1
|
Santos RX, Lee SH, Lofthouse R, Melis V, Robinson L, Leith M, Dreesen E, Armstrong P, Vorley T, Goatman EA, Hull C, Riedel G, Wischik CM, Harrington CR. Hydromethylthionine sustains truncated tau-dependent inflammation-lowering effects in mouse brain. FEBS J 2025; 292:2602-2623. [PMID: 39960781 PMCID: PMC12103070 DOI: 10.1111/febs.70021] [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: 10/23/2024] [Revised: 12/04/2024] [Accepted: 01/31/2025] [Indexed: 05/25/2025]
Abstract
Tauopathies are a heterogeneous mixture of neurodegenerative disorders, including Alzheimer's disease and frontotemporal dementia (FTD), characterised by the accumulation of tau filaments in brain tissue. Tau protein aggregation is inhibited by hydromethylthionine (HMT), an effect that appeared to be prevented in clinical trials for subjects already receiving acetylcholinesterase inhibitors or memantine. Since neuroinflammatory responses are associated with tauopathies, we investigated the effect of HMT on the brain immune response and inflammatory status in line 66 (L66) mice, an FTD-like model overexpressing human tau, in the presence of memantine. We determined whether HMT (5 and 15 mg·kg-1), either singly or combined with memantine (20 mg·kg-1), would have a sustained impact on neuroinflammation following the cessation of drug administration. The levels of core tau fragments in L66+/- mice (P301S/G335D-hTau) were decreased in a dose-dependent manner 12 weeks after the last administration of HMT, an effect that was not affected by memantine. HMT lowered the levels of tumour necrosis factor alpha (TNF-α), thus favouring an environment conducive to neuronal protection and repair. HMT sustained increased microglial reactivity after its discontinuation, which may assist in the removal of tau aggregates, but co-administration with memantine prevented the HMT-sustained activation of microglia. These findings indicate that HMT has a beneficial effect in reducing neuroinflammation that accompanies a decrease in the accumulation of truncated tau species and that these benefits are not susceptible to interference by memantine. In turn, the nature of drug interference between HMT and memantine seems to be independent of tau and related to microglia reactivity.
Collapse
Affiliation(s)
- Renato X. Santos
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
| | - Sophie H. Lee
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
| | - Richard Lofthouse
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
- Scottish Biologics FacilityUniversity of AberdeenUK
| | - Valeria Melis
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
| | - Lianne Robinson
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
| | - Michael Leith
- Department of Chemistry, School of Natural and Computing SciencesUniversity of AberdeenUK
| | - Eline Dreesen
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
| | - Paul Armstrong
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
| | - Thomas Vorley
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
| | - Elizabeth A. Goatman
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
| | | | - Gernot Riedel
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
| | - Claude M. Wischik
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
- TauRx Therapeutics Ltd.AberdeenUK
| | - Charles R. Harrington
- Institute of Medical Sciences, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenUK
- TauRx Therapeutics Ltd.AberdeenUK
| |
Collapse
|
2
|
Scott O, Saran E, Freeman SA. The spectrum of lysosomal stress and damage responses: from mechanosensing to inflammation. EMBO Rep 2025; 26:1425-1439. [PMID: 40016424 PMCID: PMC11933331 DOI: 10.1038/s44319-025-00405-9] [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: 10/07/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 03/01/2025] Open
Abstract
Cells and tissues turn over their aged and damaged components in order to adapt to a changing environment and maintain homeostasis. These functions rely on lysosomes, dynamic and heterogeneous organelles that play essential roles in nutrient redistribution, metabolism, signaling, gene regulation, plasma membrane repair, and immunity. Because of metabolic fluctuations and pathogenic threats, lysosomes must adapt in the short and long term to maintain functionality. In response to such challenges, lysosomes deploy a variety of mechanisms that prevent the breaching of their membrane and escape of their contents, including pathogen-associated molecules and hydrolases. While transient permeabilization of the lysosomal membrane can have acute beneficial effects, supporting inflammation and antigen cross-presentation, sustained or repeated lysosomal perforations have adverse metabolic and transcriptional consequences and can lead to cell death. This review outlines factors contributing to lysosomal stress and damage perception, as well as remedial processes aimed at addressing lysosomal disruptions. We conclude that lysosomal stress plays widespread roles in human physiology and pathology, the understanding and manipulation of which can open the door to novel therapeutic strategies.
Collapse
Affiliation(s)
- Ori Scott
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada
- Division of Clinical Immunology and Allergy, Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Ekambir Saran
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada
| | - Spencer A Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Gao A, Lv J, Su Y. The Inflammatory Mechanism of Parkinson's Disease: Gut Microbiota Metabolites Affect the Development of the Disease Through the Gut-Brain Axis. Brain Sci 2025; 15:159. [PMID: 40002492 PMCID: PMC11853208 DOI: 10.3390/brainsci15020159] [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: 01/07/2025] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Parkinson's disease is recognized as the second most prevalent neurodegenerative disorder globally, with its incidence rate projected to increase alongside ongoing population growth. However, the precise etiology of Parkinson's disease remains elusive. This article explores the inflammatory mechanisms linking gut microbiota to Parkinson's disease, emphasizing alterations in gut microbiota and their metabolites that influence the disease's progression through the bidirectional transmission of inflammatory signals along the gut-brain axis. Building on this mechanistic framework, this article further discusses research methodologies and treatment strategies focused on gut microbiota metabolites, including metabolomics detection techniques, animal model investigations, and therapeutic approaches such as dietary interventions, probiotic treatments, and fecal transplantation. Ultimately, this article aims to elucidate the relationship between gut microbiota metabolites and the inflammatory mechanisms underlying Parkinson's disease, thereby paving the way for novel avenues in the research and treatment of this condition.
Collapse
Affiliation(s)
| | | | - Yanwei Su
- Department of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (A.G.); (J.L.)
| |
Collapse
|
4
|
Shahidehpour RK, Nelson PT, Katsumata Y, Bachstetter AD. Exploring the link between dystrophic microglia and the spread of Alzheimer's neuropathology. Brain 2025; 148:89-101. [PMID: 39101580 PMCID: PMC11706277 DOI: 10.1093/brain/awae258] [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: 05/17/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 08/06/2024] Open
Abstract
Genetics and other data modalities indicate that microglia play a critical role in Alzheimer's disease progression, but details of the disease-driving influence of microglia are poorly understood. Microglial cells can be parsed into subtypes based on their histological appearance. One subtype of microglia, termed dystrophic microglia, is characterized structurally by fragmented processes and cytoplasmic decay, and their presence has been associated with ageing and neurodegeneration. Recent studies suggest that the interaction between tau proteins and amyloid-β might induce dystrophic changes in microglia, potentially linking amyloid-β and tau pathologies to their effects on these microglia. We developed a study of human brains to test the hypothesis that dystrophic microglia are involved in Alzheimer's disease progression. We speculated that if their presence is unique to Alzheimer's disease neuropathological change, they would be substantially more common in Alzheimer's disease neuropathological change than in neurodegenerative diseases characterized by other proteinopathies, e.g. α-synuclein or transactive response (TAR) DNA-binding protein 43 kDa (TDP-43) pathology. Our analyses used histologically stained sections from five human brain regions of 64 individuals across six disease states, from healthy controls to advanced Alzheimer's disease stages, including comparative conditions such as Lewy body disease and limbic-predominant age-related TDP-43 encephalopathy neuropathological change. Using stereological sampling and digital pathology, we assessed populations of ramified, hypertrophic and dystrophic microglia. We found a significant increase in dystrophic microglia in areas affected early by Alzheimer's disease neuropathological change, suggesting a disease-specific role in neuropathology. Mediation analysis and structural equation modelling suggest that dystrophic microglia might impact the regional spread of Alzheimer's disease neuropathological change. In the mediation model, tau was found to be the initiating factor leading to the development of dystrophic microglia, which was then associated with the spread of amyloid-β and tau. These results suggest that a loss of the protective role of microglia could contribute to the spread of Alzheimer's disease neuropathological change and indicate that further research into preserving microglial function might be warranted.
Collapse
Affiliation(s)
- Ryan K Shahidehpour
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Department of Pathology and Laboratory Medicine, Division of Neuropathology, University of Kentucky, Lexington, KY 40536, USA
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Biostatistics, University of Kentucky, Lexington, KY 40536, USA
| | - Adam D Bachstetter
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
5
|
Chunowski P, Otto-Ślusarczyk D, Duszyńska-Wąs K, Drzewińska A, Załęski A, Madetko-Alster N, Wiercińska-Drapało A, Struga M, Alster P. Possible Impact of Peripheral Inflammatory Factors and Interleukin-1β (IL-1β) on Cognitive Functioning in Progressive Supranuclear Palsy-Richardson Syndrome (PSP-RS) and Progressive Supranuclear Palsy-Predominant Parkinsonism (PSP-P). Int J Mol Sci 2024; 25:13211. [PMID: 39684921 DOI: 10.3390/ijms252313211] [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: 11/22/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Progressive supranuclear palsy (PSP) is a tauopathic atypical parkinsonian syndrome. Recent studies suggest that inflammation may play a role in PSP pathogenesis, highlighting markers like the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and cytokines such as IL-1β and IL-6. This study aimed to assess the relationship between peripheral inflammatory markers and psychological abnormalities in PSP-RS and PSP-P patients. The study included 24 participants: 12 with PSP-RS, 12 with PSP-P, and 12 controls. Cognitive function was assessed using the Montreal Cognitive Assessment (MoCA); however, the executive functions were evaluated using the Frontal Assessment Battery (FAB), while inflammatory markers such as IL-1β, IL6, NLR, and PLR were measured. The parameter correlation was executed using Spearman's correlation (rs). The analysis revealed significant negative correlations between NLR and MoCA (rs = -0.48), as well as between PLR and MoCA (rs = -0.60). The negative correlation between IL-1β and MoCA was statistically significant but relatively weak. This study highlights the relevance of inflammatory markers such as NLR and PLR in reflecting cognitive decline in PSP patients, with IL-1β potentially playing a protective role in cognitive function.
Collapse
Affiliation(s)
- Patryk Chunowski
- Department of Neurology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | | | | | | | - Andrzej Załęski
- Department of Infectious and Tropical Diseases and Hepatology, Medical University of Warsaw, Wolska 37, 01-201 Warsaw, Poland
| | | | - Alicja Wiercińska-Drapało
- Department of Infectious and Tropical Diseases and Hepatology, Medical University of Warsaw, Wolska 37, 01-201 Warsaw, Poland
| | - Marta Struga
- Department of Biochemistry, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Piotr Alster
- Department of Neurology, Medical University of Warsaw, 03-242 Warsaw, Poland
| |
Collapse
|
6
|
Basheer N, Muhammadi MK, Freites CL, Avila M, Momand MUD, Hryntsova N, Smolek T, Katina S, Zilka N. TLR4-mediated chronic neuroinflammation has no effect on tangle pathology in a tauopathy mouse model. Front Aging Neurosci 2024; 16:1468602. [PMID: 39503044 PMCID: PMC11536299 DOI: 10.3389/fnagi.2024.1468602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/26/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Alzheimer's disease (AD) is marked by the accumulation of fibrillary aggregates composed of pathological tau protein. Although neuroinflammation is frequently observed in conjunction with tau pathology, current preclinical evidence does not sufficiently establish a direct causal role in tau tangle formation. This study aimed to evaluate whether chronic Toll-like receptor 4 (TLR4) stimulation, induced by a high dose of lipopolysaccharide (LPS, 5 mg/kg), exacerbates neurofibrillary tangle (NFT) pathology in a transgenic mouse model of tauopathy that expresses human truncated 151-391/3R tau, an early feature of sporadic AD. Methods We utilized a transgenic mouse model of tauopathy subjected to chronic TLR4 stimulation via weekly intraperitoneal injections of LPS over nine consecutive weeks. Neurofibrillary tangle formation, microglial activation, and tau hyperphosphorylation in the brainstem and hippocampus were assessed through immunohistochemistry, immunofluorescence, and detailed morphometric analysis of microglia. Results Chronic LPS treatment led to a significant increase in the number of Iba-1+ microglia in the LPS-treated group compared to the sham group (p < 0.0001). Notably, there was a 1.5- to 1.7-fold increase in microglia per tangle-bearing neuron in the LPS-treated group. These microglia exhibited a reactive yet exhausted phenotype, characterized by a significant reduction in cell area (p < 0.0001) without significant changes in other morphometric parameters, such as perimeter, circumference, solidity, aspect ratio, or arborization degree. Despite extensive microglial activation, there was no observed reduction in tau hyperphosphorylation or a decrease in tangle formation in the brainstem, where pathology predominantly develops in this model. Discussion These findings suggest that chronic TLR4 stimulation in tau-transgenic mice results in significant microglial activation but does not influence tau tangle formation. This underscores the complexity of the relationship between neuroinflammation and tau pathology, indicating that additional mechanisms may be required for neuroinflammation to directly contribute to tau tangle formation.
