02/20/2022] [Accepted: 02/28/2022] [Indexed: 10/18/2022]
Abstract
Tauopathies are a group of neurodegenerative diseases among which are many of the most prevalent and with higher incidence worldwide, such as Alzheimer's disease (AD). According to the World Health Organization, this set of diseases will continue to increase their incidence, affecting millions of people by 2050. All of them are characterized by aberrant aggregation of tau protein in neurons and glia that are distributed in different brain regions according to their susceptibility. Numerous studies reveal that synaptic regulation not only has a neuronal component, but glia plays a fundamental role in it beyond its neuroinflammatory role. Despite this, it has not been emphasized how the glial inclusions of tau in this cell type directly affect this and many other essential functions, whose alterations have been related to the development of tauopathies. In this way, this review shows how tau inclusions in glia influence the synaptic dysfunctions that result in the cognitive symptoms characteristic of tauopathies. Thus, the mechanisms affected by inclusions in neurons, astrocytes, and oligodendrocytes are unraveled.
Collapse
Affiliation(s)
- Marta Caamaño-Moreno
- Instituto de investigaciones Biomédicas I+12, Hospital 12 de Octubre, Madrid, Spain
| | - Ricardo Gargini
- Instituto de investigaciones Biomédicas I+12, Hospital 12 de Octubre, Madrid, Spain; Neurooncology Unit, Instituto de Salud Carlos III-UFIEC, 28220 Madrid, Spain.
| |
Collapse
|
8
|
Ismael S, Sindi G, Colvin RA, Lee D. Activity-dependent release of phosphorylated human tau from Drosophila neurons in primary culture. J Biol Chem 2021; 297:101108. [PMID: 34473990 PMCID: PMC8455371 DOI: 10.1016/j.jbc.2021.101108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 08/11/2021] [Accepted: 08/19/2021] [Indexed: 11/23/2022] Open
Abstract
Neuronal activity can enhance tau release and thus accelerate tauopathies. This activity-dependent tau release can be used to study the progression of tau pathology in Alzheimer's disease (AD), as hyperphosphorylated tau is implicated in AD pathogenesis and related tauopathies. However, our understanding of the mechanisms that regulate activity-dependent tau release from neurons and the role that tau phosphorylation plays in modulating activity-dependent tau release is still rudimentary. In this study, Drosophila neurons in primary culture expressing human tau (hTau) were used to study activity-dependent tau release. We found that hTau release was markedly increased by 50 mM KCl treatment for 1 h. A similar level of release was observed using optogenetic techniques, where genetically targeted neurons were stimulated for 30 min using blue light (470 nm). Our results showed that activity-dependent release of phosphoresistant hTauS11A was reduced when compared with wildtype hTau. In contrast, release of phosphomimetic hTauE14 was increased upon activation. We found that released hTau was phosphorylated in its proline-rich and C-terminal domains using phosphorylation site-specific tau antibodies (e.g., AT8). Fold changes in detectable levels of total or phosphorylated hTau in cell lysates or following immunopurification from conditioned media were consistent with preferential release of phosphorylated hTau after light stimulation. This study establishes an excellent model to investigate the mechanism of activity-dependent hTau release and to better understand the role of phosphorylated tau release in the pathogenesis of AD since it relates to alterations in the early stage of neurodegeneration associated with increased neuronal activity.
Collapse
Affiliation(s)
- Sazan Ismael
- Neuroscience Program, Department of Biological Sciences, and Molecular and Cellular Biology Interdisciplinary Graduate Program, Ohio University, Athens, Ohio, USA
| | - Ghadir Sindi
- Neuroscience Program, Department of Biological Sciences, and Molecular and Cellular Biology Interdisciplinary Graduate Program, Ohio University, Athens, Ohio, USA
| | - Robert A Colvin
- Neuroscience Program, Department of Biological Sciences, and Molecular and Cellular Biology Interdisciplinary Graduate Program, Ohio University, Athens, Ohio, USA
| | - Daewoo Lee
- Neuroscience Program, Department of Biological Sciences, and Molecular and Cellular Biology Interdisciplinary Graduate Program, Ohio University, Athens, Ohio, USA.
