1
|
Men Y, Hirayama S, Ao S, Sakurai Y, Shibata Y, Lo M, Sato Y, Murata S. ESCRT-I and PTPN23 mediate microautophagy of ubiquitylated tau aggregates. J Cell Biol 2025; 224:e202406120. [PMID: 40197510 PMCID: PMC11977513 DOI: 10.1083/jcb.202406120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 12/09/2024] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
Protein aggregates are degraded by both the autophagy-lysosomal and the ubiquitin-proteasome pathways. Macroautophagy and microautophagy, two forms of the autophagy-lysosomal pathway, are widely conserved across eukaryotes. While macroautophagy has been extensively studied in the context of degradation of protein aggregates, microautophagy remains less explored. Here, we identify the UBAP1-containing ESCRT-I complex and PTPN23 as new regulators for degradation of aggregated proteins through an unbiased genome-wide CRISPR knockout screen, using a cell line expressing tau repeat domain (tauRD) aggregates. ESCRT-I recognizes ubiquitylated tauRD via the UEV domain of TSG101. The accessory protein PTPN23, instead of ESCRT-II, bridges ESCRT-I and ESCRT-III to complete the endosomal microautophagy of ubiquitylated tauRD aggregates. Our results uncover the molecular mechanism underlying the degradation of tau aggregates by endosomal microautophagy.
Collapse
Affiliation(s)
- Yusen Men
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shoshiro Hirayama
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shinpei Ao
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakurai
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuri Shibata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Megan Lo
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yusuke Sato
- Department of Chemistry and Biotechnology and Center for Research on Green Sustainable Chemistry, Graduate School of Engineering, Tottori University, Tottori, Japan
| | - Shigeo Murata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Van Alstyne M, Pratt J, Parker R. Diverse influences on tau aggregation and implications for disease progression. Genes Dev 2025; 39:555-581. [PMID: 40113250 PMCID: PMC12047666 DOI: 10.1101/gad.352551.124] [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] [Indexed: 03/22/2025]
Abstract
Tau is an intrinsically disordered protein that accumulates in fibrillar aggregates in neurodegenerative diseases. The misfolding of tau can be understood as an equilibrium between different states and their propensity to form higher-order fibers, which is affected by several factors. First, modulation of the biochemical state of tau due to ionic conditions, post-translational modifications, cofactors, and interacting molecules or assemblies can affect the formation and structure of tau fibrils. Second, cellular processes impact tau aggregation through modulating stability, clearance, disaggregation, and transport. Third, through interactions with glial cells, the neuronal microenvironment can affect intraneuronal conditions with impacts on tau fibrilization and toxicity. Importantly, tau fibrils propagate through the brain via a "prion-like" manner, contributing to disease progression. This review highlights the biochemical and cellular pathways that modulate tau aggregation and discusses implications for pathobiology and tau-directed therapeutic approaches.
Collapse
Affiliation(s)
- Meaghan Van Alstyne
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado 80301, USA
- Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, Colorado 80301, USA
| | - James Pratt
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado 80301, USA
| | - Roy Parker
- Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado 80301, USA;
- Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, Colorado 80301, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado 80301, USA
| |
Collapse
|
3
|
Xiao X, Liu H, Yao R, Li Y, Liao X, Liu Y, Zhou Y, Wang J, Tang B, Jiao B, Li J, Shen L, Luo S. LMTK2 and CRB1 are two novel risk genes for Alzheimer's disease in Han Chinese. J Prev Alzheimers Dis 2025; 12:100087. [PMID: 39922756 DOI: 10.1016/j.tjpad.2025.100087] [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: 12/27/2024] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent neurodegenerative disease with a substantial genetic background. However, its underlying genetic architecture remains to be elucidated. METHODS In this study, we performed whole-exome sequencing in 282 familial and/or early-onset AD patients and 1086 cognitively normal controls in the Han Chinse populations. According to minor allele frequency, variants were divided into common variants (MAF ≥ 0.01) and rare variants (MAF < 0.01). Common variant-based association analysis and gene-based association test aggregating rare variants were performed by PLINK 1.9 and Sequence Kernel Association Test-Optimal, respectively. We replicated the significant results by using the same AD samples and controls from whole genome sequencing (n = 1879). Furthermore, we determined the functions of the novel AD risk genes in vitro. RESULTS Common variants association analysis revealed that APOE rs429358 reached statistical whole-exome significance. Gene-level aggregation testing identified that rare damaging variants in LMTK2 and CRB1 conferred risk to AD. All variants are located in highly conserved amino acid regions and are predicted to be damaging. Furthermore, functional studies showed that LMTK2 rare damaging variants (R234P and S974G) enhanced tau phosphorylation levels, tau aggregates formation, and Aβ generation. Meanwhile, the CRB1 Y556X variant caused incomplete translation of CRB1 protein and increased the Aβ42 level and Aβ42/Aβ40 ratio. CONCLUSION Our findings indicated that LMTK2 and CRB1 are two novel AD risk genes in Han Chinese, which may provide promising targets for diagnosis and intervention.
Collapse
Affiliation(s)
- Xuewen Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, PR China
| | - Hui Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Rui Yao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Yunni Li
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, PR China
| | - Xinxin Liao
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, PR China; Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Yingzi Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Yafang Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, PR China; Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, PR China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, PR China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, PR China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, PR China
| | - Jinchen Li
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, PR China; Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Changsha, PR China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, PR China
| | - Shilin Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, PR China; Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, PR China; Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, PR China.
| |
Collapse
|
4
|
Angera IJ, Xu X, Rajewski BH, Hallinan GI, Zhang X, Ghetti B, Vidal R, Jiang W, Del Valle JR. Macrocyclic β-arch peptides that mimic the structure and function of disease-associated tau folds. Nat Chem 2025:10.1038/s41557-025-01805-z. [PMID: 40307419 DOI: 10.1038/s41557-025-01805-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025]
Abstract
Tauopathies are a class of neurodegenerative disorders that feature tau protein aggregates in the brain. Misfolded tau has the capacity to seed the fibrillization of soluble tau, leading to the prion-like spread of aggregates. Within these filaments, tau protomers always exhibit a cross-β amyloid structure. However, distinct cross-β amyloid folds correlate with specific diseases. An understanding of how these conformations impact seeding activity remains elusive. Identifying the minimal epitopes required for transcellular propagation of tau aggregates represents a key step towards more relevant models of disease progression. Here we implement a diversity-oriented peptide macrocyclization approach towards miniature tau, or 'mini-tau', proteomimetics that can seed the aggregation of tau in engineered cells and primary neurons. Structural elucidation of one such seed-competent macrocycle reveals remarkable conformational congruence with core folds from patient-derived extracts of tau. The ability to impart β-arch form and function through peptide stapling has broad-ranging implications for the minimization and mimicry of pathological tau and other amyloid proteins that drive neurodegeneration.
Collapse
Affiliation(s)
- Isaac J Angera
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Xueyong Xu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Benjamin H Rajewski
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Grace I Hallinan
- Department of Pathology & Laboratory Medicine and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiaoqi Zhang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Bernardino Ghetti
- Department of Pathology & Laboratory Medicine and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ruben Vidal
- Department of Pathology & Laboratory Medicine and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wen Jiang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Juan R Del Valle
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
5
|
Chongtham A, Ramakrishnan A, Farinas M, Broekaart DWM, Seo JH, Zhu CW, Sano M, Shen L, Pereira AC. Neocortical tau propagation is a mediator of clinical heterogeneity in Alzheimer's disease. Mol Psychiatry 2025:10.1038/s41380-025-02998-y. [PMID: 40234685 DOI: 10.1038/s41380-025-02998-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025]
Abstract
Heterogeneity in progression of clinical dementia obstructs the general therapeutic potential of current treatments for Alzheimer's disease (AD). Though the mechanisms of this heterogeneity remain unclear, the characterization of bioactive tau species and factors that regulate their seeding behavior might give valuable insight as pathological tau is well correlated with cognitive impairment. Here, we conducted an innovative investigation into the molecular basis of widespread, connectivity-based tau propagation that begins in the inferior temporal gyrus (ITG) and spreads to neocortical areas such as the prefrontal cortex (PFC). Biochemical analysis of human postmortem ITG and PFC tissues revealed individual variability in tau seeding, which correlated with cognitive decline, particularly in the ITG, a region known for promoting accelerated tau propagation. Notably, this study presents the first evidence that site-specific phosphorylation and isoform composition of both aggregation-prone high-molecular-weight (HMW) tau and the relatively unexplored, yet potentially crucial in AD progression low-molecular-weight (LMW) tau significantly contribute to tau propagation and cognitive decline. Our findings underscore the importance of comprehensively considering diverse tau forms including both HMW and LMW tau species in understanding AD progression. Additionally, these results are the first to identify distinct morphological strains within the AD brain associated with differing seeding propensity, potentially enabling patient stratification based on their tau profile. Furthermore, RNA-seq analyses of gene expression patterns in the ITG revealed molecular heterogeneity associated with tau seeding potential. Patients with higher levels of seed-competent tau displayed greater impairments in synaptic and neural plasticity, and increased neuroinflammation. This multidisciplinary study offers novel insights into various molecular mechanisms driving AD progression, suggesting potential molecular targets for early intervention and improved patient subtyping, which is critical for developing precision medicine approaches.
Collapse
Affiliation(s)
- Anjalika Chongtham
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marissa Farinas
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diede W M Broekaart
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joon Ho Seo
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolyn W Zhu
- Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
- Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Sano
- James J. Peters VA Medical Center, Bronx, NY, USA
- Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ana C Pereira
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Sanford Grossman Interdisciplinary Program I Neural Circuitry and Immune Function, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
6
|
Domene-Serrano I, Cuadros R, García-Escudero V, Vallejo-Bedia F, Santa-María I, Vallés-Saiz L, Hernandez F, Avila J. Shapeshifter W-Tau Peptide Inhibits Tau Aggregation and Disintegrates Paired Helical Filaments. Biochemistry 2025; 64:1841-1851. [PMID: 40140976 PMCID: PMC12004447 DOI: 10.1021/acs.biochem.4c00809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/28/2025]
Abstract
Tauopathies comprise a range of neurodegenerative conditions characterized by the aberrant accumulation of tau protein clumps in the brain. These aggregates are formed by different tau splicing isoforms. Here, we analyzed the role of a specific intron-derived peptide called the W-Tau peptide on the polymerization-depolymerization of tau filaments. This peptide originates from a new isoform of the tau protein, named W-Tau, which is formed due to the retention of intron 12. AlphaFold3 (AF3)-based in silico investigations suggested that the W-Tau peptide interacts with tau monomers. Our in vitro experiments confirmed these predictions and showed that the W-Tau peptide inhibited tau aggregation. In addition, the W-Tau peptide disrupted preexisting paired helical filaments (PHFs) isolated from postmortem brain samples of patients with Alzheimer's disease, thereby supporting its potential therapeutic value. The effectiveness of the W-Tau peptide was demonstrated by the decrease in tau aggregation observed after cotransfection of the W-Tau peptide and PHF seeds, as demonstrated by analysis involving a fluorescence resonance energy transfer (FRET) cell biosensor. The W-Tau peptide breaks PHFs by selectively attaching to their ends, causing the structures to unwind and convert into circle-like formations. Considering the potential neuroprotective effects against tauopathies, the W-Tau isoform and its peptide are interesting candidates for future therapeutic interventions.
Collapse
Affiliation(s)
- Indalo Domene-Serrano
- Centro
de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid 28049, Spain
- Facultad
de Ciencias Experimentales, Universidad
Francisco de Vitoria, Pozuelo de Alarcon, Madrid 28223, Spain
| | - Raquel Cuadros
- Centro
de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid 28049, Spain
| | - Vega García-Escudero
- Centro
de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid 28049, Spain
- Departamento
de Anatomía, Histología y Neurociencia, School of Medicine, Autonoma de Madrid University (UAM), Arzobispo Morcillo, 4, Madrid 28029, Spain
| | | | - Ismael Santa-María
- Facultad
de Ciencias Experimentales, Universidad
Francisco de Vitoria, Pozuelo de Alarcon, Madrid 28223, Spain
| | - Laura Vallés-Saiz
- Centro
de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid 28049, Spain
| | - Félix Hernandez
- Centro
de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid 28049, Spain
| | - Jesús Avila
- Centro
de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid 28049, Spain
- Center
for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
7
|
Nguyen T, Panwar V, Jamale V, Perny A, Dusek C, Cai Q, Kapur P, Danuser G, Rajaram S. Autonomous learning of pathologists' cancer grading rules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643999. [PMID: 40166226 PMCID: PMC11956981 DOI: 10.1101/2025.03.18.643999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Deep learning (DL) algorithms have demonstrated remarkable proficiency in histopathology classification tasks, presenting an opportunity to discover disease-related features escaping visual inspection. However, the "black box" nature of DL obfuscates the basis of the classification. Here, we develop an algorithm for interpretable Deep Learning (IDL) that sheds light on the links between tissue morphology and cancer biology. We make use of a generative model trained to represent images via a combination of a semantic latent space and a noise vector to capture low level image details. We traversed the latent space so as to induce prototypical image changes associated with the disease state, which we identified via a second DL model. Applied to a dataset of clear cell renal cell carcinoma (ccRCC) tissue images the AI system pinpoints nuclear size and nucleolus density in tumor cells (but not other cell types) as the decisive features of tumor progression from grade 1 to grade 4 - mirroring the rules that have been used for decades in the clinic and are taught in textbooks. Moreover, the AI system posits a decrease in vasculature with increasing grade. While the association has been illustrated by some previous reports, the correlation is not part of currently implemented grading systems. These results indicate the potential of IDL to autonomously formalize the connection between the histopathological presentation of a disease and the underlying tissue architectural drivers.