Collapse
Affiliation(s)
- Neha Basheer
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - Carlos Leandro Freites
- Institute of Histology and Embryology of Mendoza (IHEM), National University of Cuyo, National Scientific and Technical Research Council (CONICET), Mendoza, Argentina
| | - Martin Avila
- Institute of Histology and Embryology of Mendoza (IHEM), National University of Cuyo, National Scientific and Technical Research Council (CONICET), Mendoza, Argentina
| | - Miraj Ud Din Momand
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Natalia Hryntsova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Stanislav Katina
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Mathematics and Statistics, Faculty of Science, Masaryk University, Brno, Czechia
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
7
|
Zhang S, Gao Z, Feng L, Li M. Prevention and Treatment Strategies for Alzheimer's Disease: Focusing on Microglia and Astrocytes in Neuroinflammation. J Inflamm Res 2024; 17:7235-7259. [PMID: 39421566 PMCID: PMC11484773 DOI: 10.2147/jir.s483412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease characterized by its insidious onset and progressive development, making it the most common form of dementia. Despite its prevalence, the exact causes and mechanisms responsible for AD remain unclear. Recent studies have highlighted that inflammation in the central nervous system (CNS) plays a crucial role in both the initiation and progression of AD. Neuroinflammation, an immune response within the CNS triggered by glial cells in response to various stimuli, such as nerve injury, infection, toxins, or autoimmune reactions, has emerged as a significant factor alongside amyloid deposition and neurofibrillary tangles (NFTs) commonly associated with AD. This article aims to provide an overview of the most recent research regarding the involvement of neuroinflammation in AD, with a particular focus on elucidating the specific mechanisms involving microglia and astrocytes. By exploring these intricate processes, a new theoretical framework can be established to further probe the impact of neuroinflammation on the development and progression of AD. Through a deeper understanding of these underlying mechanisms, potential targets for therapeutic interventions and novel treatment strategies can be identified in the ongoing battle against AD.
Collapse
Affiliation(s)
- Shenghao Zhang
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| | - Zhejianyi Gao
- Department of Orthopaedics, Fushun Hospital of Chinese Medicine, Fushun, Liaoning Province, 113008, People’s Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
- Shandong Key Laboratory of TCM Multi-Targets Intervention and Disease Control, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, 271000, People’s Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| |
Collapse
|
8
|
Alster P, Otto-Ślusarczyk D, Kutyłowski M, Migda B, Wiercińska-Drapało A, Jabłońska J, Struga M, Madetko-Alster N. The associations between common neuroimaging parameters of Progressive Supranuclear Palsy in magnetic resonance imaging and non-specific inflammatory factors - pilot study. Front Immunol 2024; 15:1458713. [PMID: 39176092 PMCID: PMC11338774 DOI: 10.3389/fimmu.2024.1458713] [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: 07/02/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Progressive Supranuclear Palsy is an atypical parkinsonism based on tauopathic pathology. Growing interest is associated with the pathomechanism of this disease. Among theories analyzing this issue can be mentioned the one highlighting the significance of inflammation. In this study authors examined 14 patients with PSP-Richardson syndrome (PSP-RS) and 13 healthy volunteers using laboratory testing based on the analysis of interleukins 1 and 6 (IL-1 and IL-6), tau in the cerebrospinal fluid (CSF) and non-specific parameters of peripheral inflammation in the serum (IL-1, IL-6, neutrophils, lymphocytes, monocytes, platelets and the ratios based on the factors). All of the patients underwent neuroimaging using magnetic resonance imaging using 3 Tesla. The serum levels of IL-1 were positively correlated with the area of the mesencephalon, suggesting that higher levels of IL-1 are not linked with atrophic changes in this region, whereas serum levels IL-6 was positively correlated with frontal horn width and negatively correlated with superior cerebellar area. Additionally IL-6 in the serum was found to be correlated with neutrophil-to-high density lipoprotein ratio. The observations were not confirmed in the analysis of the levels of interleukins in the CSF. To the best of our knowledge this work is one of the first analyzing this issue. The outcome of the work shows that the role of interleukins associated with microglial activation may possibly differ in the context of neurodegenerative changes, moreover the role of peripheral inflammation in PSP requires further analysis.
Collapse
Affiliation(s)
- Piotr Alster
- Department of Neurology, Medical University of Warsaw, Warsaw, Masovian, Poland
| | | | - Michał Kutyłowski
- Department of Diagnostic Imaging, Mazowiecki Hospital Brodnowski, Warsaw, Poland
| | - Bartosz Migda
- Diagnostic Ultrasound Lab, Department of Pediatric Radiology, Medical University of Warsaw, Warsaw, Masovian, Poland
| | - Alicja Wiercińska-Drapało
- Department of Infectious and Tropical Diseases and Hepatology, Medical University of Warsaw, Warsaw, Masovian, Poland
| | - Joanna Jabłońska
- Department of Infectious and Tropical Diseases and Hepatology, Medical University of Warsaw, Warsaw, Masovian, Poland
| | - Marta Struga
- Department of Biochemistry, Medical University of Warsaw, Warsaw, Masovian, Poland
| | | |
Collapse
|
9
|
Papageorgakopoulou MA, Bania A, Lagogianni IA, Birmpas K, Assimakopoulou M. The Role of Glia Telomere Dysfunction in the Pathogenesis of Central Nervous System Diseases. Mol Neurobiol 2024; 61:5868-5881. [PMID: 38240992 PMCID: PMC11249767 DOI: 10.1007/s12035-024-03947-6] [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: 10/10/2023] [Accepted: 01/09/2024] [Indexed: 07/16/2024]
Abstract
Maintaining the telomere length is decisive for the viability and homeostasis process of all the cells of an organism, including human glial cells. Telomere shortening of microglial cells has been widely associated with the onset and progression of neurodegenerative diseases such as Parkinson's and Alzheimer's disease. Additionally, traumatic brain injury appears to have a positive correlation with the telomere-shortening process of microglia, and telomere length can be used as a non-invasive biomarker for the clinical management of these patients. Moreover, telomere involvement through telomerase reactivation and homologous recombination also known as the alternative lengthening of telomeres (ALT) has been described in gliomagenesis pathways, and particular focus has been given in the translational significance of these mechanisms in gliomas diagnosis and prognostic classification. Finally, glia telomere shortening is implicated in some psychiatric diseases. Given that telomere dysfunction of glial cells is involved in the central nervous system (CNS) disease pathogenesis, it represents a promising drug target that could lead to the incorporation of new tools in the medicinal arsenal for the management of so far incurable conditions.
Collapse
Affiliation(s)
| | - Angelina Bania
- School of Medicine, University of Patras, 26504, Patras, Greece
| | | | | | - Martha Assimakopoulou
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, Preclinical Medicine Department Building, 1 Asklipiou, 26504, Patras, Greece.
| |
Collapse
|
10
|
Yan H, Wang W, Cui T, Shao Y, Li M, Fang L, Feng L. Advances in the Understanding of the Correlation Between Neuroinflammation and Microglia in Alzheimer's Disease. Immunotargets Ther 2024; 13:287-304. [PMID: 38881647 PMCID: PMC11180466 DOI: 10.2147/itt.s455881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease with a subtle and progressive onset and is the most common type of dementia. However, its etiology and pathogenesis have not yet been fully elucidated. The common pathological manifestations of AD include extraneuronal β-amyloid deposition (Aβ), intraneuronal tau protein phosphorylation leading to the formation of 'neurofibrillary tangles' (NFTs), neuroinflammation, progressive loss of brain neurons/synapses, and glucose metabolism disorders. Current treatment approaches for AD primarily focus on the 'Aβ cascade hypothesis and abnormal aggregation of hyperphosphorylation of tau proteins', but have shown limited efficacy. Therefore, there is an ongoing need to identify more effective treatment targets for AD. The central nervous system (CNS) inflammatory response plays a key role in the occurrence and development of AD. Neuroinflammation is an immune response activated by glial cells in the CNS that usually occurs in response to stimuli such as nerve injury, infection and toxins or in response to autoimmunity. Neuroinflammation ranks as the third most prominent pathological feature in AD, following Aβ and NFTs. In recent years, the focus on the role of neuroinflammation and microglia in AD has increased due to the advancements in genome-wide association studies (GWAS) and sequencing technology. Furthermore, research has validated the pivotal role of microglia-mediated neuroinflammation in the progression of AD. Therefore, this article reviews the latest research progress on the role of neuroinflammation triggered by microglia in AD in recent years, aiming to provide a new theoretical basis for further exploring the role of neuroinflammation in the process of AD occurrence and development.
Collapse
Affiliation(s)
- Huiying Yan
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Wei Wang
- Department of Intensive Care Unit, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Tingting Cui
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Yanxin Shao
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, People's Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Limei Fang
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| |
Collapse
|
11
|
Ichikawa-Escamilla E, Velasco-Martínez RA, Adalid-Peralta L. Progressive Supranuclear Palsy Syndrome: An Overview. IBRO Neurosci Rep 2024; 16:598-608. [PMID: 38800085 PMCID: PMC11126858 DOI: 10.1016/j.ibneur.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/27/2024] [Indexed: 05/29/2024] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disease, commonly observed as a movement disorder in the group of parkinsonian diseases. The term PSP usually refers to PSP-Richardson's syndrome (PSP-RS), the most typical clinical presentation. However, the broad concept of progressive supranuclear palsy syndrome (PSP-S) applies to a set of clinical entities that share a pathophysiological origin and some symptoms. According to its clinical predominance, PSP-S is divided into subtypes. PSP-S has clinical similarities with Parkinson's disease, and both pathologies are classified in the group of parkinsonisms, but they do not share pathophysiological traits. By contrast, the pathophysiology of corticobasal syndrome (CBS) depends on tau expression and shares similarities with PSP-S in both pathophysiology and clinical picture. An involvement of the immune system has been proposed as a cause of neurodegeneration. The role of neuroinflammation in PSP-S has been studied by neuroimaging, among other methods. As it is the case in other neurodegenerative pathologies, microglial cells have been attributed a major role in PSP-S. While various studies have explored the detection and use of possible inflammatory biomarkers in PSP-S, no significant advances have been made in this regard. This review is aimed at highlighting the most relevant information on neuroinflammation and peripheral inflammation in the development and progression of PSP-S, to lay the groundwork for further research on the pathophysiology, potential biomarkers, and therapeutic strategies for PSP-S.
Collapse
Affiliation(s)
- Eduardo Ichikawa-Escamilla
- Laboratorio de Reprogramación Celular del Instituto de Fisiología Celular, UNAM, en el Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suarez”, Mexico City 14269, Mexico
| | - Rodrigo A. Velasco-Martínez
- Laboratorio de Reprogramación Celular del Instituto de Fisiología Celular, UNAM, en el Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suarez”, Mexico City 14269, Mexico
| | - Laura Adalid-Peralta
- Laboratorio de Reprogramación Celular del Instituto de Fisiología Celular, UNAM, en el Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suarez”, Mexico City 14269, Mexico
| |
Collapse
|
12
|
Nguyen HD, Kim WK, Huong Vu G. Molecular mechanisms implicated in protein changes in the Alzheimer's disease human hippocampus. Mech Ageing Dev 2024; 219:111930. [PMID: 38554950 DOI: 10.1016/j.mad.2024.111930] [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: 12/03/2023] [Revised: 01/18/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
This study aimed to elucidate the specific biochemical pathways linked to changes in proteins in the Alzheimer's disease (AD) human hippocampus. Our data demonstrate a constant rise in the expression of four proteins (VGF, GFAP, HSPB1, and APP) across all eleven studies. Notably, UBC was the most centrally involved and had increased expression in the hippocampus tissue of individuals with AD. Modified proteins in the hippocampal tissue were found to activate the innate immune system and disrupt communication across chemical synapses. Four hub proteins (CD44, APP, ITGB2, and APOE) are connected to amyloid plaques, whereas two hub proteins (RPL24 and RPS23) are related to neurofibrillary tangles (NFTs). The presence of modified proteins was discovered to trigger the activation of microglia and decrease the functioning of ribosomes and mitochondria in the hippocampus. Three significant microRNAs (hsa-miR-106b-5p, hsa-miR-17-5p, and hsa-miR-16-5p) and transcription factors (MYT1L, PIN1, and CSRNP3) have been discovered to improve our understanding of the alterations in proteins within the hippocampal tissues that lead to the progression of AD. These findings establish a path for possible treatments for AD to employ therapeutic strategies that specifically focus on the proteins or processes linked to the illness.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Department of Pharmacy, College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea; Division of Microbiology, Tulane National Primate Research Center, Tulane University, Louisiana, USA.
| | - Woong-Ki Kim
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Louisiana, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Giang Huong Vu
- Department of Public Health, Hong Bang Health Center, Hai Phong, Vietnam
| |
Collapse
|
13
|
Shah A, Prasad S, Indoria A, Pal PK, Saini J, Ingalhalikar M. Free water imaging in Parkinson's disease and atypical parkinsonian disorders. J Neurol 2024; 271:2521-2528. [PMID: 38265472 DOI: 10.1007/s00415-024-12184-9] [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: 10/31/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Free water (FW)-corrected diffusion measures are more precise compared to standard diffusion measures. This study comprehensively evaluates FW and corrected diffusion metrics for whole brain white and deep gray matter (WM, GM) structures in patients with Parkinson's disease (PD), progressive supranuclear palsy (PSP) and multiple system atrophy (MSA) and attempts to ascertain the probable patterns of WM abnormalities. METHOD Diffusion MRI was acquired for subjects with PD (n = 133), MSA (n = 25), PSP (n = 30) and matched healthy controls (HC) (n = 99, n = 24, n = 12). Diffusion metrics of FA, MD, AD, RD were generated and FW, corrected FA maps were calculated using a bi-tensor model. TBSS was carried out at 5000 permutations with significance at p < 0.05. For GM, diffusivity maps were extracted from the basal ganglia, and analyzed at an FDR with p < 0.05. RESULTS Compared to HC, PD showed focal changes in FW. MSA showed changes in the cerebellum and brainstem, and PSP showed increase in FW involving supratentorial WM and midbrain. All three showed increased substantia nigra FW. MSA, PSP demonstrated increased FW in bilateral putamen. PD showed increased FW in left GP externa, and bilateral thalamus. Compared to HC, MSA had increased FW in bilateral GP interna, and left thalamic. PSP had an additional increase in FW of the right GP externa, right GP interna, and bilateral thalamus. CONCLUSION The present study demonstrated definitive differences in the patterns of FW alterations between PD and atypical parkinsonian disorders suggesting the possibility of whole brain FW maps being used as markers for diagnosis of these disorders.