| |
Collapse
|
9
|
Zhang H, Cao Y, Ma L, Wei Y, Li H. Possible Mechanisms of Tau Spread and Toxicity in Alzheimer's Disease. Front Cell Dev Biol 2021; 9:707268. [PMID: 34395435 PMCID: PMC8355602 DOI: 10.3389/fcell.2021.707268] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/09/2021] [Indexed: 12/31/2022] Open
Abstract
Tau is a protein that associates with microtubules (MTs) and promotes their assembly and stability. The protein loses its ability to bind MTs in tauopathies, and detached tau can misfold and induce the pathological changes that characterize Alzheimer’s disease (AD). A growing body of evidence indicates that tauopathies can spread between cells or connected regions. Pathological tau transmission in the brain of patients with AD and other tauopathies is due to the spread of various tau species along neuroanatomically connected regions in a “prion-like” manner. This complex process involves multiple steps of secretion, cellular uptake, transcellular transfer, and/or seeding, but the precise mechanisms of tau pathology propagation remain unclear. This review summarizes the current evidence on the nature of propagative tau species and the possible steps involved in the process of tau pathology spread, including detachment from MTs, degradations, and secretion, and discusses the different mechanisms underlying the spread of tau pathology.
Collapse
Affiliation(s)
- Huiqin Zhang
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Ma
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yun Wei
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Li
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Esteras N, Kopach O, Maiolino M, Lariccia V, Amoroso S, Qamar S, Wray S, Rusakov DA, Jaganjac M, Abramov AY. Mitochondrial ROS control neuronal excitability and cell fate in frontotemporal dementia. Alzheimers Dement 2021; 18:318-338. [PMID: 34057756 DOI: 10.1002/alz.12394] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/15/2021] [Accepted: 05/06/2021] [Indexed: 12/29/2022]
Abstract
INTRODUCTION The second most common form of early-onset dementia-frontotemporal dementia (FTD)-is often characterized by the aggregation of the microtubule-associated protein tau. Here we studied the mechanism of tau-induced neuronal dysfunction in neurons with the FTD-related 10+16 MAPT mutation. METHODS Live imaging, electrophysiology, and redox proteomics were used in 10+16 induced pluripotent stem cell-derived neurons and a model of tau spreading in primary cultures. RESULTS Overproduction of mitochondrial reactive oxygen species (ROS) in 10+16 neurons alters the trafficking of specific glutamate receptor subunits via redox regulation. Increased surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA) receptors containing GluA1 and NR2B subunits leads to impaired glutamatergic signaling, calcium overload, and excitotoxicity. Mitochondrial antioxidants restore the altered response and prevent neuronal death. Importantly, extracellular 4R tau induces the same pathological response in healthy neurons, thus proposing a mechanism for disease propagation. DISCUSSION These results demonstrate mitochondrial ROS modulate glutamatergic signaling in FTD, and suggest a new therapeutic strategy.