Collapse
Affiliation(s)
- Thuong Nguyen
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vandana Panwar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vipul Jamale
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Averi Perny
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cecilia Dusek
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qi Cai
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Satwik Rajaram
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
8
|
Welikovitch LA, Mate de Gerando A, Khasnavis A, Bhavsar H, Meltzer JC, Buée L, Chibnik LB, Bussiere T, Hyman BT. Tau, synapse loss and gliosis progress in an Alzheimer's mouse model after amyloid-β immunotherapy. Brain 2025; 148:1316-1328. [PMID: 39504519 PMCID: PMC11967717 DOI: 10.1093/brain/awae345] [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: 06/10/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Preclinical studies assessing drugs for Alzheimer's disease (AD) are conducted in animal models that usually display only one neuropathological feature of AD, whereas patients present with a complex combination of comorbidities and neuropathologies. Importantly, it is well established that amyloid-β (Aβ) plaque and tau tangle accumulation interact in a phase-dependent manner, making it difficult to predict how targeting one might influence the other, as well as downstream degeneration. We developed a transgenic mouse model, APP/PS1xTau22, with progressive cortical Aβ deposition and hippocampal tau neurofibrillary inclusions, to investigate how both neuropathologies act jointly to bring about neural degeneration, synapse loss and glial phenotypes. We then assessed whether applying murine chimeric aducanumab, an anti-amyloid immunotherapy, could impact the synergistic relationship between amyloid and tau. Drug treatment resulted in a ∼70% reduction in Aβ deposition in hippocampal and cortical areas and produced a robust peri-plaque microglial and astrocytic response. Removing amyloid from the brain did not reverse or slow tau pathology or alter synapse loss. Our findings suggest that, once the interaction between amyloid and tau is set in motion, reducing plaque burden by Aβ immunotherapy may stimulate glial responses, but is insufficient to curb degenerative phenotypes in this model.
Collapse
Affiliation(s)
- Lindsay A Welikovitch
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Anastasie Mate de Gerando
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Anita Khasnavis
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Harshil Bhavsar
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Jonah C Meltzer
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition Lille, F-59000 Lille, France
| | - Lori B Chibnik
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | | | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
9
|
Sandhof CA, Murray HFB, Silva MC, Haggarty SJ. Targeted protein degradation with bifunctional molecules as a novel therapeutic modality for Alzheimer's disease & beyond. Neurotherapeutics 2025; 22:e00499. [PMID: 39638711 PMCID: PMC12047403 DOI: 10.1016/j.neurot.2024.e00499] [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: 09/16/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
Alzheimer's disease (AD) is associated with memory and cognitive impairment caused by progressive degeneration of neurons. The events leading to neuronal death are associated with the accumulation of aggregating proteins in neurons and glia of the affected brain regions, in particular extracellular deposition of amyloid plaques and intracellular formation of tau neurofibrillary tangles. Moreover, the accumulation of pathological tau proteoforms in the brain concurring with disease progression is a key feature of multiple neurodegenerative diseases, called tauopathies, like frontotemporal dementia (FTD) where autosomal dominant mutations in the tau encoding MAPT gene provide clear evidence of a causal role for tau dysfunction. Observations from disease models, post-mortem histology, and clinical evidence have demonstrated that pathological tau undergoes abnormal post-translational modifications, misfolding, oligomerization, changes in solubility, mislocalization, and intercellular spreading. Despite extensive research, there are few disease-modifying or preventative therapeutics for AD and none for other tauopathies. Challenges faced in tauopathy drug development include an insufficient understanding of pathogenic mechanisms of tau proteoforms, limited specificity of agents tested, and inadequate levels of brain exposure, altogether underscoring the need for innovative therapeutic modalities. In recent years, the development of experimental therapeutic modalities, such as targeted protein degradation (TPD) strategies, has shown significant and promising potential to promote the degradation of disease-causing proteins, thereby reducing accumulation and aggregation. Here, we review all modalities of TPD that have been developed to target tau in the context of AD and FTD, as well as other approaches that with innovation could be adapted for tau-specific TPD.
Collapse
Affiliation(s)
- C Alexander Sandhof
- Department of Neurology, Precision Therapeutics Unit, Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Heide F B Murray
- Department of Neurology, Precision Therapeutics Unit, Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - M Catarina Silva
- Department of Neurology, Precision Therapeutics Unit, Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Stephen J Haggarty
- Department of Neurology, Precision Therapeutics Unit, Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
10
|
Puangmalai N, Aday AE, Samples M, Bhatt N, Cascio FL, Marcatti M, Park SJ, Fung L, Jerez C, Penalva LO, Zhao Y, Hao H, Lugano D, Kayed R, Montalbano M. Pathogenic oligomeric Tau alters neuronal RNA processes through the formation of nuclear heteromeric amyloids with RNA-binding protein Musashi1. Prog Neurobiol 2025; 247:102742. [PMID: 40064283 PMCID: PMC11984483 DOI: 10.1016/j.pneurobio.2025.102742] [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: 10/15/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
Alzheimer's disease (AD) is marked by cytoplasmic proteinopathies, primarily involving misfolded Tau protein. Pathogenic Tau species, such as soluble oligomers and fibrils, disrupt RNA metabolism, though the mechanisms are unclear. Recent research indicates that RNA has a crucial role in Tau aggregation. Our study builds on this by noting significant co-deposition of RNA-Binding Proteins (RBPs) with Tau in AD and Frontotemporal dementia (FTLD) brains. Using molecular and cellular techniques, we investigate the interaction between RNA dynamics and Tau aggregation, focusing on the localization and aggregation of Tau and RBPs, particularly Musashi (MSI), within neuronal nuclei. Through cyto-fluorometric, biochemical, and cellular assays, we reveal the importance of Tau/RBP interplay in primary cortical neurons expressing wild-type and mutant Tau. Pathogenic Tau oligomers alter MSI protein localization and function, causing cytoplasmic and nuclear aggregation. Mass spectrometry of the MSI1 nuclear interactome in Tau models shows disrupted RNA metabolism pathways, including ribosomal biogenesis, RNA splicing, and protein folding. Moreover, RNA immunoprecipitation assay revealed a remarkable impact of mutant P301L Tau on MSI1 ability to bind RNA targets. These findings highlight potential targets for early neurodegenerative therapeutic interventions.
Collapse
Affiliation(s)
- Nicha Puangmalai
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| | - Abbigael E Aday
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| | - Madison Samples
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| | - Filippa Lo Cascio
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| | - Michela Marcatti
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| | - Suhyeorn J Park
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| | - Leiana Fung
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| | - Luiz O Penalva
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229, USA; Department of Cell Systems and Anatomy, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229, USA.
| | - Yingxin Zhao
- Department of Internal Medicine, The University of Texas Medical Branch, 301 University Blvd, Galveston TX 77555, USA.
| | - Haiping Hao
- Director, UTMB Next Gen Sequencing Core, Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | - Doreen Lugano
- KEMRI-Wellcome Trust Research Programme, P.O. Box 230, Kilifi Kenya.
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| | - Mauro Montalbano
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA; Departments of Neurology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA.
| |
Collapse
|
11
|
Morderer D, Wren MC, Liu F, Kouri N, Maistrenko A, Khalil B, Pobitzer N, Salemi MR, Phinney BS, Bu G, Zhao N, Dickson DW, Murray ME, Rossoll W. Probe-dependent Proximity Profiling (ProPPr) Uncovers Similarities and Differences in Phospho-Tau-Associated Proteomes Between Tauopathies. Mol Neurodegener 2025; 20:32. [PMID: 40082954 PMCID: PMC11905455 DOI: 10.1186/s13024-025-00817-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/25/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Tauopathies represent a diverse group of neurodegenerative disorders characterized by the abnormal aggregation of the microtubule-associated protein tau. Despite extensive research, the mechanisms underlying the diversity of neuronal and glial tau pathology in different tauopathies are poorly understood. While there is a growing understanding of tauopathy-specific differences in tau isoforms and fibrillar structures, the specific composition of heterogenous tau lesions remains unknown. Here we study the protein composition of tau aggregates in four major tauopathies: Alzheimer's disease (AD), corticobasal degeneration (CBD), Pick's disease (PiD), and progressive supranuclear palsy (PSP). METHODS We developed an approach for in situ proximity labeling and isolation of aggregate-associated proteins using glass slides with formalin-fixed paraffin-embedded (FFPE) human postmortem brain tissue, termed Probe-dependent Proximity Profiling (ProPPr). We used ProPPr for the analysis of proteomes associated with AT8-positive cellular lesions from frontal cortices. Isolated proximity proteomes were analyzed by data-independent acquisition mass spectrometry. Co-immunofluorescence staining and quantitative data analysis for selected proteins in human brain tissue was performed to further investigate associations with diverse tau pathologies. RESULTS Proteomics data analysis identified numerous common and tauopathy-specific proteins associated with phospho-tau aggregates. Extensive validations of candidates through quantitative immunofluorescence imaging of distinct aggregates across disease cases demonstrate successful implementation of ProPPr for unbiased discovery of aggregate-associated proteins in in human brain tissue. Our results reveal the association of retromer complex component vacuolar protein sorting-associated protein 35 (VPS35) and lysosome-associated membrane glycoprotein 2 (LAMP2) with specific types of phospho-tau lesions in tauopathies. Furthermore, we discovered a disease-specific association of certain proteins with distinct pathological lesions, including glycogen synthase kinase alpha (GSK3α), ferritin light chain (FTL), and the neuropeptide precursor VGF. Notably, the identification of FTL-positive microglia in CBD astrocytic plaques indicate their potential role in the pathogenesis of these lesions. CONCLUSIONS Our findings demonstrate the suitability of the ProPPr approach in FFPE brain tissue for unbiased discovery of local proteomes that provide valuable insights into the underlying proteomic landscape of tauopathies, shedding light on the molecular mechanisms underlying tau pathology. This first comprehensive characterization of tau-associated proteomes in a range of distinct tauopathies enhances our understanding of disease heterogeneity and mechanisms, informing strategies for the development of diagnostic biomarkers and targeted therapies.
Collapse
Affiliation(s)
- Dmytro Morderer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Melissa C Wren
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Feilin Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Naomi Kouri
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Bilal Khalil
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Nora Pobitzer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Brett S Phinney
- Proteomics Core, University of California Davis, Davis, CA, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Present address: Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | |
Collapse
|
12
|
Huang M, McEwan WA. Sensitive detection and propagation of brain-derived tau assemblies in HEK293-based wild-type tau seeding assays. J Biol Chem 2025; 301:108245. [PMID: 39880096 PMCID: PMC11910105 DOI: 10.1016/j.jbc.2025.108245] [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: 09/23/2024] [Revised: 12/28/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
The assembly of tau into filaments defines tauopathies, a group of neurodegenerative diseases including Alzheimer's disease (AD), Pick's disease (PiD), corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP). The seeded aggregation of tau has been modeled in cell culture using pro-aggregate modifications such as truncation of N- and C-termini and point mutations within the microtubule-binding repeat domain. This limits the applicability of research findings to sporadic disease, where aggregates contain wild-type, full-length tau. We describe a sensitive and specific biosensor assays for brain-derived tau species utilizing wild-type 0N3R and 0N4R tau expressed in HEK293 cells. We further generate a cell line that propagates AD-templated insoluble tau which is hyperphosphorylated at disease-relevant sites and retains a seeding profile similar to AD. We propose these systems as an advance over existing cell-based seeding assays, as they display specificity to the conformation and isoform composition of sporadic human disease.