Collapse
Affiliation(s)
- Apurva Shah
- Symbiosis Center for Medical Image Analysis and Symbiosis Institute of Technology, Symbiosis International University, Lavale, Mulshi, Pune, 412115, Maharashtra, India
| | - Shweta Prasad
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Hosur Road, Bengaluru, 560029, Karnataka, India
| | - Abhilasha Indoria
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Hosur Road, Bengaluru, 560029, Karnataka, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Hosur Road, Bengaluru, 560029, Karnataka, India
| | - Jitender Saini
- Department of Neuroimaging and Interventional Radiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Hosur Road, Bengaluru, 560029, Karnataka, India
| | - Madhura Ingalhalikar
- Symbiosis Center for Medical Image Analysis and Symbiosis Institute of Technology, Symbiosis International University, Lavale, Mulshi, Pune, 412115, Maharashtra, India.
| |
Collapse
|
14
|
Kim YA, Mellen M, Kizil C, Santa-Maria I. Mechanisms linking cerebrovascular dysfunction and tauopathy: Adding a layer of epiregulatory complexity. Br J Pharmacol 2024; 181:879-895. [PMID: 37926507 DOI: 10.1111/bph.16280] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 09/11/2023] [Accepted: 09/30/2023] [Indexed: 11/07/2023] Open
Abstract
Intracellular accumulation of hyperphosphorylated misfolded tau proteins are found in many neurodegenerative tauopathies, including Alzheimer's disease (AD). Tau pathology can impact cerebrovascular physiology and function through multiple mechanisms. In vitro and in vivo studies have shown that alterations in the blood-brain barrier (BBB) integrity and function can result in synaptic abnormalities and neuronal damage. In the present review, we will summarize how tau proteostasis dysregulation contributes to vascular dysfunction and, conversely, we will examine the factors and pathways leading to tau pathological alterations triggered by cerebrovascular dysfunction. Finally, we will highlight the role epigenetic and epitranscriptomic factors play in regulating the integrity of the cerebrovascular system and the progression of tauopathy including a few observartions on potential therapeutic interventions. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
- Yoon A Kim
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Marian Mellen
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain
| | - Caghan Kizil
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Ismael Santa-Maria
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain
| |
Collapse
|
15
|
Borrego–Écija S, Pérez‐Millan A, Antonell A, Fort‐Aznar L, Kaya‐Tilki E, León‐Halcón A, Lladó A, Molina‐Porcel L, Balasa M, Juncà‐Parella J, Vitorica J, Venero JL, Deierborg T, Boza‐Serrano A, Sánchez‐Valle R. Galectin-3 is upregulated in frontotemporal dementia patients with subtype specificity. Alzheimers Dement 2024; 20:1515-1526. [PMID: 38018380 PMCID: PMC10984429 DOI: 10.1002/alz.13536] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 11/30/2023]
Abstract
INTRODUCTION Neuroinflammation is a major contributor to the progression of frontotemporal dementia (FTD). Galectin-3 (Gal-3), a microglial activation regulator, holds promise as a therapeutic target and potential biomarker. Our study aimed to investigate Gal-3 levels in patients with FTD and assess its diagnostic potential. METHODS We examined Gal-3 levels in brain, serum, and cerebrospinal fluid (CSF) samples of patients with FTD and controls. Multiple linear regressions between Gal-3 levels and other FTD markers were explored. RESULTS Gal-3 levels were increased significantly in patients with FTD, mainly across brain tissue and CSF, compared to controls. Remarkably, Gal-3 levels were higher in cases with tau pathology than TAR-DNA Binding Protein 43 (TDP-43) pathology. Only MAPT mutation carriers displayed increased Gal-3 levels in CSF samples, which correlated with total tau and 14-3-3. DISCUSSION Our findings underscore the potential of Gal-3 as a diagnostic marker for FTD, particularly in MAPT cases, and highlights the relation of Gal-3 with neuronal injury markers.
Collapse
Affiliation(s)
- Sergi Borrego–Écija
- Alzheimer's disease and other cognitive disorders Unit. Service of Neurology, Fundació Recerca Clínic Barcelona‐IDIBAPSHospital Clínic de BarcelonaBarcelonaSpain
| | - Agnès Pérez‐Millan
- Alzheimer's disease and other cognitive disorders Unit. Service of Neurology, Fundació Recerca Clínic Barcelona‐IDIBAPSHospital Clínic de BarcelonaBarcelonaSpain
- Institut of Neurosciences. Faculty of Medicine and Medical SciencesUniversity of BarcelonaBarcelonaSpain
| | - Anna Antonell
- Alzheimer's disease and other cognitive disorders Unit. Service of Neurology, Fundació Recerca Clínic Barcelona‐IDIBAPSHospital Clínic de BarcelonaBarcelonaSpain
| | - Laura Fort‐Aznar
- Alzheimer's disease and other cognitive disorders Unit. Service of Neurology, Fundació Recerca Clínic Barcelona‐IDIBAPSHospital Clínic de BarcelonaBarcelonaSpain
| | - Elif Kaya‐Tilki
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad de Sevilla, Sevilla, SpainSevillaSpain
| | - Alberto León‐Halcón
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad de Sevilla, Sevilla, SpainSevillaSpain
- Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevillaSpain
| | - Albert Lladó
- Alzheimer's disease and other cognitive disorders Unit. Service of Neurology, Fundació Recerca Clínic Barcelona‐IDIBAPSHospital Clínic de BarcelonaBarcelonaSpain
- Institut of Neurosciences. Faculty of Medicine and Medical SciencesUniversity of BarcelonaBarcelonaSpain
| | - Laura Molina‐Porcel
- Alzheimer's disease and other cognitive disorders Unit. Service of Neurology, Fundació Recerca Clínic Barcelona‐IDIBAPSHospital Clínic de BarcelonaBarcelonaSpain
| | - Mircea Balasa
- Alzheimer's disease and other cognitive disorders Unit. Service of Neurology, Fundació Recerca Clínic Barcelona‐IDIBAPSHospital Clínic de BarcelonaBarcelonaSpain
| | - Jordi Juncà‐Parella
- Alzheimer's disease and other cognitive disorders Unit. Service of Neurology, Fundació Recerca Clínic Barcelona‐IDIBAPSHospital Clínic de BarcelonaBarcelonaSpain
| | - Javier Vitorica
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad de Sevilla, Sevilla, SpainSevillaSpain
- Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevillaSpain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Jose Luis Venero
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad de Sevilla, Sevilla, SpainSevillaSpain
- Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevillaSpain
| | - Tomas Deierborg
- Department of Experimental Medical Sciences, Experimental Neuroinflammatory LabLund UniversityLundSweden
| | - Antonio Boza‐Serrano
- Alzheimer's disease and other cognitive disorders Unit. Service of Neurology, Fundació Recerca Clínic Barcelona‐IDIBAPSHospital Clínic de BarcelonaBarcelonaSpain
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad de Sevilla, Sevilla, SpainSevillaSpain
- Instituto de Biomedicina de SevillaIBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevillaSpain
| | - Raquel Sánchez‐Valle
- Alzheimer's disease and other cognitive disorders Unit. Service of Neurology, Fundació Recerca Clínic Barcelona‐IDIBAPSHospital Clínic de BarcelonaBarcelonaSpain
- Institut of Neurosciences. Faculty of Medicine and Medical SciencesUniversity of BarcelonaBarcelonaSpain
| |
Collapse
|
16
|
Vellingiri B, Balasubramani K, Iyer M, Raj N, Elangovan A, Song K, Yeo HC, Jayakumar N, Kinoshita M, Thangarasu R, Narayanasamy A, Dayem AA, Prajapati VK, Gopalakrishnan AV, Cho SG. Role of Telomeres and Telomerase in Parkinson's Disease-A New Theranostics? Adv Biol (Weinh) 2023; 7:e2300097. [PMID: 37590305 DOI: 10.1002/adbi.202300097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/19/2023] [Indexed: 08/19/2023]
Abstract
Parkinson's disease (PD) is a complex condition that is significantly influenced by oxidative stress and inflammation. It is also suggested that telomere shortening (TS) is regulated by oxidative stress which leads to various diseases including age-related neurodegenerative diseases like PD. Thus, it is anticipated that PD would result in TS of peripheral blood mononuclear cells (PBMCs). Telomeres protect the ends of eukaryotic chromosomes preserving them against fusion and destruction. The TS is a normal process because DNA polymerase is unable to replicate the linear ends of the DNA due to end replication complications and telomerase activity in various cell types counteracts this process. PD is usually observed in the aged population and progresses over time therefore, disparities among telomere length in PBMCs of PD patients are recorded and it is still a question whether it has any useful role. Here, the likelihood of telomere attrition in PD and its implications concerning microglia activation, ageing, oxidative stress, and the significance of telomerase activators are addressed. Also, the possibility of telomeres and telomerase as a diagnostic and therapeutic biomarker in PD is discussed.
Collapse
Affiliation(s)
- Balachandar Vellingiri
- Stem Cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Kiruthika Balasubramani
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Mahalaxmi Iyer
- Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore, Tamil Nadu, 641021, India
| | - Neethu Raj
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Ajay Elangovan
- Stem Cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Kwonwoo Song
- Department of Stem Cell and Regenerative Biotechnology, Molecular and Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, 05029, Republic of Korea
| | - Han-Cheol Yeo
- Department of Stem Cell and Regenerative Biotechnology, Molecular and Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, 05029, Republic of Korea
| | - Namitha Jayakumar
- Department of Biotechnology, Sri Ramakrishna College of Arts and Science, Coimbatore, Tamil Nadu, 641006, India
| | - Masako Kinoshita
- Department of Neurology, National Hospital Organization Utano National Hospital, Ondoyama-Cho, Narutaki, Ukyo-Ku, Kyoto, 616-8255, Japan
| | - Ravimanickam Thangarasu
- Department of Zoology, School of Science, Tamil Nadu Open University, Saidapet, Chennai, 600015, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular and Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, 05029, Republic of Korea
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular and Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, 05029, Republic of Korea
| |
Collapse
|
17
|
Gao C, Jiang J, Tan Y, Chen S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct Target Ther 2023; 8:359. [PMID: 37735487 PMCID: PMC10514343 DOI: 10.1038/s41392-023-01588-0] [Citation(s) in RCA: 370] [Impact Index Per Article: 185.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 09/23/2023] Open
Abstract
Microglia activation is observed in various neurodegenerative diseases. Recent advances in single-cell technologies have revealed that these reactive microglia were with high spatial and temporal heterogeneity. Some identified microglia in specific states correlate with pathological hallmarks and are associated with specific functions. Microglia both exert protective function by phagocytosing and clearing pathological protein aggregates and play detrimental roles due to excessive uptake of protein aggregates, which would lead to microglial phagocytic ability impairment, neuroinflammation, and eventually neurodegeneration. In addition, peripheral immune cells infiltration shapes microglia into a pro-inflammatory phenotype and accelerates disease progression. Microglia also act as a mobile vehicle to propagate protein aggregates. Extracellular vesicles released from microglia and autophagy impairment in microglia all contribute to pathological progression and neurodegeneration. Thus, enhancing microglial phagocytosis, reducing microglial-mediated neuroinflammation, inhibiting microglial exosome synthesis and secretion, and promoting microglial conversion into a protective phenotype are considered to be promising strategies for the therapy of neurodegenerative diseases. Here we comprehensively review the biology of microglia and the roles of microglia in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, multiple system atrophy, amyotrophic lateral sclerosis, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration, dementia with Lewy bodies and Huntington's disease. We also summarize the possible microglia-targeted interventions and treatments against neurodegenerative diseases with preclinical and clinical evidence in cell experiments, animal studies, and clinical trials.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jingwen Jiang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yuyan Tan
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
18
|
Dutta D, Jana M, Paidi RK, Majumder M, Raha S, Dasarathy S, Pahan K. Tau fibrils induce glial inflammation and neuropathology via TLR2 in Alzheimer's disease-related mouse models. J Clin Invest 2023; 133:e161987. [PMID: 37552543 PMCID: PMC10503811 DOI: 10.1172/jci161987] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
Glial activation and inflammation coincide with neurofibrillary tangle (NFT) formation in neurons. However, the mechanism behind the interaction between tau fibrils and glia is poorly understood. Here, we found that tau preformed fibrils (PFFs) caused induction of inflammation in microglia by specifically activating the TLR2/MyD88, but not the TLR4/MyD88, pathway. Accordingly, the WT TLR2-interacting domain of MyD88 (wtTIDM) peptide inhibited tau PFF-induced activation of the TLR2/MyD88/NF-κB pathway, resulting in reduced inflammation. Nasal administration of wtTIDM in P301S tau-expressing PS19 mice was found to inhibit gliosis and inflammatory markers, as well as to reduce pathogenic tau in the hippocampus, resulting in improved cognitive behavior in PS19 mice. The inhibitory effect of wtTIDM on tau pathology was absent in PS19 mice lacking TLR2, reinforcing the essential involvement of TLR2 in wtTIDM-mediated effects in vivo. Studying the mechanism further, we found that the tau promoter harbored a potential NF-κB-binding site and that proinflammatory molecules increased transcription of tau in neurons via NF-κB. These results suggest that tau-induced neuroinflammation and neuropathology require TLR2 and that neuroinflammation directly upregulates tau in neurons via NF-κB, highlighting a direct connection between inflammation and tauopathy.