Collapse
Affiliation(s)
- Noemí Esteras
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Olga Kopach
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Marta Maiolino
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche,", Ancona, Italy
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche,", Ancona, Italy
| | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche,", Ancona, Italy
| | - Seema Qamar
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Selina Wray
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Dmitri A Rusakov
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Morana Jaganjac
- Qatar Analytics & BioResearch Lab, Anti-Doping Lab Qatar, Doha, Qatar.,Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Andrey Y Abramov
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
11
|
De La-Rocque S, Moretto E, Butnaru I, Schiavo G. Knockin' on heaven's door: Molecular mechanisms of neuronal tau uptake. J Neurochem 2020; 156:563-588. [PMID: 32770783 PMCID: PMC8432157 DOI: 10.1111/jnc.15144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022]
Abstract
Since aggregates of the microtubule‐binding protein tau were found to be the main component of neurofibrillary tangles more than 30 years ago, their contribution to neurodegeneration in Alzheimer's disease (AD) and tauopathies has become well established. Recent work shows that both tau load and its distribution in the brain of AD patients correlate with cognitive decline more closely compared to amyloid plaque deposition. In addition, the amyloid cascade hypothesis has been recently challenged because of disappointing results of clinical trials designed to treat AD by reducing beta‐amyloid levels, thus fuelling a renewed interest in tau. There is now robust evidence to indicate that tau pathology can spread within the central nervous system via a prion‐like mechanism following a stereotypical pattern, which can be explained by the trans‐synaptic inter‐neuronal transfer of pathological tau. In the receiving neuron, tau has been shown to take multiple routes of internalisation, which are partially dependent on its conformation and aggregation status. Here, we review the emerging mechanisms proposed for the uptake of extracellular tau in neurons and the requirements for the propagation of its pathological conformers, addressing how they gain access to physiological tau monomers in the cytosol. Furthermore, we highlight some of the key mechanistic gaps of the field, which urgently need to be addressed to expand our understanding of tau propagation and lead to the identification of new therapeutic strategies for tauopathies.
Collapse
Affiliation(s)
- Samantha De La-Rocque
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edoardo Moretto
- UK Dementia Research Institute, University College London, London, UK
| | - Ioana Butnaru
- UK Dementia Research Institute, University College London, London, UK
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
12
|
Loeffler DA, Aasly JO, LeWitt PA, Coffey MP. What Have We Learned from Cerebrospinal Fluid Studies about Biomarkers for Detecting LRRK2 Parkinson's Disease Patients and Healthy Subjects with Parkinson's-Associated LRRK2 Mutations? JOURNAL OF PARKINSONS DISEASE 2020; 9:467-488. [PMID: 31322581 PMCID: PMC6700639 DOI: 10.3233/jpd-191630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common known cause of autosomal dominant Parkinson’s disease (PD) and sporadic PD (sPD). The clinical presentation of LRRK2 PD is similar to sPD, and except for genetic testing, no biochemical or imaging markers can differentiate LRRK2 PD from sPD. Discovery of such biomarkers could indicate neuropathological mechanisms that are unique to or increased in LRRK2 PD. This review discusses findings in 17 LRRK2 - related CSF studies found on PubMed. Most of these studies compared analyte concentrations between four diagnostic groups: LRRK2 PD patients, sPD patients, asymptomatic control subjects carrying PD-associated LRRK2 mutations (LRRK2 CTL), and healthy control subjects lacking LRRK2 mutations (CTL). Analytes examined in these studies included Aβ1-42, tau, α-synuclein, oxidative stress markers, autophagy-related proteins, pteridines, neurotransmitter metabolites, exosomal LRRK2 protein, RNA species, inflammatory cytokines, mitochondrial DNA (mtDNA), and intermediary metabolites. FINDINGS: Pteridines, α-synuclein, mtDNA, 5-hydroxyindolacetic acid, β-D-glucose, lamp2, interleukin-8, and vascular endothelial growth factor were suggested to differentiate LRRK2 PD from sPD patients; 8-hydroxy-2’-deoxyguanosine (8-OHdG), 8-isoprostane (8-ISO), 2-hydroxybutyrate, mtDNA, lamp2, and neopterin may differentiate between LRRK2 CTL and LRRK2 PD subjects; and soluble oligomeric α-synuclein, 8-OHdG, and 8-ISO might differentiate LRRK2 CTL from CTL subjects. CONCLUSIONS: The low numbers of investigations of each analyte, small sample sizes, and methodological differences limit conclusions that can be drawn from these studies. Further investigations are indicated to determine the validity of the analytes identified in these studies as possible biomarkers for LRRK2 PD patients and/or LRRK2 CTL subjects.