Collapse
Affiliation(s)
- Melissa Huang
- UK Dementia Research Institute at the University of Cambridge, Department of Clinical Neurosciences, Cambridge, United Kingdom.
| | - William A McEwan
- UK Dementia Research Institute at the University of Cambridge, Department of Clinical Neurosciences, Cambridge, United Kingdom.
| |
Collapse
|
13
|
Wen T, Meng L, Liu H, Zhang Q, Dai L, Huang L, Dan L, Zhu K, Luo J, Zhang Z. Fibrinogen-tau Aggregates Exacerbate Tau Pathology and Memory Deficits in Alzheimer's Disease Model Mice. Neurosci Bull 2025:10.1007/s12264-025-01366-8. [PMID: 39971888 DOI: 10.1007/s12264-025-01366-8] [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: 07/08/2024] [Accepted: 10/30/2024] [Indexed: 02/21/2025] Open
Abstract
Vascular damage plays a significant role in the onset and progression of Alzheimer's disease (AD). However, the precise molecular mechanisms underlying the induction of neuronal injury by vascular damage remain unclear. The present study aimed to examine the impact of fibrinogen (Fg) on tau pathology. The results showed that Fg deposits in the brains of tau P301S transgenic mice interact with tau, enhancing the cytotoxicity of pathological tau aggregates and promoting tau phosphorylation and aggregation. Notably, Fg-modified tau fibrils caused enhanced neuronal apoptosis and synaptic damage compared to unmodified fibrils. Furthermore, intrahippocampal injection of Fg-modified tau fibrils worsened the tau pathology, neuroinflammation, synaptic damage, neuronal apoptosis, and cognitive dysfunction in tau P301S mice compared to controls. The present study provides compelling evidence linking Fg and tau, thereby connecting cerebrovascular damage to tau pathology in AD. Consequently, inhibiting Fg-mediated tau pathology could potentially impede the progression of AD.
Collapse
Affiliation(s)
- Tingting Wen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Han Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qian Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liqin Huang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liang Dan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Kedong Zhu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiaying Luo
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
14
|
Locskai LF, Gill T, Tan SAW, Burton AH, Alyenbaawi H, Burton EA, Allison WT. A larval zebrafish model of traumatic brain injury: optimizing the dose of neurotrauma for discovery of treatments and aetiology. Biol Open 2025; 14:bio060601. [PMID: 39936823 PMCID: PMC11849975 DOI: 10.1242/bio.060601] [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: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 02/13/2025] Open
Abstract
Traumatic brain injuries (TBI) are diverse with heterogeneous injury pathologies, which creates challenges for the clinical treatment and prevention of secondary pathologies such as post-traumatic epilepsy and subsequent dementias. To develop pharmacological strategies that treat TBI and prevent complications, animal models must capture the spectrum of TBI severity to better understand pathophysiological events that occur during and after injury. To address such issues, we improved upon our recent larval zebrafish TBI paradigm emphasizing titrating to different injury levels. We observed coordination between an increase in injury level and clinically relevant injury phenotypes including post-traumatic seizures (PTS) and tau aggregation. This preclinical TBI model is simple to implement, allows dosing of injury levels to model diverse pathologies, and can be scaled to medium- or high-throughput screening.
Collapse
Affiliation(s)
- Laszlo F. Locskai
- Department of Biological Sciences, University of Alberta, Edmonton AB, T6G 2E9, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton AB, T6G 2M8, Canada
| | - Taylor Gill
- Department of Biological Sciences, University of Alberta, Edmonton AB, T6G 2E9, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton AB, T6G 2M8, Canada
| | - Samantha A. W. Tan
- Department of Biological Sciences, University of Alberta, Edmonton AB, T6G 2E9, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton AB, T6G 2M8, Canada
| | - Alexander H. Burton
- Departments of Chemical and Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hadeel Alyenbaawi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - Edward A. Burton
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Geriatric Research, Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA 15213, USA
| | - W. Ted Allison
- Department of Biological Sciences, University of Alberta, Edmonton AB, T6G 2E9, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton AB, T6G 2M8, Canada
- Department of Medical Genetics, University of Alberta, Edmonton AB, T6G 2H7, Canada
| |
Collapse
|
15
|
Cohen‐Adiv S, Amer‐Sarsour F, Berdichevsky Y, Boxer E, Goldstein O, Gana‐Weisz M, Tripathi U, Rike WA, Prag G, Gurevich T, Giladi N, Stern S, Orr‐Urtreger A, Friedmann‐Morvinski D, Ashkenazi A. TMEM16F regulates pathologic α-synuclein secretion and spread in cellular and mouse models of Parkinson's disease. Aging Cell 2025; 24:e14387. [PMID: 39487963 PMCID: PMC11822650 DOI: 10.1111/acel.14387] [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: 04/15/2024] [Revised: 09/14/2024] [Accepted: 10/01/2024] [Indexed: 11/04/2024] Open
Abstract
One of the main hallmarks of Parkinson's disease (PD) pathology is the spread of the aggregate-prone protein α-synuclein (α-syn), which can be detected in the plasma and cerebrospinal fluid of patients as well as in the extracellular environment of neuronal cells. The secreted α-syn can exhibit "prion-like" behavior and transmission to naïve cells can promote conformational changes and pathology. The precise role of plasma membrane proteins in the pathologic process of α-syn is yet to be fully resolved. The TMEM16 family of lipid scramblases and ion channels has been recently associated with cancer and infectious diseases but is less known for its role in aging-related diseases. To elucidate the role of TMEM16F in α-syn spread, we transduced neurons derived from TMEM16F knockout mice with a reporter system that enables the distinction between donor and recipient neurons of pathologic α-synA53T. We found that the spread of α-synA53T was reduced in neurons derived from TMEM16F-knockout mice. These findings were recapitulated in vivo in a mouse model of PD, where attenuated α-synA53T spread was observed when TMEM16F was ablated. Moreover, we identified a single nucleotide polymorphism in TMEM16F of Ashkenazi Jewish PD patients resulting in a missense Ala703Ser mutation with enhanced lipid scramblase activity. This mutation is associated with altered regulation of α-synA53T extracellular secretion in cellular models of PD. Our study highlights TMEM16F as a novel regulator of α-syn spread and as a potential therapeutic target in synucleinopathies.
Collapse
Affiliation(s)
- Stav Cohen‐Adiv
- The Department of Cell and Developmental Biology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Fatima Amer‐Sarsour
- The Department of Cell and Developmental Biology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Yevgeny Berdichevsky
- The Department of Cell and Developmental Biology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Emily Boxer
- The School of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| | - Orly Goldstein
- Laboratory of Biomarkers and Genomics of Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Mali Gana‐Weisz
- Laboratory of Biomarkers and Genomics of Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Utkarsh Tripathi
- Sagol Department of Neurobiology, Faculty of Natural SciencesUniversity of HaifaHaifaIsrael
| | - Wote Amelo Rike
- Sagol Department of Neurobiology, Faculty of Natural SciencesUniversity of HaifaHaifaIsrael
| | - Gali Prag
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- School of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
| | - Tanya Gurevich
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Movement Disorders Division, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Nir Giladi
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
- Brain DivisionTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural SciencesUniversity of HaifaHaifaIsrael
| | - Avi Orr‐Urtreger
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
- Laboratory of Biomarkers and Genomics of Neurodegeneration, Neurological InstituteTel Aviv Sourasky Medical CenterTel AvivIsrael
- Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
| | - Dinorah Friedmann‐Morvinski
- The School of Neurobiology, Biochemistry and Biophysics, the George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| | - Avraham Ashkenazi
- The Department of Cell and Developmental Biology, Faculty of Medical and Health SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
16
|
da Costa Caiado MJ, Dolga AM, den Dunnen WFA. Iron(ing) out parkinsonisms: The interplay of proteinopathy and ferroptosis in Parkinson's disease and tau-related parkinsonisms. Redox Biol 2025; 79:103478. [PMID: 39721496 PMCID: PMC11732237 DOI: 10.1016/j.redox.2024.103478] [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: 11/01/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Parkinsonian syndromes are characterised by similar motor-related symptomology resulting from dopaminergic neuron damage. While Parkinson's disease (PD) is the most prevalent parkinsonism, we also focus on two other variants, Progressive supranuclear palsy (PSP) and Corticobasal degeneration (CBD). Due to the clinical similarities of these parkinsonisms, and since definite diagnoses are only possible post-mortem, effective therapies and novel biomarkers of disease are scarce. Thus, we explore the current findings relating to the relationship of parkinsonism proteinopathy (α-synuclein in PD, and tau in PSP/CBD) paralleled to a specific form of cell death, ferroptosis. Ferroptosis is characterised by iron-induced lipid peroxidation and several markers of this pathway have been identified to control intracellular iron fluctuations. However, in parkinsonism, these mechanisms are thought to become dysfunctional. Although both proteinopathies have been linked to ferroptosis, much less is known about ferroptotic cell death and tau in the context of PSP/CBD. Interestingly, clinical trials targeting iron have recently shown conflicting results which begs to question the complexity of the ferroptotic pathway and alludes to the need for exploring other ferroptosis-related machinery as possible therapeutic targets. Overall, we address the literature gap in parkinsonism proteinopathy and ferroptosis, and its relevance to understanding disease pathophysiology and aetiology.
Collapse
Affiliation(s)
- Maria João da Costa Caiado
- Graduate School of Medical Sciences (GSMS) and Research School of Behavioural and Cognitive Neurosciences (BCN), University of Groningen, 9713 GZ, Groningen, the Netherlands; Department of Pathology and Medical Biology, University Medical Centre Groningen (UMCG), Hanzeplein 1, 9713 GZ, Groningen, the Netherlands; Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, the Netherlands.
| | - Amalia M Dolga
- Department of Pathology and Medical Biology, University Medical Centre Groningen (UMCG), Hanzeplein 1, 9713 GZ, Groningen, the Netherlands; Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV, Groningen, the Netherlands
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, University Medical Centre Groningen (UMCG), Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| |
Collapse
|
17
|
Chang X, Tse AM, Fayzullina M, Albanese A, Kim M, Wang CF, Zheng Z, Joshi RV, Williams CK, Magaki SD, Vinters HV, Jones JO, Haworth IS, Seidler PM. Monoaminergic neurotransmitters are bimodal effectors of tau aggregation. SCIENCE ADVANCES 2025; 11:eadr8055. [PMID: 39888993 PMCID: PMC11784839 DOI: 10.1126/sciadv.adr8055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/31/2024] [Indexed: 02/02/2025]
Abstract
Neurotransmitters (NTs) mediate trans-synaptic signaling, and disturbances in their levels are linked to aging and brain disorders. Here, we ascribe an additional function for NTs in mediating intracellular protein aggregation by interaction with cytosolic protein fibrils. Cell-based seeding experiments revealed monoaminergic NTs as inhibitors of tau. Seeding is a disease-relevant mechanism involving catalysis by fibrils, leading to the aggregation of proteins in Alzheimer's disease and other neurodegenerative diseases. Chemotyping small molecules with varied backbone structures revealed determinants of aggregation inhibitors and catalysts. Among those identified were monoaminergic NTs. Dose titrations revealed bimodal effects indicative of fibril disaggregation, with aggregation catalysis occurring at low ratios of NTs and inhibited seeding ensuing at higher concentrations. Bimodal effects by NTs extend from in vitro systems to dopaminergic neurons, suggesting that pharmacotherapies that modify intracellular NT levels could shape the neuronal protein aggregation environment.
Collapse
Affiliation(s)
- Xinmin Chang
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Amanda M. Tse
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Marina Fayzullina
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Angela Albanese
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Medical Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Minchan Kim
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Conner F. Wang
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Zipeng Zheng
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Ruchira V. Joshi
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Christopher K. Williams
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shino D. Magaki
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Harry V. Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Brain Research Institute, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Jeremy O. Jones
- Simulations Plus Inc., 42505 10th Street West, Lancaster, CA 93534-7059, USA
| | - Ian S. Haworth
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Paul M. Seidler
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
18
|
Hyde VR, Zhou C, Fernandez JR, Chatterjee K, Ramakrishna P, Lin A, Fisher GW, Çeliker OT, Caldwell J, Bender O, Sauer PJ, Lugo-Martinez J, Bar DZ, D'Aiuto L, Shemesh OA. Anti-herpetic tau preserves neurons via the cGAS-STING-TBK1 pathway in Alzheimer's disease. Cell Rep 2025; 44:115109. [PMID: 39753133 DOI: 10.1016/j.celrep.2024.115109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease (AD) diagnosis relies on the presence of extracellular β-amyloid (Aβ) and intracellular hyperphosphorylated tau (p-tau). Emerging evidence suggests a potential link between AD pathologies and infectious agents, with herpes simplex virus 1 (HSV-1) being a leading candidate. Our investigation, using metagenomics, mass spectrometry, western blotting, and decrowding expansion pathology, detects HSV-1-associated proteins in human brain samples. Expression of the herpesvirus protein ICP27 increases with AD severity and strongly colocalizes with p-tau but not with Aβ. Modeling in human brain organoids shows that HSV-1 infection elevates tau phosphorylation. Notably, p-tau reduces ICP27 expression and markedly decreases post-infection neuronal death from 64% to 7%. This modeling prompts investigation into the cGAS-STING-TBK1 pathway products, nuclear factor (NF)-κB and IRF-3, which colocalizes with ICP27 and p-tau in AD. Furthermore, experimental activation of STING enhances tau phosphorylation, while TBK1 inhibition prevents it. Together, these findings suggest that tau phosphorylation acts as an innate immune response in AD, driven by cGAS-STING.