Collapse
Affiliation(s)
- Debashis Dutta
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Malabendu Jana
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Ramesh Kumar Paidi
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Moumita Majumder
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sumita Raha
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Sridevi Dasarathy
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
19
|
Rao YL, Ganaraja B, Suresh PK, Joy T, Ullal SD, Manjrekar PA, Murlimanju BV, Sharma BG. Effect of resveratrol and combination of resveratrol and donepezil on the expression of microglial cells and astrocytes in Wistar albino rats of colchicine-induced Alzheimer's disease. 3 Biotech 2023; 13:319. [PMID: 37641690 PMCID: PMC10460340 DOI: 10.1007/s13205-023-03743-4] [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: 06/09/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
Aim The goal was to evaluate the effect of resveratrol (RS) and combination therapy of RS and donepezil (DPZ), on the numerical expression of microglial cells and astrocytes, in the frontal cortex, regions of the hippocampus in colchicine-induced Alzheimer's disease (AD) model. Methods The study involved male albino Wistar rats of three months, age and consisted of 6 groups, with six animals each. The immunohistochemical staining with mouse monoclonal anti-human CD 68 and mouse monoclonal anti-GFAP was performed to assess the number of microglial cells and astrocytes, respectively. Results AD group showed an increase in the number of microglia, and the numbers declined in the treatment groups, RS 10, RS 20, RS10/10 and DPZ + RS (p < 0.001). Astrocyte count was increased in the treatment groups in contrast to the AD group (p < 0.05). The DPZ + RS combination group revealed substantial elevation in the number of astrocytes and decreased microglial number among all the groups (p < 0.001). Conclusion RS administration has diminished the microglial number and elevated the number of astrocytes. The elevated reactive astrocytes have decreased the microglial population. However, the limitation of our study is utilizing the colchicine for the induction of neurodegeneration. Using the transgenic models of AD may give a better insight into the pathogenesis and effect of RS. Another limitation of this study is the administration of RS and DPZ through different routes. The prospects of this research include studying the probiotic nature of RS and the effect of RS in other neurodegenerative disorders.
Collapse
Affiliation(s)
- Y. Lakshmisha Rao
- Department of Anatomy, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - B. Ganaraja
- Department of Physiology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - Pooja K. Suresh
- Department of Pathology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - Teresa Joy
- Department of Anatomy, American University of Antigua College of Medicine, University Park, Jabberwock Beach Road, Coolidge, Antigua, West Indies Antigua and Barbuda
| | - Sheetal D. Ullal
- Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - Poornima A. Manjrekar
- Department of Biochemistry, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - B. V. Murlimanju
- Department of Anatomy, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - B. Gaurav Sharma
- Senior Registrar in Trauma and Orthopaedic Surgery, Hampshire Hospitals NHS Foundation Trust, Basingstoke and North Hampshire Hospital, Aldermaston Road, Basingstoke, RG24 9NA UK
| |
Collapse
|
20
|
Wang L, Wei Y, Sun Z, Jiang LH, Yin Y, Zheng P, Fu Y, Wang H, Li C, Wang JZ. DpdtpA, A Multi-metal Ion Chelator, Attenuates Tau Phosphorylation and Microglial Inflammatory Response via Regulating the PI3K/AKT/GSK-3β Signal Pathways. Neuroscience 2023; 526:196-203. [PMID: 37419407 DOI: 10.1016/j.neuroscience.2023.07.004] [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: 03/02/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023]
Abstract
Tau protein hyperphosphorylation and formation of intracellular neurofibrillary tangles (NFTs) are one of the histopathological hallmarks of Alzheimer's disease (AD) and positively correlated with the severity of AD symptoms. NFTs contain a large number of metal ions that play an important role in regulating tau protein phosphorylation and AD progression. Extracellular tau induces primary phagocytosis of stressed neurons and neuronal loss by activating microglia. Here, we studied the effects of a multi-metal ion chelator, DpdtpA, on tau-induced microglial activation and inflammatory responses and the underlying mechanisms. Treatment with DpdtpA attenuated the increase in the expression of NF-κB and production of inflammatory cytokines, IL-1β, IL-6 and IL-10, in rat microglial cells induced by expression of human tau40 proteins. Treatment with DpdtpA also suppressed tau protein expression and phosphorylation. Moreover, treatment with DpdtpA prevented tau-induced activation of glycogen synthase kinase-3β (GSK-3β) and inhibition of phosphatidylinositol-3-hydroxy kinase (PI3K)/AKT. Collectively, these results show that DpdtpA can attenuate tau phosphorylation and inflammatory responses of microglia by regulating the PI3K/AKT/GSK-3β signal pathways, providing a new option to alleviate neuroinflammation for the treatment of AD.
Collapse
Affiliation(s)
- Lu Wang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China; Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China.
| | - Yingjuan Wei
- Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Zhenzhou Sun
- Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Xinxiang Medical University, Xinxiang, China
| | - Yaling Yin
- Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Panpan Zheng
- Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Yun Fu
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| | - Hongwei Wang
- Department of Physiology and Pathophysiology, Xinxiang Medical University, Xinxiang, China
| | - Changzheng Li
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang, China
| | - Jian-Zhi Wang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China; Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Leńska-Mieciek M, Madetko-Alster N, Alster P, Królicki L, Fiszer U, Koziorowski D. Inflammation in multiple system atrophy. Front Immunol 2023; 14:1214677. [PMID: 37426656 PMCID: PMC10327640 DOI: 10.3389/fimmu.2023.1214677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Misfolding protein aggregation inside or outside cells is the major pathological hallmark of several neurodegenerative diseases. Among proteinopathies are neurodegenerative diseases with atypical Parkinsonism and an accumulation of insoluble fibrillary alpha-synuclein (synucleinopathies) or hyperphosphorylated tau protein fragments (tauopathies). As there are no therapies available to slow or halt the progression of these disea ses, targeting the inflammatory process is a promising approach. The inflammatory biomarkers could also help in the differential diagnosis of Parkinsonian syndromes. Here, we review inflammation's role in multiple systems atrophy pathogenesis, diagnosis, and treatment.
Collapse
Affiliation(s)
- Marta Leńska-Mieciek
- Department of Neurology and Epileptology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | | | - Piotr Alster
- Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Królicki
- Department of Nuclear Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Urszula Fiszer
- Department of Neurology and Epileptology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | | |
Collapse
|
22
|
Gao L, Huang C, Li H, Wu S, Zhou X, Ying C. Exploring the molecular targets for Type 2 diabetes-induced Alzheimer's disease through bioinformatics analysis. Epigenomics 2023; 15:619-633. [PMID: 37554106 DOI: 10.2217/epi-2023-0149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023] Open
Abstract
Aim: The purpose of this study was to elucidate the potential mechanisms of Alzheimer's disease (AD) induced by Type 2 diabetes mellitus (T2DM) through bioinformatics analysis, to provide new treatment targets for this disease. Methods: We used weighted gene coexpression network analysis and differentially expressed genes analysis to identify significantly differentially expressed genes shared by T2DM and AD. Molecular docking was used to predict possible protein targets for T2DM-induced AD. Results: The direct interaction of CD44 and STAT3 may play a significant role in the development of T2DM-induced AD. Conclusion: A new approach to treating T2DM-associated AD may be provided by these hub genes and their predicted molecular targets.
Collapse
Affiliation(s)
- Lin Gao
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Chengyu Huang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Hui Li
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Shidi Wu
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Xiaoyan Zhou
- Xuzhou Engineering Research Center of Medical Genetics & Transformation, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Changjiang Ying
- Xuzhou Engineering Research Center of Medical Genetics & Transformation, Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| |
Collapse
|
23
|
Kemppainen S, Huber N, Willman RM, Zamora A, Mäkinen P, Martiskainen H, Takalo M, Haapasalo A, Sobrino T, González Gómez MA, Piñeiro Y, Rivas J, Himmelreich U, Hiltunen M. Organotypic Hippocampal Slice Cultures from Adult Tauopathy Mice and Theragnostic Evaluation of Nanomaterial Phospho-TAU Antibody-Conjugates. Cells 2023; 12:1422. [PMID: 37408256 DOI: 10.3390/cells12101422] [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: 04/21/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 07/07/2023] Open
Abstract
Organotypic slice culture models surpass conventional in vitro methods in many aspects. They retain all tissue-resident cell types and tissue hierarchy. For studying multifactorial neurodegenerative diseases such as tauopathies, it is crucial to maintain cellular crosstalk in an accessible model system. Organotypic slice cultures from postnatal tissue are an established research tool, but adult tissue-originating systems are missing, yet necessary, as young tissue-originating systems cannot fully model adult or senescent brains. To establish an adult-originating slice culture system for tauopathy studies, we made hippocampal slice cultures from transgenic 5-month-old hTau.P301S mice. In addition to the comprehensive characterization, we set out to test a novel antibody for hyperphosphorylated TAU (pTAU, B6), with and without a nanomaterial conjugate. Adult hippocampal slices retained intact hippocampal layers, astrocytes, and functional microglia during culturing. The P301S-slice neurons expressed pTAU throughout the granular cell layer and secreted pTAU to the culture medium, whereas the wildtype slices did not. Additionally, cytotoxicity and inflammation-related determinants were increased in the P301S slices. Using fluorescence microscopy, we showed target engagement of the B6 antibody to pTAU-expressing neurons and a subtle but consistent decrease in intracellular pTAU with the B6 treatment. Collectively, this tauopathy slice culture model enables measuring the extracellular and intracellular effects of different mechanistic or therapeutic manipulations on TAU pathology in adult tissue without the hindrance of the blood-brain barrier.
Collapse
Affiliation(s)
- Susanna Kemppainen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Nadine Huber
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Roosa-Maria Willman
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Ana Zamora
- Molecular Imaging and Photonics, KU Leuven, 3001 Leuven, Belgium
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Petra Mäkinen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Henna Martiskainen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Annakaisa Haapasalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Manuel Antonio González Gómez
- Institute of Materials, Applied Physics Department, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Yolanda Piñeiro
- Institute of Materials, Applied Physics Department, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José Rivas
- Institute of Materials, Applied Physics Department, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
24
|
Yang J, Ou W, Jagadeesan N, Simanauskaite J, Sun J, Castellanos D, Cribbs DH, Sumbria RK. The Effects of a Blood-Brain Barrier Penetrating Erythropoietin in a Mouse Model of Tauopathy. Pharmaceuticals (Basel) 2023; 16:ph16040558. [PMID: 37111315 PMCID: PMC10141171 DOI: 10.3390/ph16040558] [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: 02/23/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Erythropoietin (EPO), a hematopoietic neurotrophin, is a potential therapeutic for Alzheimer's disease (AD) but has limited blood-brain barrier (BBB) permeability. EPO fused to a chimeric transferrin receptor monoclonal antibody (cTfRMAb) enters the brain via TfR-mediated transcytosis across the BBB. We previously showed that cTfRMAb-EPO is protective in a mouse model of amyloidosis, but its effects on tauopathy are not known. Given that amyloid and tau pathology are characteristics of AD, the effects of cTfRMAb-EPO were studied in a tauopathy mouse model (PS19). Six-month-old PS19 mice were injected intraperitoneally with either saline (PS19-Saline; n = 9) or cTfRMAb-EPO (PS19-cTfRMAb-EPO, 10 mg/kg; n = 10); every two or three days on alternate weeks for 8 weeks. Age-matched, saline-treated, wildtype littermates (WT-Saline; n = 12) were injected using the same protocol. After 8 weeks, locomotion, hyperactivity, and anxiety were assessed via the open-field test, and brains were harvested and sectioned. Cerebral cortex, hippocampus, amygdala, and entorhinal cortex sections were analyzed for phospho-tau (AT8) and microgliosis (Iba1). Hippocampal cellular density (H&E) was also assessed. PS19-Saline mice were hyperactive and less anxious compared to WT-Saline mice, and these behavioral phenotypes were significantly reduced in the PS19-cTfRMAb-EPO mice compared to the PS19-Saline mice. cTfRMAb-EPO significantly reduced AT8 load by ≥50% in all of the brain regions analyzed and microgliosis in the entorhinal cortex and amygdala compared to the PS19-Saline mice. Hippocampal pyramidal and granule cell layer density did not differ significantly between the PS19-cTfRMAb-EPO and PS19-Saline mice. This proof-of-concept study demonstrates the therapeutic effects of the BBB-penetrating cTfRMAb-EPO in PS19 mice.