Collapse
Affiliation(s)
- David A Loeffler
- Department of Neurology, Beaumont Hospital-Royal Oak, Beaumont Health, Royal Oak, MI, USA
| | - Jan O Aasly
- Department of Neurology, St. Olav's Hospital, Trondheim, Norway
| | - Peter A LeWitt
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mary P Coffey
- Department of Biostatistics, Beaumont Hospital-Royal Oak, Beaumont Health, Royal Oak, MI, USA
| |
Collapse
|
13
|
A system-level approach to investigate alloxan-induced toxicity in microtubule-binding protein to lead type 2 diabetes mellitus. Mol Divers 2020; 25:911-924. [PMID: 32219736 DOI: 10.1007/s11030-020-10075-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/12/2020] [Indexed: 12/24/2022]
Abstract
Microtubule-associated protein tau (MAPT) is a key protein, which is mainly identified as an essential factor for microtubule dynamics and neuronal outgrowth. Though tau has several functions, regulation of insulin signaling is one among them to control type 2 diabetes. Abnormal expression of tau protein leads to hyperphosphorylation and is known as tauopathies. The presence of alloxan occurs in refined wheat flour, especially in various baking products such as parotta, a well-known South Indian dish. In this study, the reduced form of alloxan called dialuric acid can enter the beta cells of islets of Langerhans and binds MAPT to induce toxicity by hyperphosphorylating the tau protein, which ultimately causes destruction to pancreatic beta cells, and it leads to diabetes mellitus. Here, the toxic effects of dialuric acid targeting MAPT through in silico computational predictions have been investigated. The 3D structure of MAPT protein was constructed through I-Tasser, and it has been refined and validated by GalaxyRefine and PROCHECK. The structure of ligand was retrieved from PubChem. Molecular docking was accomplished by AutoDock 4.2 software, and the results indicate the strong binding affinity between dialuric acid and MAPT protein, and it showed a binding free energy (∆G) of - 3.72 kcal/mol. Dialuric acid binds with the active region SER 232 of MAPT whereby it hyperphosphorylates the protein to become toxic. Also, ADMET results strongly suggest that the compound dialuric acid possesses toxic property, and similarly, Ames test confirmed that it was found to be mutagenic. Thus, our results strongly revealed that dialuric acid was found to be toxic which could be able to damage the beta cells of the pancreas and abates insulin signaling, and finally, it leads to DM.
Collapse
|
14
|
From the prion-like propagation hypothesis to therapeutic strategies of anti-tau immunotherapy. Acta Neuropathol 2020; 139:3-25. [PMID: 31686182 PMCID: PMC6942016 DOI: 10.1007/s00401-019-02087-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/18/2019] [Accepted: 10/19/2019] [Indexed: 12/15/2022]
Abstract
The term “propagon” is used to define proteins that may transmit misfolding in vitro, in tissues or in organisms. Among propagons, misfolded tau is thought to be involved in the pathogenic mechanisms of various “tauopathies” that include Alzheimer's disease, progressive supranuclear palsy, and argyrophilic grain disease. Here, we review the available data in the literature and point out how the prion-like tau propagation has been extended from Alzheimer's disease to tauopathies. First, in Alzheimer’s disease, the progression of tau aggregation follows stereotypical anatomical stages which may be considered as spreading. The mechanisms of the propagation are now subject to intensive and controversial research. It has been shown that tau may be secreted in the interstitial fluid in an active manner as reflected by high and constant concentration of extracellular tau during Alzheimer’s pathology. Animal and cell models have been devised to mimic tau seeding and propagation, and despite their limitations, they have further supported to the prion-like propagation hypothesis. Finally, such new ways of thinking have led to different therapeutic strategies in anti-tau immunotherapy among tauopathies and have stimulated new clinical trials. However, it appears that the prion-like propagation hypothesis mainly relies on data obtained in Alzheimer’s disease. From this review, it appears that further studies are needed (1) to characterize extracellular tau species, (2) to find the right pathological tau species to target, (3) to follow in vivo tau pathology by brain imaging and biomarkers and (4) to interpret current clinical trial results aimed at reducing the progression of these pathologies. Such inputs will be essential to have a comprehensive view of these promising therapeutic strategies in tauopathies.