Collapse
Affiliation(s)
- Vanesa R Hyde
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaoming Zhou
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Juan R Fernandez
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Krishnashis Chatterjee
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Pururav Ramakrishna
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Amanda Lin
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Gregory W Fisher
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Orhan Tunç Çeliker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jill Caldwell
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Omer Bender
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Peter Joseph Sauer
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose Lugo-Martinez
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel Z Bar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Leonardo D'Aiuto
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Or A Shemesh
- School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
19
|
Vaquer-Alicea J, Manon VA, Bommareddy V, Kunach P, Gupta A, Monistrol J, Perez VA, Tran HT, Saez-Calveras N, Du S, Batra S, Stoddard D, White CL, Joachimiak LA, Shahmoradian SH, Diamond MI. Functional classification of tauopathy strains reveals the role of protofilament core residues. SCIENCE ADVANCES 2025; 11:eadp5978. [PMID: 39841851 PMCID: PMC11753436 DOI: 10.1126/sciadv.adp5978] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
Distinct tau amyloid assemblies underlie diverse tauopathies but defy rapid classification. Cell and animal experiments indicate tau functions as a prion, as different strains propagated in cells cause unique, transmissible neuropathology after inoculation. Strain amplification requires compatibility of the monomer and amyloid template. We used cryo-electron microscopy to study one cell-based yellow fluorescent protein (YFP)-tagged strain, resolving its amyloid nature. We then used sequential alanine (Ala) substitution (scan) within tau repeat domain (RD) to measure incorporation to preexisting tau RD-YFP aggregates. This robustly discriminated strains, defining sequences critical for monomer incorporation. We then created 3R/4R or 4R wild-type RD (amino acids 246 to 408) biosensors. Ala scan of recombinant tau seeds with the Alzheimer's disease (AD) fold matched that of AD homogenate. We scanned 22 brain lysates comprising four tauopathies. This clustered cases by neuropathological syndrome, revealed the role of amino acids in protofilament folds, and allowed strain discrimination based on amino acid requirements for prion replication.
Collapse
Affiliation(s)
- Jaime Vaquer-Alicea
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Victor A. Manon
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vaibhav Bommareddy
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Peter Kunach
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ankit Gupta
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jim Monistrol
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Valerie A. Perez
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hung Tri Tran
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nil Saez-Calveras
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Siling Du
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sushobhna Batra
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Daniel Stoddard
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Charles L. White
- Department of Pathology, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lukasz A. Joachimiak
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sarah H. Shahmoradian
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marc I. Diamond
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
20
|
Lo Cascio F, Park S, Sengupta U, Puangmalai N, Bhatt N, Shchankin N, Jerez C, Moreno N, Bittar A, Xavier R, Zhao Y, Wang C, Fu H, Ma Q, Montalbano M, Kayed R. Brain-derived tau oligomer polymorphs: distinct aggregations, stability profiles, and biological activities. Commun Biol 2025; 8:53. [PMID: 39809992 PMCID: PMC11733013 DOI: 10.1038/s42003-025-07499-w] [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: 09/26/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Aggregation of microtubule-associated tau protein is a distinct hallmark of several neurodegenerative disorders such as Alzheimer's disease (AD), dementia with Lewy bodies (DLB), and progressive supranuclear palsy (PSP). Tau oligomers are suggested to be the primary neurotoxic species that initiate aggregation and propagate prion-like structures. Furthermore, different diseases are shown to have distinct structural characteristics of aggregated tau, denoted as polymorphs. Here, we investigate the structural and functional differences of amplified brain-derived tau oligomers (aBDTOs) from AD, DLB, and PSP. Our results indicate that the aBDTOs possess different structural and morphological features that impact neuronal function, gene regulation, and ultimately disease progression. The distinct tau oligomeric polymorphs may thus contribute to the development of clinical phenotypes and shape the progression of diseases. Our results can provide insight into developing personalized therapy to target a specific neurotoxic tau polymorph.
Collapse
Affiliation(s)
- Filippa Lo Cascio
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Suhyeorn Park
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nikita Shchankin
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Naomi Moreno
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Alice Bittar
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rhea Xavier
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yingxin Zhao
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Cankun Wang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Hongjun Fu
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Mauro Montalbano
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA.
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA.
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
21
|
Oldani EG, Stillman NH, Dohoney RA, Baysah CZ, Kumar S. Inhibition of Phosphorylated Alpha-Synuclein Aggregation by Synthetic Protein Mimetics and Foldamers. ACS Chem Neurosci 2025; 16:152-160. [PMID: 39719105 DOI: 10.1021/acschemneuro.4c00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024] Open
Abstract
The formation of Lewy bodies (LB) is a pathological hallmark for synucleinopathies, which is an umbrella term for many diseases, including Parkinson's disease, Lewy body dementia, and multiple system atrophy. One of the main components of LB is the aggregates of phosphorylated modification of α-Synuclein at residue 129 (αS-129), a neuronal protein expressed in the dopaminergic neurons in the brain. There are equivocal results about the role of αS-129, suggesting its involvement in both potentiating pathology and a functional role to rescue pathology. Regardless, a potential therapeutic strategy for LB-based pathologies could be the identification of inhibitors of both αS and αS-129 aggregation. However, to the best of our knowledge, there are no reports of ligands that can potently inhibit the aggregation of αS-129. Our group has recently identified potent antagonists of αS aggregation based on the oligopyridylamide (synthetic protein mimetics) and oligoquinoline (foldamers) scaffolds. Both ligands were potent antagonists of αS aggregation-mediated disease phenotypes in various PD models. Here, we tested both ligands against αS-129 aggregation and the coaggregation of αS and αS-129 (αS/αS-129). Both ligands were potent antagonists of αS-129 aggregation and coaggregation of αS/αS-129 in biophysical and cellular models of PD. Both ligands rescued cell toxicity mediated by the coaggregation of αS/αS-129. To the best of our knowledge, these are the first ligands that potently inhibit the major component of LB. This finding will aid in the development of therapeutic insights into aggregation-related synucleinopathies.
Collapse
Affiliation(s)
- Emily G Oldani
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
- Molecular and Cellular Biophysics Program, University of Denver, Denver, Colorado 80210, United States
| | - Nicholas H Stillman
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Ryan A Dohoney
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Charles Zuwu Baysah
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
| | - Sunil Kumar
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- The Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado 80210, United States
- Molecular and Cellular Biophysics Program, University of Denver, Denver, Colorado 80210, United States
| |
Collapse
|
22
|
Batra S, Vaquer-Alicea J, Valdez C, Taylor SP, Manon VA, Vega AR, Kashmer OM, Kolay S, Lemoff A, Cairns NJ, White CL, Diamond MI. VCP regulates early tau seed amplification via specific cofactors. Mol Neurodegener 2025; 20:2. [PMID: 39773263 PMCID: PMC11707990 DOI: 10.1186/s13024-024-00783-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Neurodegenerative tauopathies may progress based on seeding by pathological tau assemblies, whereby an aggregate is released from one cell, gains entry to an adjacent or connected cell, and serves as a specific template for its own replication in the cytoplasm. Seeding into the complex cytoplasmic milieu happens within hours, implying the existence of unknown factors that regulate this process. METHODS We used proximity labeling to identify proteins that control seed amplification within 5 h of seed exposure. We fused split-APEX2 to the C-terminus of tau repeat domain (RD) to reconstitute peroxidase activity 5 h after seeded intracellular tau aggregation. Valosin containing protein (VCP/p97) was the top hit. VCP harbors dominant mutations that underlie two neurodegenerative diseases, multisystem proteinopathy and vacuolar tauopathy, but its mechanistic role is unclear. We used immortalized cells and human neurons to study the effects of VCP on tau seeding. We exposed cells to fibrils or brain homogenates in cell culture media and measured effects on uptake and induction of intracellular tau aggregation following various genetic and pharmacological manipulations of VCP. RESULTS VCP knockdown reduced tau seeding. Chemical inhibitors had opposing effects on seeding in HEK293T tau biosensor cells and human neurons: ML-240 increased seeding efficiency, whereas NMS-873 decreased it. The inhibitors only functioned when administered within 8 h of seed exposure, indicating a role for VCP early in seed processing. We screened 30 VCP co-factors in HEK293T biosensor cells by genetic knockout or knockdown. Reduction of ATXN3, NSFL1C, UBE4B, NGLY1, and OTUB1 decreased tau seeding, as did NPLOC4, which also uniquely increased soluble tau levels. By contrast, reduction of FAF2 increased tau seeding. CONCLUSIONS Divergent effects on tau seeding of chemical inhibitors and cofactor reduction indicate that VCP regulates this process. This is consistent with a cytoplasmic processing complex centered on VCP that directs seeds acutely towards degradation vs. amplification.
Collapse
Affiliation(s)
- Sushobhna Batra
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Jaime Vaquer-Alicea
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Clarissa Valdez
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Skyler P Taylor
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Victor A Manon
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Anthony R Vega
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Omar M Kashmer
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Sourav Kolay
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Nigel J Cairns
- Department of Clinical and Biological Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Charles L White
- Department of Pathology, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 6124 Harry Hines Blvd, Dallas, TX, NS8.334, United States.
- Department of Neurology, Dallas, United States.
| |
Collapse
|
23
|
Zeng Z, Tsay K, Vijayan V, Frost MP, Prakash S, Quddus A, Albert A, Vigers M, Srivastava M, Woerman AL, Han S. Passaging Human Tauopathy Patient Samples in Cells Generates Heterogeneous Fibrils with a Subpopulation Adopting Disease Folds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.07.19.549721. [PMID: 37502998 PMCID: PMC10370138 DOI: 10.1101/2023.07.19.549721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The discovery by cryo-electron microscopy (cryo-EM) that the neu-ropathological hallmarks of different tauopathies, including Alzheimer's disease, corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP), are caused by unique misfolded conformations of the protein tau is among the most profound developments in neurodegenerative disease research. To capitalize on these discoveries for therapeutic development, one must achieve in vitro replication of tau fibrils that adopt the representative tauopathy disease folds, which represents a grand challenge for the field. A widely used approach has been seeded propagation using pathological tau fibrils derived from post-mortem patient samples in biosensor cells that expresses a fragment of the tau protein known as K18, or Tau4RD, containing the microtubule-binding repeat domain of tau as the substrate. The new insights from cryo-EM raised the question of whether the Tau4RD fragment is capable of adopting characteristic tau folds found in CBD and PSP patient fibrils, and whether cell-passaged and amplified tau fibrils can be used as seeds to achieve cell-free assembly of recombinant 4R tau into fibrils without the addition of cofactors. Using Double Electron Electron Resonance (DEER) spectroscopy, we discovered that cell-passaged pathological seeds generate heterogeneous fibrils that are, however, distinct between the CBD and PSP lysate-seeded fibrils, and vastly different from heparin-induced tau fibril structures. Moreover, the lysate-seeded fibrils contain a characteristic sub-population that resembles the disease fold corresponding to the respective starting patient sample. These findings indicate that templated propagation using CBD and PSP patient-derived fibrils is possible with a tau fragment that does not contain the entire pathological fibril core, but also that additional mechanisms must be tuned to converge on a homogeneous fibril population.
Collapse
|
24
|
Shyong J, Wang J, Huynh QDT, Fayzullina M, Yuan B, Lee CK, Minehan T, Seidler PM, Wang CCC. Discovery of penicillic acid as a chemical probe against tau aggregation in Alzheimer's disease. Chem Sci 2024; 15:20467-20477. [PMID: 39583559 PMCID: PMC11583217 DOI: 10.1039/d4sc05469e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder proven to be caused by the aggregation of protein tau into fibrils, resulting in neuronal death. The irreparable neuronal damage leads to irreversible symptoms with no cure; therefore, disaggregation of these tau fibrils could be targeted as a therapeutic approach to AD. Here we have developed a fungal natural product library to screen for secondary metabolites that have bioactive potential towards AD tau. Our initial screenings indicate that penicillic acid demonstrates anti-aggregation activity towards tau, while further in vitro experiments reveal that penicillic acid directly inhibits tau by disaggregating fibrils. Although penicillic acid possesses blood-brain barrier penetrability properties that are computationally predicted to be favorable, it is presumed to contain some mutagenic effects as well. To address this, we used the backbone of penicillic acid as a chemical probe to discover similar compounds that can inhibit AD tau aggregation with limited mutagenicity. This work suggests the potential of discovering chemical probes through natural product screening for small-molecule drug discovery of tauopathies.