Collapse
Affiliation(s)
- Joshua Yang
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Dr, Claremont, CA 91711, USA
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | - Weijun Ou
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | - Nataraj Jagadeesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | | | - Jiahong Sun
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | - Demi Castellanos
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Dr, Claremont, CA 91711, USA
| | - David H Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Rachita K Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
- Department of Neurology, University of California, Irvine, CA 92868, USA
| |
Collapse
|
25
|
Torok J, Anand C, Verma P, Raj A. Connectome-based biophysics models of Alzheimer's disease diagnosis and prognosis. Transl Res 2023; 254:13-23. [PMID: 36031051 PMCID: PMC11019890 DOI: 10.1016/j.trsl.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022]
Abstract
With the increasing prevalence of Alzheimer's disease (AD) among aging populations and the limited therapeutic options available to slow or reverse its progression, the need has never been greater for improved diagnostic tools for identifying patients in the preclinical and prodomal phases of AD. Biophysics models of the connectome-based spread of amyloid-beta (Aβ) and microtubule-associated protein tau (τ) have enjoyed recent success as tools for predicting the time course of AD-related pathological changes. However, given the complex etiology of AD, which involves not only connectome-based spread of protein pathology but also the interactions of many molecular and cellular players over multiple spatiotemporal scales, more robust, complete biophysics models are needed to better understand AD pathophysiology and ultimately provide accurate patient-specific diagnoses and prognoses. Here we discuss several areas of active research in AD whose insights can be used to enhance the mathematical modeling of AD pathology as well as recent attempts at developing improved connectome-based biophysics models. These efforts toward a comprehensive yet parsimonious mathematical description of AD hold great promise for improving both the diagnosis of patients at risk for AD and our mechanistic understanding of how AD progresses.
Collapse
Affiliation(s)
- Justin Torok
- Department of Radiology, University of California, San Francisco, San Francisco, California.
| | - Chaitali Anand
- Department of Radiology, University of California, San Francisco, San Francisco, California
| | - Parul Verma
- Department of Radiology, University of California, San Francisco, San Francisco, California
| | - Ashish Raj
- Department of Radiology, University of California, San Francisco, San Francisco, California; Department of Bioengineering, University of California, Berkeley and University of California, San Francisco, Berkeley, California; Department of Radiology, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
26
|
Lau V, Ramer L, Tremblay MÈ. An aging, pathology burden, and glial senescence build-up hypothesis for late onset Alzheimer's disease. Nat Commun 2023; 14:1670. [PMID: 36966157 PMCID: PMC10039917 DOI: 10.1038/s41467-023-37304-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 03/07/2023] [Indexed: 03/27/2023] Open
Abstract
Alzheimer's disease (AD) predominantly occurs as a late onset (LOAD) form involving neurodegeneration and cognitive decline with progressive memory loss. Risk factors that include aging promote accumulation of AD pathologies, such as amyloid-beta and tau aggregates, as well as inflammation and oxidative stress. Homeostatic glial states regulate and suppress pathology buildup; inflammatory states exacerbate pathology by releasing pro-inflammatory cytokines. Multiple stresses likely induce glial senescence, which could decrease supportive functions and reinforce inflammation. In this perspective, we hypothesize that aging first drives AD pathology burden, whereafter AD pathology putatively induces glial senescence in LOAD. We hypothesize that increasing glial senescence, particularly local senescent microglia accumulation, sustains and drives perpetuating buildup and spread of AD pathologies, glial aging, and further senescence. We predict that increasing glial senescence, particularly local senescent microglia accumulation, also transitions individuals from healthy cognition into mild cognitive impairment and LOAD diagnosis. These pathophysiological underpinnings may centrally contribute to LOAD onset, but require further mechanistic investigation.
Collapse
Affiliation(s)
- Victor Lau
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| | - Leanne Ramer
- Department of Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, BC, Canada.
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada.
- Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada.
- The Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
| |
Collapse
|
27
|
Woollacott IOC, Swift IJ, Sogorb‐Esteve A, Heller C, Knowles K, Bouzigues A, Russell LL, Peakman G, Greaves CV, Convery R, Heslegrave A, Rowe JB, Borroni B, Galimberti D, Tiraboschi P, Masellis M, Tartaglia MC, Finger E, van Swieten JC, Seelaar H, Jiskoot L, Sorbi S, Butler CR, Graff C, Gerhard A, Laforce R, Sanchez‐Valle R, de Mendonça A, Moreno F, Synofzik M, Vandenberghe R, Ducharme S, Ber IL, Levin J, Otto M, Pasquier F, Santana I, Zetterberg H, Rohrer JD, the Genetic FTD Initiative, GENFI NelsonAnnabelBocchettaMartinaCashDavidThomasDavid L.ToddEmilyBenotmaneHanyaNicholasJenniferSamraKiranShafeiRachelleTimberlakeCarolynCopeThomasRittmanTimothyBenussiAlbertoPremiEnricoGasparottiRobertoArchettiSilvanaGazzinaStefanoCantoniValentinaArighiAndreaFenoglioChiaraScarpiniElioFumagalliGiorgioBorracciVittoriaRossiGiacominaGiacconeGiorgioDi FedeGiuseppeCaroppoPaolaPrioniSaraRedaelliVeronicaTang‐WaiDavidRogaevaEkaterinaCastelo‐BrancoMiguelFreedmanMorrisKerenRonBlackSandraMitchellSaraShoesmithChristenBarthaRobartRademakersRosaPoosJackiePapmaJanne M.GianniniLuciavan MinkelenRickPijnenburgYolandeNacmiasBenedettaFerrariCamillaPolitoCristinaLombardiGemmaBessiValentinaVeldsmanMicheleAnderssonChristinThonbergHakanÖijerstedtLinnJelicVesnaThompsonPaulLangheinrichTobiasLladóAlbertAntonellAnnaOlivesJaumeBalasaMirceaBargallóNuriaBorrego‐EcijaSergiVerdelhoAnaMarutaCarolinaFerreiraCatarina B.MiltenbergerGabrieldo CoutoFrederico SimõesGabilondoAlazneGorostidiAnaVillanuaJorgeCañadaMartaTaintaMikelZulaicaMirenBarandiaranMyriamAlvesPatriciaBenderBenjaminWilkeCarloGrafLisaVogelsAnnickVandenbulckeMathieuVan DammePhilipBruffaertsRosePoesenKoenRosa‐NetoPedroGauthierSergeCamuzatAgnèsBriceAlexisBertrandAnneFunkiewiezAurélieRinaldiDaisySaracinoDarioColliotOlivierSayahSabrinaPrixCatharinaWlasichElisabethWagemannOliviaLoosliSandraSchöneckerSonjaHoegenTobiasLombardiJolinaAnderl‐StraubSarahRollinAdelineKuchcinskiGregoryBertouxMaximeLebouvierThibaudDeramecourtVincentSantiagoBeatrizDuroDianaLeitãoMaria JoãoAlmeidaMaria RosarioTábuas‐PereiraMiguelAfonsoSónia. CSF glial markers are elevated in a subset of patients with genetic frontotemporal dementia. Ann Clin Transl Neurol 2022; 9:1764-1777. [PMID: 36245297 PMCID: PMC9639635 DOI: 10.1002/acn3.51672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Neuroinflammation has been shown to be an important pathophysiological disease mechanism in frontotemporal dementia (FTD). This includes activation of microglia, a process that can be measured in life through assaying different glia-derived biomarkers in cerebrospinal fluid. However, only a few studies so far have taken place in FTD, and even fewer focusing on the genetic forms of FTD. METHODS We investigated the cerebrospinal fluid concentrations of TREM2, YKL-40 and chitotriosidase using immunoassays in 183 participants from the Genetic FTD Initiative (GENFI) study: 49 C9orf72 (36 presymptomatic, 13 symptomatic), 49 GRN (37 presymptomatic, 12 symptomatic) and 23 MAPT (16 presymptomatic, 7 symptomatic) mutation carriers and 62 mutation-negative controls. Concentrations were compared between groups using a linear regression model adjusting for age and sex, with 95% bias-corrected bootstrapped confidence intervals. Concentrations in each group were correlated with the Mini-Mental State Examination (MMSE) score using non-parametric partial correlations adjusting for age. Age-adjusted z-scores were also created for the concentration of markers in each participant, investigating how many had a value above the 95th percentile of controls. RESULTS Only chitotriosidase in symptomatic GRN mutation carriers had a concentration significantly higher than controls. No group had higher TREM2 or YKL-40 concentrations than controls after adjusting for age and sex. There was a significant negative correlation of chitotriosidase concentration with MMSE in presymptomatic GRN mutation carriers. In the symptomatic groups, for TREM2 31% of C9orf72, 25% of GRN, and 14% of MAPT mutation carriers had a concentration above the 95th percentile of controls. For YKL-40 this was 8% C9orf72, 8% GRN and 0% MAPT mutation carriers, whilst for chitotriosidase it was 23% C9orf72, 50% GRN, and 29% MAPT mutation carriers. CONCLUSIONS Although chitotriosidase concentrations in GRN mutation carriers were the only significantly raised glia-derived biomarker as a group, a subset of mutation carriers in all three groups, particularly for chitotriosidase and TREM2, had elevated concentrations. Further work is required to understand the variability in concentrations and the extent of neuroinflammation across the genetic forms of FTD. However, the current findings suggest limited utility of these measures in forthcoming trials.