Collapse
|
15
|
Webster JM, Darling AL, Uversky VN, Blair LJ. Small Heat Shock Proteins, Big Impact on Protein Aggregation in Neurodegenerative Disease. Front Pharmacol 2019; 10:1047. [PMID: 31619995 PMCID: PMC6759932 DOI: 10.3389/fphar.2019.01047] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
Misfolding, aggregation, and aberrant accumulation of proteins are central components in the progression of neurodegenerative disease. Cellular molecular chaperone systems modulate proteostasis, and, therefore, are primed to influence aberrant protein-induced neurotoxicity and disease progression. Molecular chaperones have a wide range of functions from facilitating proper nascent folding and refolding to degradation or sequestration of misfolded substrates. In disease states, molecular chaperones can display protective or aberrant effects, including the promotion and stabilization of toxic protein aggregates. This seems to be dependent on the aggregating protein and discrete chaperone interaction. Small heat shock proteins (sHsps) are a class of molecular chaperones that typically associate early with misfolded proteins. These interactions hold proteins in a reversible state that helps facilitate refolding or degradation by other chaperones and co-factors. These sHsp interactions require dynamic oligomerization state changes in response to diverse cellular triggers and, unlike later steps in the chaperone cascade of events, are ATP-independent. Here, we review evidence for modulation of neurodegenerative disease-relevant protein aggregation by sHsps. This includes data supporting direct physical interactions and potential roles of sHsps in the stewardship of pathological protein aggregates in brain. A greater understanding of the mechanisms of sHsp chaperone activity may help in the development of novel therapeutic strategies to modulate the aggregation of pathological, amyloidogenic proteins. sHsps-targeting strategies including modulators of expression or post-translational modification of endogenous sHsps, small molecules targeted to sHsp domains, and delivery of engineered molecular chaperones, are also discussed.
Collapse
Affiliation(s)
- Jack M Webster
- Department of Molecular Medicine, USF Byrd Institute, University of South Florida, Tampa, FL, United States
| | - April L Darling
- Department of Molecular Medicine, USF Byrd Institute, University of South Florida, Tampa, FL, United States
| | - Vladimir N Uversky
- Department of Molecular Medicine, USF Byrd Institute, University of South Florida, Tampa, FL, United States
| | - Laura J Blair
- Department of Molecular Medicine, USF Byrd Institute, University of South Florida, Tampa, FL, United States
| |
Collapse
|
16
|
Fichou Y, Al-Hilaly YK, Devred F, Smet-Nocca C, Tsvetkov PO, Verelst J, Winderickx J, Geukens N, Vanmechelen E, Perrotin A, Serpell L, Hanseeuw BJ, Medina M, Buée L, Landrieu I. The elusive tau molecular structures: can we translate the recent breakthroughs into new targets for intervention? Acta Neuropathol Commun 2019; 7:31. [PMID: 30823892 PMCID: PMC6397507 DOI: 10.1186/s40478-019-0682-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 02/20/2019] [Indexed: 12/11/2022] Open
Abstract
Insights into tau molecular structures have advanced significantly in recent years. This field has been the subject of recent breakthroughs, including the first cryo-electron microscopy structures of tau filaments from Alzheimer’s and Pick’s disease inclusions, as well as the structure of the repeat regions of tau bound to microtubules. Tau structure covers various species as the tau protein itself takes many forms. We will here address a range of studies that help to define the many facets of tau protein structures and how they translate into pathogenic forms. New results shed light on previous data that need now to be revisited in order to up-date our knowledge of tau molecular structure. Finally, we explore how these data can contribute the important medical aspects of this research - diagnosis and therapeutics.
Collapse
|