Collapse
Affiliation(s)
- Jennifer Shyong
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California Los Angeles California 90089 USA
| | - Jinliang Wang
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California Los Angeles California 90089 USA
| | - Quoc-Dung Tran Huynh
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University Taipei 11031 Taiwan
| | - Marina Fayzullina
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California Los Angeles California 90089 USA
| | - Bo Yuan
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California Los Angeles California 90089 USA
| | - Ching-Kuo Lee
- School of Pharmacy, College of Pharmacy, Taipei Medical University Taipei 11031 Taiwan
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University Taipei 11031 Taiwan
| | - Thomas Minehan
- Department of Chemistry and Biochemistry, California State University, Northridge Northridge California 91330 USA
| | - Paul M Seidler
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California Los Angeles California 90089 USA
| | - Clay C C Wang
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California Los Angeles California 90089 USA
- Department of Chemistry, University of Southern California, Dornsife College of Letters, Arts, and Sciences Los Angeles California 90089 USA
| |
Collapse
|
25
|
Wang J, Williams CK, DeTure MA, Magaki SD, Dickson DW, Vinters HV, Seidler PM. Tau seeds catalyze fibril-type structures from GFP tau biosensor cells. Structure 2024; 32:2251-2258.e3. [PMID: 39488204 DOI: 10.1016/j.str.2024.10.013] [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: 04/13/2024] [Revised: 08/06/2024] [Accepted: 10/08/2024] [Indexed: 11/04/2024]
Abstract
Fibril-type aggregates of tau occur in Alzheimer's disease (AD) and dozens of tauopathies. Fibrils catalyze aggregation by prion-like seeding, which in part underlies disease progression. Seeding by recombinant and brain-derived tau fibrils is measured using biosensor cells that express aggregation-prone tau mutants fused with fluorescent reporter proteins. Seeding results in a punctated phenotype that is well established, but evidence that fluorescent tau fusion proteins from biosensor cells assemble into fibril-type structures is lacking. We investigated the effects of seeding on fibril formation by biosensor cells. Fluorescent punctated cell phenotypes that were catalyzed persisted with varying stabilities. Seeded cells bearing punctated phenotypes yielded sarkosyl-insoluble fibrils, although non-seeded cells did not. ImmunoEM of cell-purified fibrils shows that GFP localizes to the proteolytically sensitive fuzzy coat of tau fibrils. The presented data offer compelling evidence that fluorescent puncta are fibril-type aggregates of tau that result from prion-like seeding.
Collapse
Affiliation(s)
- Jinliang Wang
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California Mann School of Pharmacy and Pharmaceutical Sciences, 1985 Zonal Avenue, Los Angeles, CA 90089-9121, USA
| | - Christopher K Williams
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Michael A DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Shino D Magaki
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Paul M Seidler
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California Mann School of Pharmacy and Pharmaceutical Sciences, 1985 Zonal Avenue, Los Angeles, CA 90089-9121, USA.
| |
Collapse
|
26
|
Eid S, Lee S, Verkuyl CE, Almanza D, Hanna J, Shenouda S, Belotserkovsky A, Zhao W, Watts JC. The importance of prion research. Biochem Cell Biol 2024; 102:448-471. [PMID: 38996387 DOI: 10.1139/bcb-2024-0018] [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] [Indexed: 07/14/2024] Open
Abstract
Over the past four decades, prion diseases have received considerable research attention owing to their potential to be transmitted within and across species as well as their consequences for human and animal health. The unprecedented nature of prions has led to the discovery of a paradigm of templated protein misfolding that underlies a diverse range of both disease-related and normal biological processes. Indeed, the "prion-like" misfolding and propagation of protein aggregates is now recognized as a common underlying disease mechanism in human neurodegenerative disorders such as Alzheimer's and Parkinson's disease, and the prion principle has led to the development of novel diagnostic and therapeutic strategies for these illnesses. Despite these advances, research into the fundamental biology of prion diseases has declined, likely due to their rarity and the absence of an acute human health crisis. Given the past translational influence, continued research on the etiology, pathogenesis, and transmission of prion disease should remain a priority. In this review, we highlight several important "unsolved mysteries" in the prion disease research field and how solving them may be crucial for the development of effective therapeutics, preventing future outbreaks of prion disease, and understanding the pathobiology of more common human neurodegenerative disorders.
Collapse
Affiliation(s)
- Shehab Eid
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Seojin Lee
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Claire E Verkuyl
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Dustin Almanza
- Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Joseph Hanna
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Sandra Shenouda
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Ari Belotserkovsky
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Wenda Zhao
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Powell W, Nahum M, Pankratz K, Herlory M, Greenwood J, Poliyenko D, Holland P, Jing R, Biggerstaff L, Stowell MHB, Walczak MA. Post-Translational Modifications Control Phase Transitions of Tau. ACS CENTRAL SCIENCE 2024; 10:2145-2161. [PMID: 39634209 PMCID: PMC11613296 DOI: 10.1021/acscentsci.4c01319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 12/07/2024]
Abstract
The self-assembly of Tau into filaments, which mirror the structures observed in Alzheimer's disease (AD) brains, raises questions about the role of AD-specific post-translational modifications (PTMs) in the formation of paired helical filaments (PHFs). To investigate this, we developed a synthetic approach to produce Tau(291-391) featuring N-acetyllysine, phosphoserine, phosphotyrosine, and N-glycosylation at positions commonly modified in post-mortem AD brains. Using various electron and optical microscopy techniques, we discovered that these modifications generally hinder the in vitro assembly of Tau into PHFs. Interestingly, while acetylation's effect on Tau assembly displayed variability, either promoting or inhibiting phase transitions in cofactor-free aggregation, heparin-induced aggregation, and RNA-mediated liquid-liquid phase separation (LLPS), phosphorylation uniformly mitigated these processes. Our observations suggest that PTMs, particularly those situated outside the rigid core, are pivotal in the nucleation of PHFs. Moreover, with heparin-induced aggregation leading to the formation of heterogeneous aggregates, most AD-specific PTMs appeared to decelerate aggregation. The impact of acetylation on RNA-induced LLPS was notably site-dependent, whereas phosphorylation consistently reduced LLPS across all proteoforms examined. These insights underscore the complex interplay between site-specific PTMs and environmental factors in modulating Tau aggregation kinetics, highlighting the role of PTMs located outside the ordered filament core in driving the self-assembly.
Collapse
Affiliation(s)
- Wyatt
C. Powell
- Department
of Chemistry, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - McKinley Nahum
- Department
of Chemistry, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Karl Pankratz
- Department
of Chemistry, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Morgane Herlory
- Department
of Chemistry, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - James Greenwood
- Department
of Chemistry, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Darya Poliyenko
- Department
of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Patrick Holland
- Department
of Chemistry, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Ruiheng Jing
- Department
of Chemistry, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Luke Biggerstaff
- Department
of Chemistry, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Michael H. B. Stowell
- Department
of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| | - Maciej A. Walczak
- Department
of Chemistry, University of Colorado, Boulder, Boulder, Colorado 80309, United States
| |
Collapse
|
28
|
Andrade GCD, Mota MF, Moreira-Ferreira DN, Silva JL, de Oliveira GAP, Marques MA. Protein aggregation in health and disease: A looking glass of two faces. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 145:145-217. [PMID: 40324846 DOI: 10.1016/bs.apcsb.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Protein molecules organize into an intricate alphabet of twenty amino acids and five architecture levels. The jargon "one structure, one functionality" has been challenged, considering the amount of intrinsically disordered proteins in the human genome and the requirements of hierarchical hetero- and homo-protein complexes in cell signaling. The assembly of large protein structures in health and disease is now viewed through the lens of phase separation and transition phenomena. What drives protein misfolding and aggregation? Or, more fundamentally, what hinders proteins from maintaining their native conformations, pushing them toward aggregation? Here, we explore the principles of protein folding, phase separation, and aggregation, which hinge on crucial events such as the reorganization of solvents, the chemical properties of amino acids, and their interactions with the environment. We focus on the dynamic shifts between functional and dysfunctional states of proteins and the conditions that promote protein misfolding, often leading to disease. By exploring these processes, we highlight potential therapeutic avenues to manage protein aggregation and reduce its harmful impacts on health.
Collapse
Affiliation(s)
- Guilherme C de Andrade
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology, Federal University of Rio de Janeiro, Rio De Janeiro, RJ, Brazil
| | - Michelle F Mota
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology, Federal University of Rio de Janeiro, Rio De Janeiro, RJ, Brazil
| | - Dinarte N Moreira-Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology, Federal University of Rio de Janeiro, Rio De Janeiro, RJ, Brazil
| | - Jerson L Silva
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology, Federal University of Rio de Janeiro, Rio De Janeiro, RJ, Brazil
| | - Guilherme A P de Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology, Federal University of Rio de Janeiro, Rio De Janeiro, RJ, Brazil.
| | - Mayra A Marques
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology, Federal University of Rio de Janeiro, Rio De Janeiro, RJ, Brazil.
| |
Collapse
|
29
|
Parra Bravo C, Naguib SA, Gan L. Cellular and pathological functions of tau. Nat Rev Mol Cell Biol 2024; 25:845-864. [PMID: 39014245 DOI: 10.1038/s41580-024-00753-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/18/2024]
Abstract
Tau protein is involved in various cellular processes, including having a canonical role in binding and stabilization of microtubules in neurons. Tauopathies are neurodegenerative diseases marked by the abnormal accumulation of tau protein aggregates in neurons, as seen, for example, in conditions such as frontotemporal dementia and Alzheimer disease. Mutations in tau coding regions or that disrupt tau mRNA splicing, tau post-translational modifications and cellular stress factors (such as oxidative stress and inflammation) increase the tendency of tau to aggregate and interfere with its clearance. Pathological tau is strongly implicated in the progression of neurodegenerative diseases, and the propagation of tau aggregates is associated with disease severity. Recent technological advancements, including cryo-electron microscopy and disease models derived from human induced pluripotent stem cells, have increased our understanding of tau-related pathology in neurodegenerative conditions. Substantial progress has been made in deciphering tau aggregate structures and the molecular mechanisms that underlie protein aggregation and toxicity. In this Review, we discuss recent insights into the diverse cellular functions of tau and the pathology of tau inclusions and explore the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Celeste Parra Bravo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sarah A Naguib
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
30
|
Taniguchi D, Shimonaka S, Imtiaz A, Elahi M, Hatano T, Imai Y, Hattori N. Legumain/asparaginyl endopeptidase-resistant tau fibril fold produces corticobasal degeneration-specific C-terminal tau fragment. Neurobiol Dis 2024; 201:106686. [PMID: 39353514 DOI: 10.1016/j.nbd.2024.106686] [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: 06/06/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024] Open
Abstract
Corticobasal degeneration (CBD) is a major four-repeat tauopathy along with progressive supranuclear palsy (PSP). Although detergent-insoluble 37-40-kDa carboxyl-terminal tau fragments (CTFs) are hallmarks of CBD pathology, the process of their formation is unknown. This study monitored the formation of CBD-type fibrils that exhibit astrocytic plaques, a characteristic CBD pathology, using its biochemical properties different from those of Alzheimer's disease/PSP-type fibrils. Tau fibrils from patients with CBD were amplified in non-astrocytic cultured cells, which maintained CBD-specific biochemical properties. We found that the lysosomal protease Legumain (LGMN) was involved in the generation of CBD-specific 37-40-kDa CTFs. While LGMN cleaved tau fibrils at Asn167 and Asn368 in the brain tissues of patients with Alzheimer's disease and PSP, tau fibrils from patients with CBD were predominantly resistant to cleavage at Asn368 by LGMN, resulting in the generation of CBD-specific CTFs. LGMN preference in tau fibrils was lost upon unraveling the tau fibril fold, suggesting that the CBD-specific tau fibril fold contributes to CBD-specific CTF production. From these findings, we found a way to differentiate astrocytic plaque from tufted astrocyte using the anti-Asn368 LGMN cleavage site-specific antibody. Inoculation of tau fibrils amplified in non-astrocytic cells into the mouse brain reproduced LGMN-resistant tau fibrils and recapitulated anti-Asn368-negative astrocytic plaques, which are characteristic of CBD pathology. This study supports the existence of disease-specific tau fibrils and contribute to further understanding of the tauopathy diagnosis. Our tau propagation mouse model using cellular tau seeds may contribute to uncovering disease mechanisms and screening for potential therapeutic compounds.