Collapse
Affiliation(s)
- Ione O. C. Woollacott
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
| | - Imogen J. Swift
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
- UK Dementia Research Institute at UCLLondonUnited Kingdom
| | - Aitana Sogorb‐Esteve
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
- UK Dementia Research Institute at UCLLondonUnited Kingdom
| | - Carolin Heller
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
- UK Dementia Research Institute at UCLLondonUnited Kingdom
| | - Kathryn Knowles
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
- UK Dementia Research Institute at UCLLondonUnited Kingdom
| | - Arabella Bouzigues
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
| | - Lucy L. Russell
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
| | - Georgia Peakman
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
| | - Caroline V. Greaves
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
| | - Rhian Convery
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
| | | | - James B. Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust and Medical Research Council Cognition and Brain Sciences UnitUniversity of CambridgeCambridgeUnited Kingdom
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental SciencesUniversity of MilanMilanItaly
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
| | | | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research InstituteUniversity of TorontoTorontoCanada
| | | | - Elizabeth Finger
- Department of Clinical Neurological SciencesUniversity of Western OntarioLondonOntarioCanada
| | | | - Harro Seelaar
- Department of NeurologyErasmus Medical CentreRotterdamThe Netherlands
| | - Lize Jiskoot
- Department of NeurologyErasmus Medical CentreRotterdamThe Netherlands
| | - Sandro Sorbi
- Department of NeurofarbaUniversity of FlorenceFlorenceItaly
- IRCCS Fondazione Don Carlo GnocchiFlorenceItaly
| | - Chris R. Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences DivisionUniversity of OxfordOxfordUnited Kingdom
- Department of Brain SciencesImperial College LondonUnited Kingdom
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of NeurobiologyCare Sciences and Society, Bioclinicum, Karolinska InstitutetSolnaSweden
- Unit for Hereditary Dementias, Theme AgingKarolinska University HospitalSolnaSweden
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging CentreUniversity of ManchesterManchesterUnited Kingdom
- Departments of Geriatric Medicine and Nuclear MedicineUniversity of Duisburg‐EssenEssenGermany
- Cerebral Function Unit, Manchester Centre for Clinical NeurosciencesSalford Royal NHS Foundation TrustSalfordUnited Kingdom
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de MédecineUniversité LavalQuébecCanada
| | - Raquel Sanchez‐Valle
- Alzheimer's disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I SunyerUniversity of BarcelonaBarcelonaSpain
| | | | - Fermin Moreno
- Cognitive Disorders Unit, Department of NeurologyDonostia University HospitalSan SebastianGipuzkoaSpain
- Neuroscience AreaBiodonostia Health Research InstituteSan SebastianGipuzkoaSpain
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie‐Institute for Clinical Brain Research and Center of NeurologyUniversity of TübingenTübingenGermany
- Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of NeurosciencesKU LeuvenLeuvenBelgium
- Neurology ServiceUniversity Hospitals LeuvenLeuvenBelgium
- Leuven Brain Institute, KU LeuvenLeuvenBelgium
| | - Simon Ducharme
- Douglas Mental Health University Institute, Department of PsychiatryMcGill UniversityMontrealCanada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Department of Neurology & NeurosurgeryMcGill UniversityMontrealCanada
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute – Institut du Cerveau – ICM, Inserm U1127, CNRS UMR 7225, AP‐HP ‐ Hôpital Pitié‐SalpêtrièreParisFrance
- Centre de référence des démences rares ou précoces, IM2A, Département de NeurologieAP‐HP ‐ Hôpital Pitié‐SalpêtrièreParisFrance
- Département de NeurologieAP‐HP ‐ Hôpital Pitié‐SalpêtrièreParisFrance
| | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig‐Maximilians‐UniversitätMunichGermany
- Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster of Systems NeurologyMunichGermany
| | - Markus Otto
- Department of NeurologyUniversity of UlmUlmGermany
| | - Florence Pasquier
- Univ LilleLilleFrance
- Inserm 1172LilleFrance
- CHU, CNR‐MAJ, Labex Distalz, LiCEND LilleLilleFrance
| | - Isabel Santana
- Neurology Service, Faculty of MedicineUniversity Hospital of Coimbra (HUC), University of CoimbraCoimbraPortugal
- Center for Neuroscience and Cell Biology, Faculty of MedicineUniversity of CoimbraCoimbraPortugal
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCLLondonUnited Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Hong Kong Center for Neurodegenerative DiseasesClear Water Bay, Hong KongChina
| | - Jonathan D. Rohrer
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of Neurology, Queen SquareLondonUnited Kingdom
- UK Dementia Research Institute at UCLLondonUnited Kingdom
| | | |
Collapse
|
28
|
Wu YG, Song LJ, Yin LJ, Yin JJ, Wang Q, Yu JZ, Xiao BG, Ma CG. The effects and potential of microglial polarization and crosstalk with other cells of the central nervous system in the treatment of Alzheimer's disease. Neural Regen Res 2022; 18:947-954. [PMID: 36254973 PMCID: PMC9827789 DOI: 10.4103/1673-5374.355747] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Microglia are resident immune cells in the central nervous system. During the pathogenesis of Alzheimer's disease, stimulatory factors continuously act on the microglia causing abnormal activation and unbalanced phenotypic changes; these events have become a significant and promising area of research. In this review, we summarize the effects of microglial polarization and crosstalk with other cells in the central nervous system in the treatment of Alzheimer's disease. Our literature search found that phenotypic changes occur continuously in Alzheimer's disease and that microglia exhibit extensive crosstalk with astrocytes, oligodendrocytes, neurons, and penetrated peripheral innate immune cells via specific signaling pathways and cytokines. Collectively, unlike previous efforts to modulate microglial phenotypes at a single level, targeting the phenotypes of microglia and the crosstalk with other cells in the central nervous system may be more effective in reducing inflammation in the central nervous system in Alzheimer's disease. This would establish a theoretical basis for reducing neuronal death from central nervous system inflammation and provide an appropriate environment to promote neuronal regeneration in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Yi-Ge Wu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China,Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Li-Jun Yin
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
| | - Jun-Jun Yin
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China,Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China
| | - Jie-Zhong Yu
- Institute of Brain Science/Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases/Medical School, Shanxi Datong University, Datong, Shanxi Province, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Cun-Gen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, Shanxi Province, China,Institute of Brain Science/Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases/Medical School, Shanxi Datong University, Datong, Shanxi Province, China,Correspondence to: Cun-Gen Ma, .
| |
Collapse
|
29
|
Faridar A, Vasquez M, Thome AD, Yin Z, Xuan H, Wang JH, Wen S, Li X, Thonhoff JR, Zhao W, Zhao H, Beers DR, Wong STC, Masdeu JC, Appel SH. Ex vivo expanded human regulatory T cells modify neuroinflammation in a preclinical model of Alzheimer's disease. Acta Neuropathol Commun 2022; 10:144. [PMID: 36180898 PMCID: PMC9524037 DOI: 10.1186/s40478-022-01447-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Background Regulatory T cells (Tregs) play a neuroprotective role by suppressing microglia and macrophage-mediated inflammation and modulating adaptive immune reactions. We previously documented that Treg immunomodulatory mechanisms are compromised in Alzheimer’s disease (AD). Ex vivo expansion of Tregs restores and amplifies their immunosuppressive functions in vitro. A key question is whether adoptive transfer of ex vivo expanded human Tregs can suppress neuroinflammation and amyloid pathology in a preclinical mouse model. Methods An immunodeficient mouse model of AD was generated by backcrossing the 5xFAD onto Rag2 knockout mice (5xFAD-Rag2KO). Human Tregs were expanded ex vivo for 24 days and administered to 5xFAD-Rag2KO. Changes in amyloid burden, microglia characteristics and reactive astrocytes were evaluated using ELISA and confocal microscopy. NanoString Mouse AD multiplex gene expression analysis was applied to explore the impact of ex vivo expanded Tregs on the neuroinflammation transcriptome. Results Elimination of mature B and T lymphocytes and natural killer cells in 5xFAD-Rag2KO mice was associated with upregulation of 95 inflammation genes and amplified number of reactive microglia within the dentate gyrus. Administration of ex vivo expanded Tregs reduced amyloid burden and reactive glial cells in the dentate gyrus and frontal cortex of 5xFAD-Rag2KO mice. Interrogation of inflammation gene expression documented down-regulation of pro-inflammatory cytokines (IL1A&B, IL6), complement cascade (C1qa, C1qb, C1qc, C4a/b), toll-like receptors (Tlr3, Tlr4 and Tlr7) and microglial activations markers (CD14, Tyrobp,Trem2) following Treg administration. Conclusions Ex vivo expanded Tregs with amplified immunomodulatory function, suppressed neuroinflammation and alleviated AD pathology in vivo. Our results provide preclinical evidences for Treg cell therapy as a potential treatment strategy in AD. Supplementary Information The online version contains supplementary material available at 10.1186/s40478-022-01447-z.
Collapse
Affiliation(s)
- Alireza Faridar
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Matthew Vasquez
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - Aaron D Thome
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Zheng Yin
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - Hui Xuan
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Jing Hong Wang
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Shixiang Wen
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Xuping Li
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston, TX, USA
| | - Jason R Thonhoff
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Weihua Zhao
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Hong Zhao
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - David R Beers
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Stephen T C Wong
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - Joseph C Masdeu
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Stanley H Appel
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA.
| |
Collapse
|
30
|
Odfalk KF, Bieniek KF, Hopp SC. Microglia: Friend and foe in tauopathy. Prog Neurobiol 2022; 216:102306. [PMID: 35714860 PMCID: PMC9378545 DOI: 10.1016/j.pneurobio.2022.102306] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 04/24/2022] [Accepted: 06/10/2022] [Indexed: 12/16/2022]
Abstract
Aggregation of misfolded microtubule associated protein tau into abnormal intracellular inclusions defines a class of neurodegenerative diseases known as tauopathies. The consistent spatiotemporal progression of tau pathology in Alzheimer's disease (AD) led to the hypothesis that tau aggregates spread in the brain via bioactive tau "seeds" underlying advancing disease course. Recent studies implicate microglia, the resident immune cells of the central nervous system, in both negative and positive regulation of tau pathology. Polymorphisms in genes that alter microglial function are associated with the development of AD and other tauopathies. Experimental manipulation of microglia function can alter tau pathology and microglia-mediated neuroinflammatory cascades can exacerbate tau pathology. Microglia also exert protective functions by mitigating tau spread: microglia internalize tau seeds and have the capacity to degrade them. However, when microglia fail to degrade these tau seeds there are deleterious consequences, including secretion of exosomes containing tau that can spread to neurons. This review explores the intersection of microglia and tau from the perspective of neuropathology, neuroimaging, genetics, transcriptomics, and molecular biology. As tau-targeted therapies such as anti-tau antibodies advance through clinical trials, it is critical to understand the interaction between tau and microglia.
Collapse
Affiliation(s)
- Kristian F Odfalk
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Kevin F Bieniek
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pathology and Laboratory Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
31
|
Zaki MO, El-Desouky S, Elsherbiny DA, Salama M, Azab SS. Glimepiride mitigates tauopathy and neuroinflammation in P301S transgenic mice: role of AKT/GSK3β signaling. Inflammopharmacology 2022; 30:1871-1890. [PMID: 35922737 PMCID: PMC9499917 DOI: 10.1007/s10787-022-01023-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVE Tauopathy is a group of neurodegenerative diseases in which the pathogenesis processes are related to tau protein. The imbalances between the activities of kinases and phosphatases of tau protein lead to tau hyperphosphorylation and subsequent neurodegeneration. Numerous studies suggest a strong linkage between type 2 diabetes mellitus (T2D) and neurodegenerative diseases. Therefore, finding a drug with a dual therapeutic activity against T2D and neuroprotective will be a promising idea. Hence, the potential neuroprotective effect of Glimepiride (GPD) against tauopathy was evaluated in the current study. METHODS P301S mice model was employed for tauopathy and C57BL/6 wild type mice (WT) was used as control. Phosphorylated and acetylated tau protein levels was assessed in cortex and hippocampus by western blot. Effect of GPD on tauopathy related enzymes, neuroinflammation, apoptotic markers were evaluated. Furthermore, the neuroprotective effects against anxiety like behavior and motor impairment was analyzed using Parallel rod floor and Open field tests. RESULTS GPD significantly ameliorates motor impairment, anxiety like behavior and neurodegeneration in P301S mice. Phosphorylated tau and acetylated tau were significantly decreased in both cortex and hippocampus of P301S mice via decreasing GSK3β, increasing ratio of phosphorylated-AKT to total-AKT, increasing PP2A and normalization of CDK5 levels. Furthermore, GPD treatment also decreased neuroinflammation and apoptosis by reducing NF-kB, TNF-α and caspase 3 levels. CONCLUSION The current data suggests that GPD exerts a protective effect against tauopathy, behavioural consequences, neurodegeneration, neuroinflammation and apoptosis. GPD is therefore a promising agent for the treatment of neurodegenerative diseases associated with tauopathy.
Collapse
Affiliation(s)
- Mennatallah O Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - S El-Desouky
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Doaa A Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Mohamed Salama
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Institute of Global Health and Human Ecology, The American University in Cairo, Cairo, Egypt
| | - Samar S Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
32
|
Disruption of tubulin-alpha4a polyglutamylation prevents aggregation of hyper-phosphorylated tau and microglia activation in mice. Nat Commun 2022; 13:4192. [PMID: 35858909 PMCID: PMC9300677 DOI: 10.1038/s41467-022-31776-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/30/2022] [Indexed: 11/14/2022] Open
Abstract
Dissociation of hyper-phosphorylated Tau from neuronal microtubules and its pathological aggregates, are hallmarks in the etiology of tauopathies. The Tau-microtubule interface is subject to polyglutamylation, a reversible posttranslational modification, increasing negative charge at tubulin C-terminal tails. Here, we asked whether tubulin polyglutamylation may contribute to Tau pathology in vivo. Since polyglutamylases modify various proteins other than tubulin, we generated a knock-in mouse carrying gene mutations to abolish Tuba4a polyglutamylation in a substrate-specific manner. We found that Tuba4a lacking C-terminal polyglutamylation prevents the binding of Tau and GSK3 kinase to neuronal microtubules, thereby strongly reducing phospho-Tau levels. Notably, crossbreeding of the Tuba4a knock-in mouse with the hTau tauopathy model, expressing a human Tau transgene, reversed hyper-phosphorylation and oligomerization of Tau and normalized microglia activation in brain. Our data highlight tubulin polyglutamylation as a potential therapeutic strategy in fighting tauopathies. Pathologic oligomerization of hyper-phosphorylated Tau is a hallmark of tauopathies. Here the authors show that the loss of tubulin a4 polyglutamylation reverses tau hyperphosphorylation, oligomerization and microglia activation in a tauopathy mouse.
Collapse
|
33
|
Sanchez-Varo R, Mejias-Ortega M, Fernandez-Valenzuela JJ, Nuñez-Diaz C, Caceres-Palomo L, Vegas-Gomez L, Sanchez-Mejias E, Trujillo-Estrada L, Garcia-Leon JA, Moreno-Gonzalez I, Vizuete M, Vitorica J, Baglietto-Vargas D, Gutierrez A. Transgenic Mouse Models of Alzheimer's Disease: An Integrative Analysis. Int J Mol Sci 2022; 23:5404. [PMID: 35628216 PMCID: PMC9142061 DOI: 10.3390/ijms23105404] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/10/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) constitutes the most prominent form of dementia among elderly individuals worldwide. Disease modeling using murine transgenic mice was first initiated thanks to the discovery of heritable mutations in amyloid precursor protein (APP) and presenilins (PS) genes. However, due to the repeated failure of translational applications from animal models to human patients, along with the recent advances in genetic susceptibility and our current understanding on disease biology, these models have evolved over time in an attempt to better reproduce the complexity of this devastating disease and improve their applicability. In this review, we provide a comprehensive overview about the major pathological elements of human AD (plaques, tauopathy, synaptic damage, neuronal death, neuroinflammation and glial dysfunction), discussing the knowledge that available mouse models have provided about the mechanisms underlying human disease. Moreover, we highlight the pros and cons of current models, and the revolution offered by the concomitant use of transgenic mice and omics technologies that may lead to a more rapid improvement of the present modeling battery.