Collapse
Affiliation(s)
- Daisuke Taniguchi
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Shotaro Shimonaka
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Ahmed Imtiaz
- Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Montasir Elahi
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Taku Hatano
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yuzuru Imai
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan.
| |
Collapse
|
31
|
Quinodoz SA, Jiang L, Abu-Alfa AA, Comi TJ, Zhao H, Yu Q, Wiesner LW, Botello JF, Donlic A, Soehalim E, Zorbas C, Wacheul L, Košmrlj A, Lafontaine D, Klinge S, Brangwynne CP. Mapping and engineering RNA-controlled architecture of the multiphase nucleolus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.28.615444. [PMID: 39386460 PMCID: PMC11463421 DOI: 10.1101/2024.09.28.615444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Biomolecular condensates are key features of intracellular compartmentalization. As the most prominent nuclear condensate in eukaryotes, the nucleolus is a layered multiphase liquid-like structure and the site of ribosome biogenesis. In the nucleolus, ribosomal RNAs (rRNAs) are transcribed and processed, undergoing multiple maturation steps that ultimately result in formation of the ribosomal small subunit (SSU) and large subunit (LSU). However, how rRNA processing is coupled to the layered nucleolar organization is poorly understood due to a lack of tools to precisely monitor and perturb nucleolar rRNA processing dynamics. Here, we developed two complementary approaches to spatiotemporally map rRNA processing and engineer de novo nucleoli. Using sequencing in parallel with imaging, we found that rRNA processing steps are spatially segregated, with sequential maturation of rRNA required for its outward movement through nucleolar phases. Furthermore, by generating synthetic de novo nucleoli through an engineered rDNA plasmid system in cells, we show that defects in SSU processing can alter the ordering of nucleolar phases, resulting in inside-out nucleoli and preventing rRNA outflux, while LSU precursors are necessary to build the outermost layer of the nucleolus. These findings demonstrate how rRNA is both a scaffold and substrate for the nucleolus, with rRNA acting as a programmable blueprint for the multiphase architecture that facilitates assembly of an essential molecular machine.
Collapse
Affiliation(s)
- S A Quinodoz
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 21044, USA
| | - L Jiang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - A A Abu-Alfa
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - T J Comi
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton NJ, 08544, USA
| | - H Zhao
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton NJ, 08544, USA
| | - Q Yu
- Lewis-Sigler Institute for Integrative Genomics, Princeton NJ, 08544, USA
| | - L W Wiesner
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - J F Botello
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - A Donlic
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - E Soehalim
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton NJ, 08544, USA
| | - C Zorbas
- RNA Molecular Biology, Fonds de la Recherche Scientifique (F.R.S./FNRS), Université libre de Bruxelles (ULB), Biopark campus, B-6041 Gosselies, Belgium
| | - L Wacheul
- RNA Molecular Biology, Fonds de la Recherche Scientifique (F.R.S./FNRS), Université libre de Bruxelles (ULB), Biopark campus, B-6041 Gosselies, Belgium
| | - A Košmrlj
- Department of Mechanical and Aerospace Engineering, Princeton NJ, 08544, USA
- Princeton Materials Institute, Princeton NJ, 08544, USA
| | - Dlj Lafontaine
- RNA Molecular Biology, Fonds de la Recherche Scientifique (F.R.S./FNRS), Université libre de Bruxelles (ULB), Biopark campus, B-6041 Gosselies, Belgium
| | - S Klinge
- Laboratory of Protein and Nucleic Acid Chemistry, The Rockefeller University, New York, NY 10065, USA
| | - C P Brangwynne
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton NJ, 08544, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton NJ, 08544, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 21044, USA
| |
Collapse
|
32
|
Poulidou V, Liampas I, Arnaoutoglou M, Dardiotis E, Siokas V. The Imbalance of Homocysteine, Vitamin B12 and Folic Acid in Parkinson Plus Syndromes: A Review beyond Parkinson Disease. Biomolecules 2024; 14:1213. [PMID: 39456145 PMCID: PMC11506381 DOI: 10.3390/biom14101213] [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: 06/28/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
While there is a link between homocysteine (Hcy), B12 and folic acid and neurodegeneration, especially in disorders like Parkinson's and Alzheimer's diseases, its role in Parkinson plus syndromes (PPS) has only been partially investigated. It appears that elevated Hcy, along with an imbalance of its essential vitamin cofactors, are both implicated in the development and progression of parkinsonian syndromes, which represent different disease pathologies, namely alpha-synucleinopathies and tauopathies. Attributing a potential pathogenetic role in hyperhomocysteinemia would be crucial in terms of improving the diagnostic and prognostic accuracy of these syndromes and also for providing a new target for possible therapeutic intervention. The scope of this review is to focus on vitamin imbalance in PPS, with a special emphasis on the role of Hcy, B12 and folic acid in the neurodegenerative process and their implication in the therapeutic approach of these disorders.
Collapse
Affiliation(s)
- Vasiliki Poulidou
- First Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kyriakidi 1, 54636 Thessaloniki, Greece;
| | - Ioannis Liampas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (I.L.); (E.D.)
| | - Marianthi Arnaoutoglou
- Department of Clinical Neurophysiology, School of Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kyriakidi 1, 54636 Thessaloniki, Greece;
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (I.L.); (E.D.)
| | - Vasileios Siokas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, 41100 Larissa, Greece; (I.L.); (E.D.)
| |
Collapse
|
33
|
Sala-Jarque J, Gil V, Andrés-Benito P, Martínez-Soria I, Picón-Pagès P, Hernández F, Ávila J, Lanciego JL, Nuvolone M, Aguzzi A, Gavín R, Ferrer I, Del Río JA. The cellular prion protein does not affect tau seeding and spreading of sarkosyl-insoluble fractions from Alzheimer's disease. Sci Rep 2024; 14:21622. [PMID: 39284839 PMCID: PMC11405773 DOI: 10.1038/s41598-024-72232-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 09/04/2024] [Indexed: 09/20/2024] Open
Abstract
The cellular prion protein (PrPC) plays many roles in the developing and adult brain. In addition, PrPC binds to several amyloids in oligomeric and prefibrillar forms and may act as a putative receptor of abnormal misfolded protein species. The role of PrPC in tau seeding and spreading is not known. In the present study, we have inoculated well-characterized sarkosyl-insoluble fractions of sporadic Alzheimer's disease (sAD) into the brain of adult wild-type mice (Prnp+/+), Prnp0/0 (ZH3 strain) mice, and mice over-expressing the secreted form of PrPC lacking their GPI anchor (Tg44 strain). Phospho-tau (ptau) seeding and spreading involving neurons and oligodendrocytes were observed three and six months after inoculation. 3Rtau and 4Rtau deposits from the host tau, as revealed by inoculating Mapt0/0 mice and by using specific anti-mouse and anti-human tau antibodies suggest modulation of exon 10 splicing of the host mouse Mapt gene elicited by exogenous sAD-tau. However, no tau seeding and spreading differences were observed among Prnp genotypes. Our results show that PrPC does not affect tau seeding and spreading in vivo.
Collapse
Affiliation(s)
- Julia Sala-Jarque
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri and Reixac 15-21, 08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
- Neuroscience Research Institute and Molecular, Cell and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri and Reixac 15-21, 08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Pol Andrés-Benito
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Bellvitge University Hospital-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Inés Martínez-Soria
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri and Reixac 15-21, 08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Pol Picón-Pagès
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri and Reixac 15-21, 08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Félix Hernández
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain
- Centro de Biología Molecular 'Severo Ochoa' (CBMSO) CSIC/UAM, Madrid, Spain
| | - Jesús Ávila
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain
- Centro de Biología Molecular 'Severo Ochoa' (CBMSO) CSIC/UAM, Madrid, Spain
| | - José Luis Lanciego
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain
- Department of Neurosciences, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Mario Nuvolone
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Amyloidosis Research and Treatment Center, Foundation Scientific Institute Policlinico San Matteo, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Rosalina Gavín
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri and Reixac 15-21, 08028, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Isidro Ferrer
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Bellvitge University Hospital-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Baldiri and Reixac 15-21, 08028, Barcelona, Spain.
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain.
- Ciberned (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Madrid, Spain.
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
34
|
Ansari S, Lagasca D, Dumarieh R, Xiao Y, Krishna S, Li Y, Frederick KK. In cell NMR reveals cells selectively amplify and structurally remodel amyloid fibrils. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612142. [PMID: 39314304 PMCID: PMC11419106 DOI: 10.1101/2024.09.09.612142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Amyloid forms of α-synuclein adopt different conformations depending on environmental conditions. Advances in structural biology have accelerated fibril characterization. However, it remains unclear which conformations predominate in biological settings because current methods typically not only require isolating fibrils from their native environments, but they also do not provide insight about flexible regions. To address this, we characterized α-syn amyloid seeds and used sensitivity enhanced nuclear magnetic resonance to investigate the amyloid fibrils resulting from seeded amyloid propagation in different settings. We found that the amyloid fold and conformational preferences of flexible regions are faithfully propagated in vitro and in cellular lysates. However, seeded propagation of amyloids inside cells led to the minority conformation in the seeding population becoming predominant and more ordered, and altered the conformational preferences of flexible regions. The examination of the entire ensemble of protein conformations in biological settings that is made possible with this approach may advance our understanding of protein misfolding disorders and facilitate structure-based drug design efforts.
Collapse
Affiliation(s)
- Shoyab Ansari
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390-8816
| | - Dominique Lagasca
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390-8816
| | - Rania Dumarieh
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390-8816
| | - Yiling Xiao
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390-8816
| | - Sakshi Krishna
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390-8816
| | - Yang Li
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390-8816
| | - Kendra K. Frederick
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390-8816
- Center for Alzheimer’s and Neurodegenerative Disease, UT Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
35
|
Mate de Gerando A, Khasnavis A, Welikovitch LA, Bhavsar H, Glynn C, Quittot N, Perbet R, Hyman BT. Aqueous extractable nonfibrillar and sarkosyl extractable fibrillar Alzheimer's disease tau seeds have distinct properties. Acta Neuropathol Commun 2024; 12:145. [PMID: 39252090 PMCID: PMC11382398 DOI: 10.1186/s40478-024-01849-1] [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: 07/03/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
Pathological tau fibrils in progressive supranuclear palsy, frontotemporal dementia, chronic traumatic encephalopathy, and Alzheimer's disease each have unique conformations, and post-translational modifications that correlate with unique disease characteristics. However, within Alzheimer's disease (AD), both fibrillar (sarkosyl insoluble (AD SARK tau)), and nonfibrillar (aqueous extractable high molecular weight (AD HMW tau)) preparations have been suggested to be seed-competent. We now explore if these preparations are similar or distinct in their in vivo seeding characteristics. Using an in vivo amplification and time-course paradigm we demonstrate that, for AD HMW and AD SARK tau species, the amplified material is biochemically similar to the original sample. The HMW and SARK materials also show different clearance, propagation kinetics, and propagation patterns. These data indicate the surprising co-occurrence of multiple distinct tau species within the same AD brain, supporting the idea that multiple tau conformers - both fibrillar and nonfibrillar- can impact phenotype in AD.
Collapse
Affiliation(s)
- Anastasie Mate de Gerando
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Anita Khasnavis
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Harshil Bhavsar
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Calina Glynn
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Noe Quittot
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Romain Perbet
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Cambridge, MA, USA.
| |
Collapse
|
36
|
Ayers J, Lopez TP, Steele IT, Oehler A, Roman-Albarran R, Cleveland E, Chong A, Carlson GA, Condello C, Prusiner SB. Severe neurodegeneration in brains of transgenic rats producing human tau prions. Acta Neuropathol 2024; 148:25. [PMID: 39160375 PMCID: PMC11333523 DOI: 10.1007/s00401-024-02771-5] [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: 04/09/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024]
Abstract
Both wild-type and mutant tau proteins can misfold into prions and self-propagate in the central nervous system of animals and people. To extend the work of others, we investigated the molecular basis of tau prion-mediated neurodegeneration in transgenic (Tg) rats expressing mutant human tau (P301S); this line of Tg rats is denoted Tg12099. We used the rat Prnp promoter to drive the overexpression of mutant tau (P301S) in the human 0N4R isoform. In Tg12099(+/+) rats homozygous for the transgene, ubiquitous expression of mutant human tau resulted in the progressive accumulation of phosphorylated tau inclusions, including silver-positive tangles in the frontal cortices and limbic system. Signs of central nervous system dysfunction were found in terminal Tg12099(+/+) rats exhibiting severe neurodegeneration and profound atrophy of the amygdala and piriform cortex. The greatest increases in tau prion activity were found in the corticolimbic structures. In contrast to the homozygous Tg12099(+/+) rats, we found lower levels of mutant tau in the hemizygous rats, resulting in few neuropathologic changes up to 2 years of age. Notably, these hemizygous rats could be infected by intracerebral inoculation with recombinant tau fibrils or precipitated tau prions from the brain homogenates of sick, aged homozygous Tg12099(+/+) rats. Our studies argue that the regional propagation of tau prions and neurodegeneration in the Tg12099 rats resembles that found in human primary tauopathies. These findings seem likely to advance our understanding of human tauopathies and may lead to effective therapeutics for Alzheimer's disease and other tau prion disorders.