Collapse
Affiliation(s)
- Raquel Sanchez-Varo
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Departamento Fisiologia Humana, Histologia Humana, Anatomia Patologica y Educacion Fisica y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071 Malaga, Spain
| | - Marina Mejias-Ortega
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Juan Jose Fernandez-Valenzuela
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Cristina Nuñez-Diaz
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Laura Caceres-Palomo
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Laura Vegas-Gomez
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Elisabeth Sanchez-Mejias
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Laura Trujillo-Estrada
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Juan Antonio Garcia-Leon
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Ines Moreno-Gonzalez
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Marisa Vizuete
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Departamento Bioquimica y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, Instituto de Biomedicina de Sevilla (IBIS)-Hospital Universitario Virgen del Rocio/CSIC, 41012 Seville, Spain
| | - Javier Vitorica
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
- Departamento Bioquimica y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, Instituto de Biomedicina de Sevilla (IBIS)-Hospital Universitario Virgen del Rocio/CSIC, 41012 Seville, Spain
| | - David Baglietto-Vargas
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| | - Antonia Gutierrez
- Departamento Biologia Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, 29071 Malaga, Spain; (R.S.-V.); (M.M.-O.); (J.J.F.-V.); (C.N.-D.); (L.C.-P.); (L.V.-G.); (E.S.-M.); (L.T.-E.); (J.A.G.-L.); (I.M.-G.)
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain; (M.V.); (J.V.)
| |
Collapse
|
34
|
Permanne B, Sand A, Ousson S, Nény M, Hantson J, Schubert R, Wiessner C, Quattropani A, Beher D. O-GlcNAcase Inhibitor ASN90 is a Multimodal Drug Candidate for Tau and α-Synuclein Proteinopathies. ACS Chem Neurosci 2022; 13:1296-1314. [PMID: 35357812 PMCID: PMC9026285 DOI: 10.1021/acschemneuro.2c00057] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Neurodegenerative proteinopathies are characterized by the intracellular formation of insoluble and toxic protein aggregates in the brain that are closely linked to disease progression. In Alzheimer's disease and in rare tauopathies, aggregation of the microtubule-associated tau protein leads to the formation of neurofibrillary tangles (NFT). In Parkinson's disease (PD) and other α-synucleinopathies, intracellular Lewy bodies containing aggregates of α-synuclein constitute the pathological hallmark. Inhibition of the glycoside hydrolase O-GlcNAcase (OGA) prevents the removal of O-linked N-acetyl-d-glucosamine (O-GlcNAc) moieties from intracellular proteins and has emerged as an attractive therapeutic approach to prevent the formation of tau pathology. Like tau, α-synuclein is known to be modified with O-GlcNAc moieties and in vitro these have been shown to prevent its aggregation and toxicity. Here, we report the preclinical discovery and development of a novel small molecule OGA inhibitor, ASN90. Consistent with the substantial exposure of the drug and demonstrating target engagement in the brain, the clinical OGA inhibitor ASN90 promoted the O-GlcNAcylation of tau and α-synuclein in brains of transgenic mice after daily oral dosing. Across human tauopathy mouse models, oral administration of ASN90 prevented the development of tau pathology (NFT formation), functional deficits in motor behavior and breathing, and increased survival. In addition, ASN90 slowed the progression of motor impairment and reduced astrogliosis in a frequently utilized α-synuclein-dependent preclinical rodent model of PD. These findings provide a strong rationale for the development of OGA inhibitors as disease-modifying agents in both tauopathies and α-synucleinopathies. Since tau and α-synuclein pathologies frequently co-exist in neurodegenerative diseases, OGA inhibitors represent unique, multimodal drug candidates for further clinical development.
Collapse
Affiliation(s)
- Bruno Permanne
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Astrid Sand
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Solenne Ousson
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Maud Nény
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Jennifer Hantson
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Ryan Schubert
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Christoph Wiessner
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Anna Quattropani
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| | - Dirk Beher
- Asceneuron S.A., EPFL Innovation Park, Bâtiment B, CH-1015 Lausanne, Switzerland
| |
Collapse
|
35
|
Chronic Intermittent Hypoxia Enhances Pathological Tau Seeding, Propagation, and Accumulation and Exacerbates Alzheimer-like Memory and Synaptic Plasticity Deficits and Molecular Signatures. Biol Psychiatry 2022; 91:346-358. [PMID: 34130857 PMCID: PMC8895475 DOI: 10.1016/j.biopsych.2021.02.973] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 02/19/2021] [Accepted: 02/28/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Obstructive sleep apnea, characterized by sleep fragmentation and chronic intermittent hypoxia (CIH), is a risk factor for Alzheimer's disease (AD) progression. Recent epidemiological studies point to CIH as the best predictor of developing cognitive decline and AD in older adults with obstructive sleep apnea. However, the precise underlying mechanisms remain unknown. This study was undertaken to evaluate the effect of CIH on pathological human tau seeding, propagation, and accumulation; cognition; synaptic plasticity; neuronal network excitability; and gene expression profiles in a P301S human mutant tau mouse model of AD and related tauopathies. METHODS We exposed 4- to 4.5-month-old male P301S and wild-type mice to an 8-week CIH protocol (6-min cycle: 21% O2 to 8% O2 to 21% O2, 80 cycles per 8 hours during daytime) and assessed its effect on tau pathology and various AD-related phenotypic and molecular signatures. Age- and sex-matched P301S and wild-type mice were reared in normoxia (21% O2) as experimental controls. RESULTS CIH significantly enhanced pathological human tau seeding and spread across connected brain circuitry in P301S mice; it also increased phosphorylated tau load. CIH also exacerbated memory and synaptic plasticity deficits in P301S mice. However, CIH had no effect on seizure susceptibility and network hyperexcitability in these mice. Finally, CIH exacerbated AD-related pathogenic molecular signaling in P301S mice. CONCLUSIONS CIH-induced increase in pathologic human tau seeding and spread and exacerbation of other AD-related impairments provide new insights into the role of CIH and obstructive sleep apnea in AD pathogenesis.
Collapse
|
36
|
Perea JR, García E, Vallés-Saiz L, Cuadros R, Hernández F, Bolós M, Avila J. p38 activation occurs mainly in microglia in the P301S Tauopathy mouse model. Sci Rep 2022; 12:2130. [PMID: 35136118 PMCID: PMC8826411 DOI: 10.1038/s41598-022-05980-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 01/07/2022] [Indexed: 12/26/2022] Open
Abstract
Tauopathies are a group of neurodegenerative diseases characterized by the accumulation of hyperphosphorylated tau protein in the brain. Many of these pathologies also present an inflammatory component determined by the activation of microglia, the resident immune cells of the brain. p38 MAPK is one of the molecular pathways involved in neuroinflammation. Although this kinase is expressed mainly in glia, its activation in certain neurodegenerative diseases such as Alzheimer's Disease has been associated with its ability to phosphorylate tau in neurons. Using the P301S Tauopathy mouse model, here we show that p38 activation increases during aging and that this occurs mainly in microglia of the hippocampus rather than in neurons. Furthermore, we have observed that these mice present an activated microglial variant called rod microglia. Interestingly, p38 activation in this subpopulation of microglia is decreased. On the basis of our findings, we propose that rod microglia might have a neuroprotective phenotype in the context of tau pathology.
Collapse
Affiliation(s)
- Juan R Perea
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (UAM-CSIC) (Campus de Cantoblanco), 1 Nicolás Cabrera st, 28049, Madrid, Spain.,Center for Networked Biomedical Research On Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain
| | - Esther García
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (UAM-CSIC) (Campus de Cantoblanco), 1 Nicolás Cabrera st, 28049, Madrid, Spain
| | - Laura Vallés-Saiz
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (UAM-CSIC) (Campus de Cantoblanco), 1 Nicolás Cabrera st, 28049, Madrid, Spain
| | - Raquel Cuadros
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (UAM-CSIC) (Campus de Cantoblanco), 1 Nicolás Cabrera st, 28049, Madrid, Spain
| | - Félix Hernández
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (UAM-CSIC) (Campus de Cantoblanco), 1 Nicolás Cabrera st, 28049, Madrid, Spain.,Center for Networked Biomedical Research On Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain.,Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Marta Bolós
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (UAM-CSIC) (Campus de Cantoblanco), 1 Nicolás Cabrera st, 28049, Madrid, Spain.,Center for Networked Biomedical Research On Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain
| | - Jesús Avila
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid (UAM-CSIC) (Campus de Cantoblanco), 1 Nicolás Cabrera st, 28049, Madrid, Spain. .,Center for Networked Biomedical Research On Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain.
| |
Collapse
|
37
|
Rao YL, Ganaraja B, Murlimanju BV, Joy T, Krishnamurthy A, Agrawal A. Hippocampus and its involvement in Alzheimer's disease: a review. 3 Biotech 2022; 12:55. [PMID: 35116217 PMCID: PMC8807768 DOI: 10.1007/s13205-022-03123-4] [Citation(s) in RCA: 218] [Impact Index Per Article: 72.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/16/2022] [Indexed: 12/12/2022] Open
Abstract
Hippocampus is the significant component of the limbic lobe, which is further subdivided into the dentate gyrus and parts of Cornu Ammonis. It is the crucial region for learning and memory; its sub-regions aid in the generation of episodic memory. However, the hippocampus is one of the brain areas affected by Alzheimer's (AD). In the early stages of AD, the hippocampus shows rapid loss of its tissue, which is associated with the functional disconnection with other parts of the brain. In the progression of AD, atrophy of medial temporal and hippocampal regions are the structural markers in magnetic resonance imaging (MRI). Lack of sirtuin (SIRT) expression in the hippocampal neurons will impair cognitive function, including recent memory and spatial learning. Proliferation, differentiation, and migrations are the steps involved in adult neurogenesis. The microglia in the hippocampal region are more immunologically active than the other regions of the brain. Intrinsic factors like hormones, glia, and vascular nourishment are instrumental in the neural stem cell (NSC) functions by maintaining the brain's microenvironment. Along with the intrinsic factors, many extrinsic factors like dietary intake and physical activity may also influence the NSCs. Hence, pro-neurogenic lifestyle could delay neurodegeneration.
Collapse
Affiliation(s)
- Y. Lakshmisha Rao
- Department of Anatomy, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - B. Ganaraja
- Department of Physiology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - B. V. Murlimanju
- Department of Anatomy, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, Karnataka India
| | - Teresa Joy
- Department of Anatomy, College of Medicine, American University of Antigua, Coolidge, Antigua, Antigua and Barbuda
| | - Ashwin Krishnamurthy
- Department of Anatomy, K.S. Hegde Medical Academy, Deralakatte, Nitte University, Mangalore, Karnataka India
| | - Amit Agrawal
- Department of Neurosurgery, All India Institute of Medical Sciences, Saket Nagar, Bhopal, 462020 Madhya Pradesh India
| |
Collapse
|
38
|
Ganz T, Fainstein N, Elad A, Lachish M, Goldfarb S, Einstein O, Ben-Hur T. Microbial pathogens induce neurodegeneration in Alzheimer's disease mice: protection by microglial regulation. J Neuroinflammation 2022; 19:5. [PMID: 34991645 PMCID: PMC8740456 DOI: 10.1186/s12974-021-02369-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neurodegeneration is considered the consequence of misfolded proteins' deposition. Little is known about external environmental effects on the neurodegenerative process. Infectious agent-derived pathogen-associated molecular patterns (PAMPs) activate microglia, key players in neurodegenerative diseases. We hypothesized that systemic microbial pathogens may accelerate neurodegeneration in Alzheimer's disease (AD) and that microglia play a central role in this process. METHODS We examined the effect of an infectious environment and of microbial Toll-like receptor (TLR) agonists on cortical neuronal loss and on microglial phenotype in wild type versus 5xFAD transgenic mice, carrying mutated genes associated with familial AD. RESULTS We examined the effect of a naturally bred environment on the neurodegenerative process. Earlier and accelerated cortical neuron loss occurred in 5xFAD mice housed in a natural ("dirty") environment than in a specific-pathogen-free (SPF) environment, without increasing the burden of Amyloid deposits and microgliosis. Neuronal loss occurred in a microglia-rich cortical region but not in microglia-poor CA regions of the hippocampus. Environmental exposure had no effect on cortical neuron density in wild-type mice. To model the neurodegenerative process caused by the natural infectious environment, we injected systemically the bacterial endotoxin lipopolysaccharide (LPS), a TLR4 agonist PAMP. LPS caused cortical neuronal death in 5xFAD, but not wt mice. We used the selective retinoic acid receptor α agonist Am580 to regulate microglial activation. In primary microglia isolated from 5xFAD mice, Am580 markedly attenuated TLR agonists-induced iNOS expression, without canceling their basic immune response. Intracerebroventricular delivery of Am580 in 5xFAD mice reduced significantly the fraction of (neurotoxic) iNOS + microglia and increased the fraction of (neuroprotective) TREM2 + microglia. Furthermore, intracerebroventricular delivery of Am580 prevented neurodegeneration induced by microbial TLR agonists. CONCLUSIONS Exposure to systemic infections causes neurodegeneration in brain regions displaying amyloid pathology and high local microglia density. AD brains exhibit increased susceptibility to microbial PAMPs' neurotoxicity, which accelerates neuronal death. Microglial modulation protects the brain from microbial TLR agonist PAMP-induced neurodegeneration.