Collapse
Affiliation(s)
- Jacob Ayers
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - T Peter Lopez
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Ian T Steele
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Abby Oehler
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Rigo Roman-Albarran
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Elisa Cleveland
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Alex Chong
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - George A Carlson
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA
| | - Carlo Condello
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, 94158, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, 94158, USA.
| |
Collapse
|
37
|
Zhang X, Wang J, Zhang Z, Ye K. Tau in neurodegenerative diseases: molecular mechanisms, biomarkers, and therapeutic strategies. Transl Neurodegener 2024; 13:40. [PMID: 39107835 PMCID: PMC11302116 DOI: 10.1186/s40035-024-00429-6] [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: 11/22/2023] [Accepted: 07/05/2024] [Indexed: 09/14/2024] Open
Abstract
The deposition of abnormal tau protein is characteristic of Alzheimer's disease (AD) and a class of neurodegenerative diseases called tauopathies. Physiologically, tau maintains an intrinsically disordered structure and plays diverse roles in neurons. Pathologically, tau undergoes abnormal post-translational modifications and forms oligomers or fibrous aggregates in tauopathies. In this review, we briefly introduce several tauopathies and discuss the mechanisms mediating tau aggregation and propagation. We also describe the toxicity of tau pathology. Finally, we explore the early diagnostic biomarkers and treatments targeting tau. Although some encouraging results have been achieved in animal experiments and preclinical studies, there is still no cure for tauopathies. More in-depth basic and clinical research on the pathogenesis of tauopathies is necessary.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiangyu Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430000, China.
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
38
|
Zampar S, Di Gregorio SE, Grimmer G, Watts JC, Ingelsson M. "Prion-like" seeding and propagation of oligomeric protein assemblies in neurodegenerative disorders. Front Neurosci 2024; 18:1436262. [PMID: 39161653 PMCID: PMC11330897 DOI: 10.3389/fnins.2024.1436262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
Intra- or extracellular aggregates of proteins are central pathogenic features in most neurodegenerative disorders. The accumulation of such proteins in diseased brains is believed to be the end-stage of a stepwise aggregation of misfolded monomers to insoluble cross-β fibrils via a series of differently sized soluble oligomers/protofibrils. Several studies have shown how α-synuclein, amyloid-β, tau and other amyloidogenic proteins can act as nucleating particles and thereby share properties with misfolded forms, or strains, of the prion protein. Although the roles of different protein assemblies in the respective aggregation cascades remain unclear, oligomers/protofibrils are considered key pathogenic species. Numerous observations have demonstrated their neurotoxic effects and a growing number of studies have indicated that they also possess seeding properties, enabling their propagation within cellular networks in the nervous system. The seeding behavior of oligomers differs between the proteins and is also affected by various factors, such as size, shape and epitope presentation. Here, we are providing an overview of the current state of knowledge with respect to the "prion-like" behavior of soluble oligomers for several of the amyloidogenic proteins involved in neurodegenerative diseases. In addition to providing new insight into pathogenic mechanisms, research in this field is leading to novel diagnostic and therapeutic opportunities for neurodegenerative diseases.
Collapse
Affiliation(s)
- Silvia Zampar
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Sonja E. Di Gregorio
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Gustavo Grimmer
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Joel C. Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Martin Ingelsson
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Public Health/Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
39
|
Lee E, Park H, Kim S. Transcellular transmission and molecular heterogeneity of aggregation-prone proteins in neurodegenerative diseases. Mol Cells 2024; 47:100089. [PMID: 38971320 PMCID: PMC11286998 DOI: 10.1016/j.mocell.2024.100089] [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: 05/14/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024] Open
Abstract
The accumulation of aggregation-prone proteins in a specific neuronal population is a common feature of neurodegenerative diseases, which is correlated with the development of pathological lesions in diseased brains. The formation and progression of pathological protein aggregates in susceptible neurons induce cellular dysfunction, resulting in progressive degeneration. Moreover, recent evidence supports the notion that the cell-to-cell transmission of pathological protein aggregates may be involved in the onset and progression of many neurodegenerative diseases. Indeed, several studies have identified different pathological aggregate strains. Although how these different aggregate strains form remains unclear, a variety of biomolecular compositions or cross-seeding events promoted by the presence of other protein aggregates in the cellular environment may affect the formation of different strains of pathological aggregates, which in turn can influence complex pathologies in diseased brains. In this review, we summarize the recent results regarding cell-to-cell transmission and the molecular heterogeneity of pathological aggregate strains, raising key questions for future research directions.
Collapse
Affiliation(s)
- Eunmin Lee
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk 28644, Korea
| | - Hyeonwoo Park
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk 28644, Korea
| | - Sangjune Kim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk 28644, Korea.
| |
Collapse
|
40
|
Lester E, Parker R. Tau, RNA, and RNA-Binding Proteins: Complex Interactions in Health and Neurodegenerative Diseases. Neuroscientist 2024; 30:458-472. [PMID: 36892034 DOI: 10.1177/10738584231154551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
The tau protein is a key contributor to multiple neurodegenerative diseases. The pathology of tau is thought to be related to tau's propensity to form self-templating fibrillar structures that allow tau fibers to propagate in the brain by prion-like mechanisms. Unresolved issues with respect to tau pathology are how the normal function of tau and its misregulation contribute to disease, how cofactors and cellular organelles influence the initiation and propagation of tau fibers, and determining the mechanism of tau toxicity. Herein, we review the connection between tau and degenerative diseases, the basis for tau fibrilization, and how that process interacts with cellular molecules and organelles. One emerging theme is that tau interacts with RNA and RNA-binding proteins, normally and in pathologic aggregates, which may provide insight into alterations in RNA regulation observed in disease.
Collapse
Affiliation(s)
- Evan Lester
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Roy Parker
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
- Howard Hughes Medical Institute, University of Colorado, Boulder, CO, USA
| |
Collapse
|
41
|
Martinez P, Jury-Garfe N, Patel H, You Y, Perkins A, You Y, Lee-Gosselin A, Vidal R, Lasagna-Reeves CA. Phosphorylation at serine 214 correlates with tau seeding activity in an age-dependent manner in two mouse models for tauopathies and is required for tau transsynaptic propagation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604618. [PMID: 39211286 PMCID: PMC11361173 DOI: 10.1101/2024.07.22.604618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pathological aggregation and propagation of hyperphosphorylated and aberrant forms of tau are critical features of the clinical progression of Alzheimer's disease and other tauopathies. To better understand the correlation between these pathological tau species and disease progression, we profiled the temporal progression of tau seeding activity and the levels of various phospho- and conformational tau species in the brains of two mouse models of human tauopathies. Our findings indicate that tau seeding is an early event that occurs well before the appearance of AT8-positive NFT. Specifically, we observed that tau phosphorylation in serine 214 (pTau-Ser214) positively correlates to tau seeding activity during disease progression in both mouse models. Furthermore, we found that the histopathology of pTau-Ser214 appears much earlier and has a distinct pattern and compartmentalization compared to the pathology of AT8, demonstrating the diversity of tau species within the same region of the brain. Importantly, we also observed that preventing the phosphorylation of tau at Ser214 significantly decreases tau propagation in mouse primary neurons, and seeding activity in a Drosophila model of tauopathy, suggesting a role for this tau phosphorylation in spreading pathological forms of tau. Together, these results suggest that the diverse spectrum of soluble pathological tau species could be responsible for the distinct pathological properties of tau and that it is critical to dissect the nature of the tau seed in the context of disease progression.
Collapse
|
42
|
Liu C, Meng L, Gao Y, Chen J, Zhu M, Xiong M, Xiao T, Gu X, Liu C, Li T, Zhang Z. PM2.5 triggers tau aggregation in a mouse model of tauopathy. JCI Insight 2024; 9:e176703. [PMID: 39133647 PMCID: PMC11383351 DOI: 10.1172/jci.insight.176703] [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: 10/17/2023] [Accepted: 05/31/2024] [Indexed: 09/11/2024] Open
Abstract
The aggregation and prion-like propagation of tau are the hallmarks of Alzheimer's disease (AD) and other tauopathies. However, the molecular mechanisms underlying the assembly and spread of tau pathology remain elusive. Epidemiological data show that exposure to fine particulate matter (PM2.5) is associated with an increased risk of AD. However, the molecular mechanisms remain unknown. Here, we showed that PM2.5 triggered the aggregation of tau and promoted the formation of tau fibrils. Injection of PM2.5-induced tau preformed fibrils (PFFs) into the hippocampus of tau P301S transgenic mice promoted the aggregation of tau and induced cognitive deficits and synaptic dysfunction. Furthermore, intranasal administration of PM2.5 exacerbated tau pathology and induced cognitive impairment in tau P301S mice. In conclusion, our results indicated that PM2.5 exposure promoted tau pathology and induced cognitive impairments. These results provide mechanistic insight into how PM2.5 increases the risk of AD.
Collapse
Affiliation(s)
- Congcong Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Gao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiehui Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Min Zhu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Min Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tingting Xiao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoling Gu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chaoyang Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Research Center for Environment and Health, Zhongnan University of Economics and Law, Wuhan, China
| | - Tao Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, China
| |
Collapse
|
43
|
Gaikwad S, Puangmalai N, Sonawane M, Montalbano M, Price R, Iyer MS, Ray A, Moreno S, Kayed R. Nasal tau immunotherapy clears intracellular tau pathology and improves cognitive functions in aged tauopathy mice. Sci Transl Med 2024; 16:eadj5958. [PMID: 38959324 DOI: 10.1126/scitranslmed.adj5958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/11/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
Pathological tau aggregates cause cognitive decline in neurodegenerative tauopathies, including Alzheimer's disease (AD). These aggregates are prevalent within intracellular compartments. Current tau immunotherapies have shown limited efficacy in clearing intracellular tau aggregates and improving cognition in clinical trials. In this study, we developed toxic tau conformation-specific monoclonal antibody-2 (TTCM2), which selectively recognized pathological tau aggregates in brain tissues from patients with AD, dementia with Lewy bodies (DLB), and progressive supranuclear palsy (PSP). TTCM2 potently inhibited tau-seeding activity, an essential mechanism underlying tauopathy progression. To effectively target intracellular tau aggregates and ensure rapid delivery to the brain, TTCM2 was loaded in micelles (TTCM2-ms) and administered through the intranasal route. We found that intranasally administered TTCM2-ms efficiently entered the brain in hTau-tauopathy mice, targeting pathological tau in intracellular compartments. Moreover, a single intranasal dose of TTCM2-ms effectively cleared pathological tau, elevated synaptic proteins, and improved cognitive functions in aged tauopathy mice. Mechanistic studies revealed that TTCM2-ms cleared intracellular, synaptic, and seed-competent tau aggregates through tripartite motif-containing 21 (TRIM21), an intracellular antibody receptor and E3 ubiquitin ligase known to facilitate proteasomal degradation of cytosolic antibody-bound proteins. TRIM21 was found to be essential for TTCM2-ms-mediated clearance of tau pathology. Our study collectively provides evidence of the effectiveness of nasal tau immunotherapy in targeting and clearing intracellular tau pathology through TRIM21 and enhancing cognition in aged tauopathy mice. This study could be valuable in designing effective tau immunotherapies for AD and other tauopathies.
Collapse
Affiliation(s)
- Sagar Gaikwad
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Minal Sonawane
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mauro Montalbano
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Rachel Price
- Department of Science, University "Roma Tre," Viale G. Marconi 446 00146 Rome, Italy
| | | | | | - Sandra Moreno
- Department of Science, University "Roma Tre," Viale G. Marconi 446 00146 Rome, Italy
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
44
|
Woerman AL, Bartz JC. Effect of host and strain factors on α-synuclein prion pathogenesis. Trends Neurosci 2024; 47:538-550. [PMID: 38806297 PMCID: PMC11236502 DOI: 10.1016/j.tins.2024.05.004] [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: 02/27/2024] [Revised: 04/19/2024] [Accepted: 05/04/2024] [Indexed: 05/30/2024]
Abstract
Prion diseases are a group of neurodegenerative disorders caused by misfolding of proteins into pathogenic conformations that self-template to spread disease. Although this mechanism is largely associated with the prion protein (PrP) in classical prion diseases, a growing literature indicates that other proteins, including α-synuclein, rely on a similar disease mechanism. Notably, α-synuclein misfolds into distinct conformations, or strains, that cause discrete clinical disorders including multiple system atrophy (MSA) and Parkinson's disease (PD). Because the recognized similarities between PrP and α-synuclein are increasing, this review article draws from research on PrP to identify the host and strain factors that impact disease pathogenesis, predominantly in rodent models, and focuses on key considerations for future research on α-synuclein prions.
Collapse
Affiliation(s)
- Amanda L Woerman
- Department of Microbiology, Immunology, and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, USA.
| | - Jason C Bartz
- Department of Microbiology, Immunology, and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, USA; Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE, USA.
| |
Collapse
|
45
|
Yang J, Shen N, Shen J, Yang Y, Li HL. Complicated Role of Post-translational Modification and Protease-Cleaved Fragments of Tau in Alzheimer's Disease and Other Tauopathies. Mol Neurobiol 2024; 61:4712-4731. [PMID: 38114762 PMCID: PMC11236937 DOI: 10.1007/s12035-023-03867-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023]
Abstract
Tau, a microtubule-associated protein predominantly localized in neuronal axons, plays a crucial role in promoting microtubule assembly, stabilizing their structure, and participating in axonal transport. Perturbations in tau's structure and function are implicated in the pathogenesis of neurodegenerative diseases collectively known as tauopathies, the most common disorder of which is Alzheimer's disease (AD). In tauopathies, it has been found that tau has a variety of post-translational modification (PTM) abnormalities and/or tau is cleaved into a variety of fragments by some specific proteolytic enzymes; however, the precise contributions of these abnormal modifications and fragments to disease onset and progression remain incompletely understood. Herein, we provide an overview about the involvement of distinctive abnormal tau PTMs and different tau fragments in the pathogenesis of AD and other tauopathies and discuss the involvement of proteolytic enzymes such as caspases, calpains, and asparagine endopeptidase in mediating tau cleavage while also addressing the intercellular transmission role played by tau. We anticipate that further exploration into PTMs and fragmented forms of tau will yield valuable insights for diagnostic approaches and therapeutic interventions targeting AD and other related disorders.
Collapse
Affiliation(s)
- Jie Yang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Naiting Shen
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jianying Shen
- Department of Histology and Embryology, School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong-Lian Li
- Department of Histology and Embryology, School of Basic Medicine, Key Laboratory of Education Ministry, Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
46
|
Christensen CS, Wang S, Li W, Yu D, Li HJ. Structural Variations of Prions and Prion-like Proteins Associated with Neurodegeneration. Curr Issues Mol Biol 2024; 46:6423-6439. [PMID: 39057026 PMCID: PMC11275340 DOI: 10.3390/cimb46070384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Neurodegeneration is becoming one of the leading causes of death worldwide as the population expands and grows older. There is a growing desire to understand the mechanisms behind prion proteins as well as the prion-like proteins that make up neurodegenerative diseases (NDs), including Alzheimer's disease (AD) and Parkinson's disease (PD). Both amyloid-β (Aβ) and hyperphosphorylated tau (p-tau) proteins behave in ways similar to those of the infectious form of the prion protein, PrPSc, such as aggregating, seeding, and replicating under not yet fully understood mechanisms, thus the designation of prion-like. This review aims to highlight the shared mechanisms between prion-like proteins and prion proteins in the structural variations associated with aggregation and disease development. These mechanisms largely focus on the dysregulation of protein homeostasis, self-replication, and protein aggregation, and this knowledge could contribute to diagnoses and treatments for the given NDs.
Collapse
Affiliation(s)
| | | | | | | | - Henry James Li
- School of Arts and Sciences, New York University Shanghai, 567 West Yang Si Road, Shanghai 200122, China; (C.S.C.); (S.W.); (W.L.); (D.Y.)
| |
Collapse
|
47
|
Ryder BD, Ustyantseva E, Boyer DR, Mendoza-Oliva A, Kuska MI, Wydorski PM, Macierzyńska P, Morgan N, Sawaya MR, Diamond MI, Kampinga HH, Joachimiak LA. DNAJB8 oligomerization is mediated by an aromatic-rich motif that is dispensable for substrate activity. Structure 2024; 32:662-678.e8. [PMID: 38508190 PMCID: PMC11162344 DOI: 10.1016/j.str.2024.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 01/17/2024] [Accepted: 02/22/2024] [Indexed: 03/22/2024]
Abstract
J-domain protein (JDP) molecular chaperones have emerged as central players that maintain a healthy proteome. The diverse members of the JDP family function as monomers/dimers and a small subset assemble into micron-sized oligomers. The oligomeric JDP members have eluded structural characterization due to their low-complexity, intrinsically disordered middle domains. This in turn, obscures the biological significance of these larger oligomers in protein folding processes. Here, we identified a short, aromatic motif within DNAJB8 that drives self-assembly through π-π stacking and determined its X-ray structure. We show that mutations in the motif disrupt DNAJB8 oligomerization in vitro and in cells. DNAJB8 variants that are unable to assemble bind to misfolded tau seeds more specifically and retain capacity to reduce protein aggregation in vitro and in cells. We propose a new model for DNAJB8 function in which the sequences in the low-complexity domains play distinct roles in assembly and substrate activity.
Collapse
Affiliation(s)
- Bryan D Ryder
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elizaveta Ustyantseva
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen 9713 AV, The Netherlands
| | - David R Boyer
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ayde Mendoza-Oliva
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mikołaj I Kuska
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Paweł M Wydorski
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Paulina Macierzyńska
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nabil Morgan
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Michael R Sawaya
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Harm H Kampinga
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen 9713 AV, The Netherlands
| | - Lukasz A Joachimiak
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
48
|
Buccellato FR, D'Anca M, Tartaglia GM, Del Fabbro M, Galimberti D. Frontotemporal dementia: from genetics to therapeutic approaches. Expert Opin Investig Drugs 2024; 33:561-573. [PMID: 38687620 DOI: 10.1080/13543784.2024.2349286] [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: 12/30/2023] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Frontotemporal dementia (FTD) includes a group of neurodegenerative diseases characterized clinically by behavioral disturbances and by neurodegeneration of brain anterior temporal and frontal lobes, leading to atrophy. Apart from symptomatic treatments, there is, at present, no disease-modifying cure for FTD. AREAS COVERED Three main mutations are known as causes of familial FTD, and large consortia have studied carriers of mutations, also in preclinical Phases. As genetic cases are the only ones in which the pathology can be predicted in life, compounds developed so far are directed toward specific proteins or mutations. Herein, recently approved clinical trials will be summarized, including molecules, mechanisms of action and pharmacological testing. EXPERT OPINION These studies are paving the way for the future. They will clarify whether single mutations should be addressed rather than common proteins depositing in the brain to move from genetic to sporadic FTD.
Collapse
Affiliation(s)
- Francesca R Buccellato
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Marianna D'Anca
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Gianluca Martino Tartaglia
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Del Fabbro
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
49
|
Batra S, Vaquer-Alicea JI, Valdez C, Taylor SP, Manon VA, Vega AR, Kashmer OM, Kolay S, Lemoff A, Cairns NJ, White CL, Diamond MI. VCP regulates early tau seed amplification via specific cofactors. RESEARCH SQUARE 2024:rs.3.rs-4307848. [PMID: 38826306 PMCID: PMC11142303 DOI: 10.21203/rs.3.rs-4307848/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background Neurodegenerative tauopathies may progress based on seeding by pathological tau assemblies, whereby an aggregate is released from one cell, gains entry to an adjacent or connected cell, and serves as a specific template for its own replication in the cytoplasm. In vitro seeding reactions typically take days, yet seeding into the complex cytoplasmic milieu happens within hours, implicating a machinery with unknown players that controls this process in the acute phase. Methods We used proximity labeling to identify factors that control seed amplification within 5h of seed exposure. We fused split-APEX2 to the C-terminus of tau repeat domain (RD) to reconstitute peroxidase activity 5h after seeded intracellular tau aggregation. Valosin containing protein (VCP/p97) was the top hit. VCP harbors dominant mutations that underlie two neurodegenerative diseases, multisystem proteinopathy and vacuolar tauopathy, but its mechanistic role is unclear. We used immortalized cells and human neurons to study the effects of VCP on tau seeding. We exposed cells to fibrils or brain homogenates in cell culture media and measured effects on uptake and induction of intracellular tau aggregation following various genetic and chemical manipulations of VCP. Results VCP knockdown reduced tau seeding. Chemical inhibitors had opposing effects on aggregation in HEK293T tau biosensor cells and human neurons alike: ML-240 increased seeding efficiency, whereas NMS-873 decreased it. The inhibitors were effective only when administered within 8h of seed exposure, indicating a role for VCP early in seed processing. We screened 30 VCP co-factors in HEK293T biosensor cells by genetic knockout or knockdown. Reduction of ATXN3, NSFL1C, UBE4B, NGLY1, and OTUB1 decreased tau seeding, as did NPLOC4, which also uniquely increased soluble tau levels. By contrast, reduction of FAF2 increased tau seeding. Conclusions Divergent effects on tau seeding of chemical inhibitors and cofactor reduction indicate that VCP regulates this process. This is consistent with a dedicated cytoplasmic processing complex based on VCP that directs seeds acutely towards degradation vs. amplification.
Collapse
Affiliation(s)
- Sushobhna Batra
- UT Southwestern: The University of Texas Southwestern Medical Center
| | | | - Clarissa Valdez
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Skyler P Taylor
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Victor A Manon
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Anthony R Vega
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Omar M Kashmer
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Sourav Kolay
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Andrew Lemoff
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Nigel J Cairns
- University of Exeter Faculty of Health and Life Sciences
| | - Charles L White
- UT Southwestern: The University of Texas Southwestern Medical Center
| | - Marc I Diamond
- UT Southwestern: The University of Texas Southwestern Medical Center
| |
Collapse
|
50
|
de Fátima Dos Santos Sampaio M, de Paiva YB, Sampaio TB, Pereira MG, Coimbra NC. Therapeutic applicability of cannabidiol and other phytocannabinoids in epilepsy, multiple sclerosis and Parkinson's disease and in comorbidity with psychiatric disorders. Basic Clin Pharmacol Toxicol 2024; 134:574-601. [PMID: 38477419 DOI: 10.1111/bcpt.13997] [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: 10/14/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024]
Abstract
Studies have demonstrated the neuroprotective effect of cannabidiol (CBD) and other Cannabis sativa L. derivatives on diseases of the central nervous system caused by their direct or indirect interaction with endocannabinoid system-related receptors and other molecular targets, such as the 5-HT1A receptor, which is a potential pharmacological target of CBD. Interestingly, CBD binding with the 5-HT1A receptor may be suitable for the treatment of epilepsies, parkinsonian syndromes and amyotrophic lateral sclerosis, in which the 5-HT1A serotonergic receptor plays a key role. The aim of this review was to provide an overview of cannabinoid effects on neurological disorders, such as epilepsy, multiple sclerosis and Parkinson's diseases, and discuss their possible mechanism of action, highlighting interactions with molecular targets and the potential neuroprotective effects of phytocannabinoids. CBD has been shown to have significant therapeutic effects on epilepsy and Parkinson's disease, while nabiximols contribute to a reduction in spasticity and are a frequent option for the treatment of multiple sclerosis. Although there are multiple theories on the therapeutic potential of cannabinoids for neurological disorders, substantially greater progress in the search for strong scientific evidence of their pharmacological effectiveness is needed.
Collapse
Affiliation(s)
- Maria de Fátima Dos Santos Sampaio
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), São Paulo, Brazil
- Center for Agropastoralism Sciences and Technology (CCTA), North Fluminense State University (UENF), Rio de Janeiro, Brazil
- Psychobiology Division, Behavioural Neurosciences Institute (INeC), Ribeirão Preto, São Paulo, Brazil
| | - Yara Bezerra de Paiva
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), São Paulo, Brazil
- Psychobiology Division, Behavioural Neurosciences Institute (INeC), Ribeirão Preto, São Paulo, Brazil
- NAP-USP-Neurobiology of Emotions Research Center (NuPNE), Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Tuane Bazanella Sampaio
- Pharmacology Post-Graduation Program, Health Sciences Centre, Santa Maria Federal University, Santa Maria, Brazil
| | - Messias Gonzaga Pereira
- Center for Agropastoralism Sciences and Technology (CCTA), North Fluminense State University (UENF), Rio de Janeiro, Brazil
| | - Norberto Cysne Coimbra
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), São Paulo, Brazil
- Psychobiology Division, Behavioural Neurosciences Institute (INeC), Ribeirão Preto, São Paulo, Brazil
- NAP-USP-Neurobiology of Emotions Research Center (NuPNE), Ribeirão Preto School of Medicine of the University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|