Collapse
Affiliation(s)
- Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amit Elad
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Smadar Goldfarb
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ofira Einstein
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
39
|
Hartnell IJ, Blum D, Nicoll JAR, Dorothee G, Boche D. Glial cells and adaptive immunity in frontotemporal dementia with tau pathology. Brain 2021; 144:724-745. [PMID: 33527991 DOI: 10.1093/brain/awaa457] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/06/2020] [Accepted: 10/17/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is involved in the aetiology of many neurodegenerative disorders including Alzheimer's disease, Parkinson's disease and motor neuron disease. Whether neuroinflammation also plays an important role in the pathophysiology of frontotemporal dementia is less well known. Frontotemporal dementia is a heterogeneous classification that covers many subtypes, with the main pathology known as frontotemporal lobar degeneration. The disease can be categorized with respect to the identity of the protein that causes the frontotemporal lobar degeneration in the brain. The most common subgroup describes diseases caused by frontotemporal lobar degeneration associated with tau aggregation, also known as primary tauopathies. Evidence suggests that neuroinflammation may play a role in primary tauopathies with genome-wide association studies finding enrichment of genetic variants associated with specific inflammation-related gene loci. These loci are related to both the innate immune system, including brain resident microglia, and the adaptive immune system through possible peripheral T-cell involvement. This review discusses the genetic evidence and relates it to findings in animal models expressing pathogenic tau as well as to post-mortem and PET studies in human disease. Across experimental paradigms, there seems to be a consensus regarding the involvement of innate immunity in primary tauopathies, with increased microglia and astrocyte density and/or activation, as well as increases in pro-inflammatory markers. Whilst it is less clear as to whether inflammation precedes tau aggregation or vice versa; there is strong evidence to support a microglial contribution to the propagation of hyperphosphorylated in tau frontotemporal lobar degeneration associated with tau aggregation. Experimental evidence-albeit limited-also corroborates genetic data pointing to the involvement of cellular adaptive immunity in primary tauopathies. However, it is still unclear whether brain recruitment of peripheral immune cells is an aberrant result of pathological changes or a physiological aspect of the neuroinflammatory response to the tau pathology.
Collapse
Affiliation(s)
- Iain J Hartnell
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - David Blum
- University of Lille, Inserm, CHU-Lille, UMR-S 1172-Lille Neuroscience and Cognition, Lille, France.,Alzheimer & Tauopathies, LabEx DISTALZ, France
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Guillaume Dorothee
- Inserm, Sorbonne University, UMRS 938 Saint-Antoine Research Center, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, Paris, France
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
40
|
Emre C, Do KV, Jun B, Hjorth E, Alcalde SG, Kautzmann MAI, Gordon WC, Nilsson P, Bazan NG, Schultzberg M. Age-related changes in brain phospholipids and bioactive lipids in the APP knock-in mouse model of Alzheimer's disease. Acta Neuropathol Commun 2021; 9:116. [PMID: 34187579 PMCID: PMC8244172 DOI: 10.1186/s40478-021-01216-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
Sustained brain chronic inflammation in Alzheimer’s disease (AD) includes glial cell activation, an increase in cytokines and chemokines, and lipid mediators (LMs), concomitant with decreased pro-homeostatic mediators. The inflammatory response at the onset of pathology engages activation of pro-resolving, pro-homeostatic LMs followed by a gradual decrease. We used an APP knock-in (App KI) AD mouse that accumulates β-amyloid (Aβ) and presents cognitive deficits (at 2 and 6 months of age, respectively) to investigate LMs, their precursors, biosynthetic enzymes and receptors, glial activation, and inflammatory proteins in the cerebral cortex and hippocampus at 2-, 4-, 8- and 18-month-old in comparison with wild-type (WT) mice. We used LC-mass-spectrometry and MALDI molecular imaging to analyze LMs and phospholipids, and immunochemistry for proteins. Our results revealed an age-specific lipid and cytokine profile, and glial activation in the App KI mice. Despite an early onset of Aβ pathology, pro-inflammatory and pro-resolving LMs were prominently increased only in the oldest age group. Furthermore, the LM biosynthetic enzymes increased, and their receptor expression decreased in the aged App KI mice. Arachidonic acid (AA)-containing phospholipid molecular species were elevated, correlating with decreased cPLA2 activity. MALDI molecular imaging depicted differential distribution of phospholipids according to genotype in hippocampal layers. Brain histology disclosed increased microglia proliferation starting from young age in the App KI mice, while astrocyte numbers were enhanced in older ages. Our results demonstrate that the brain lipidome is modified preferentially during aging as compared to amyloid pathology in the model studied here. However, alterations in phospholipids signal early pathological changes in membrane composition.
Collapse
|
41
|
Bandyopadhyay S. Role of Neuron and Glia in Alzheimer's Disease and Associated Vascular Dysfunction. Front Aging Neurosci 2021; 13:653334. [PMID: 34211387 PMCID: PMC8239194 DOI: 10.3389/fnagi.2021.653334] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
Amyloidogenicity and vascular dysfunction are the key players in the pathogenesis of Alzheimer’s disease (AD), involving dysregulated cellular interactions. An intricate balance between neurons, astrocytes, microglia, oligodendrocytes and vascular cells sustains the normal neuronal circuits. Conversely, cerebrovascular diseases overlap neuropathologically with AD, and glial dyshomeostasis promotes AD-associated neurodegenerative cascade. While pathological hallmarks of AD primarily include amyloid-β (Aβ) plaques and neurofibrillary tangles, microvascular disorders, altered cerebral blood flow (CBF), and blood-brain barrier (BBB) permeability induce neuronal loss and synaptic atrophy. Accordingly, microglia-mediated inflammation and astrogliosis disrupt the homeostasis of the neuro-vascular unit and stimulate infiltration of circulating leukocytes into the brain. Large-scale genetic and epidemiological studies demonstrate a critical role of cellular crosstalk for altered immune response, metabolism, and vasculature in AD. The glia associated genetic risk factors include APOE, TREM2, CD33, PGRN, CR1, and NLRP3, which correlate with the deposition and altered phagocytosis of Aβ. Moreover, aging-dependent downregulation of astrocyte and microglial Aβ-degrading enzymes limits the neurotrophic and neurogenic role of glial cells and inhibits lysosomal degradation and clearance of Aβ. Microglial cells secrete IGF-1, and neurons show a reduced responsiveness to the neurotrophic IGF-1R/IRS-2/PI3K signaling pathway, generating amyloidogenic and vascular dyshomeostasis in AD. Glial signals connect to neural stem cells, and a shift in glial phenotype over the AD trajectory even affects adult neurogenesis and the neurovascular niche. Overall, the current review informs about the interaction of neuronal and glial cell types in AD pathogenesis and its critical association with cerebrovascular dysfunction.
Collapse
Affiliation(s)
- Sanghamitra Bandyopadhyay
- Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
42
|
Casali BT, Reed-Geaghan EG. Microglial Function and Regulation during Development, Homeostasis and Alzheimer's Disease. Cells 2021; 10:957. [PMID: 33924200 PMCID: PMC8074610 DOI: 10.3390/cells10040957] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/22/2022] Open
Abstract
Microglia are the resident immune cells of the brain, deriving from yolk sac progenitors that populate the brain parenchyma during development. During development and homeostasis, microglia play critical roles in synaptogenesis and synaptic plasticity, in addition to their primary role as immune sentinels. In aging and neurodegenerative diseases generally, and Alzheimer's disease (AD) specifically, microglial function is altered in ways that significantly diverge from their homeostatic state, inducing a more detrimental inflammatory environment. In this review, we discuss the receptors, signaling, regulation and gene expression patterns of microglia that mediate their phenotype and function contributing to the inflammatory milieu of the AD brain, as well as strategies that target microglia to ameliorate the onset, progression and symptoms of AD.
Collapse
Affiliation(s)
| | - Erin G. Reed-Geaghan
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA;
| |
Collapse
|
43
|
Wang J, Hu HJ, Liu ZK, Liu JJ, Wang SS, Cheng Q, Chen HZ, Song M. Pharmacological inhibition of asparaginyl endopeptidase by δ-secretase inhibitor 11 mitigates Alzheimer's disease-related pathologies in a senescence-accelerated mouse model. Transl Neurodegener 2021; 10:12. [PMID: 33789744 PMCID: PMC8015189 DOI: 10.1186/s40035-021-00235-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Currently, there is no cure for Alzheimer's disease (AD). Therapeutics that can modify the early stage of AD are urgently needed. Recent studies have shown that the pathogenesis of AD is closely regulated by an endo/lysosomal asparaginyl endopeptidase (AEP). Inhibition of AEP has been reported to prevent neural degeneration in transgenic mouse models of AD. However, more than 90% of AD cases are age-related sporadic AD rather than hereditary AD. The therapeutic efficacy of AEP inhibition in ageing-associated sporadic AD remains unknown. METHODS The senescence-accelerated mouse prone 8 (SAMP8) was chosen as an approximate model of sporadic AD and treated with a selective AEP inhibitor,: δ-secretase inhibitor 11. Activation of AEP was determined by enzymatic activity assay. Concentration of soluble amyloid β (Aβ) in the brain was determined by ELISA. Morris water maze test was performed to assess the learning and memory-related cognitive ability. Pathological changes in the brain were explored by morphological and western blot analyses. RESULTS The enzymatic activity of AEP in the SAMP8 mouse brain was significantly higher than that in the age-matched SAMR1 mice. The half maximal inhibitory concentration (IC50) for δ-secretase inhibitor 11 to inhibit AEP in vitro is was around 150 nM. Chronic treatment with δ-secretase inhibitor 11 markedly decreased the brain AEP activity, reduced the generation of Aβ1-40/42 and ameliorated memory loss. The inhibition of AEP with this reagent not only reduced the AEP-cleaved tau fragments and tau hyperphosphorylation, but also attenuated neuroinflammation in the form of microglial activation. Moreover, treatment with δ-secretase inhibitor 11 prevented the synaptic loss and alleviated dendritic disruption in SAMP8 mouse brain. CONCLUSIONS Pharmacological inhibition of AEP can intervene and prevent AD-like pathological progress in the model of sporadic AD. The up-regulated AEP in the brain could be a promising target for early treatment of AD. The δ-secretase inhibitor 11 can be used as a lead compound for translational development of AD treatment.
Collapse
Affiliation(s)
- Ju Wang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hui-Jie Hu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zi-Kai Liu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing-Jing Liu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shan-Shan Wang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qing Cheng
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hong-Zhuan Chen
- Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Mingke Song
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
44
|
Yan M, Zheng T. Role of the endolysosomal pathway and exosome release in tau propagation. Neurochem Int 2021; 145:104988. [PMID: 33582164 DOI: 10.1016/j.neuint.2021.104988] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 02/08/2023]
Abstract
The progressive deposition of misfolded and aggregated forms of Tau protein in the brain is a pathological hallmark of tauopathies, such as Alzheimer's disease (AD) and frontotemporal degeneration (FTD). The misfolded Tau can be released into the extracellular space and internalized by neighboring cells, acting as seeds to trigger the robust conversion of soluble Tau into insoluble filamentous aggregates in a prion-like manner, ultimately contributing to the progression of the disease. However, molecular mechanisms accountable for the propagation of Tau pathology are poorly defined. We reviewed the Tau processing imbalance in endosomal, lysosomal, and exosomal pathways in AD. Increased exosome release counteracts the endosomal-lysosomal dysfunction of Tau processing but increases the number of aggregates and the propagation of Tau. This review summarizes our current understanding of the underlying tauopathy mechanisms with an emphasis on the emerging role of the endosomal-lysosomal-exosome pathways in this process. The components CHMP6, TSG101, and other components of the ESCRT complex, as well as Rab GTPase such as Rab35 and Rab7A, regulate vesicle cargoes routing from endosome to lysosome and affect Tau traffic, degradation, or secretion. Thus, the significant molecular pathways that should be potential therapeutic targets for treating tauopathies are determined.
Collapse
Affiliation(s)
- Minli Yan
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou, 310009, China
| | - Tingting Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou, 310009, China.
| |
Collapse
|
45
|
Gratuze M, Leyns CE, Sauerbeck AD, St-Pierre MK, Xiong M, Kim N, Serrano JR, Tremblay MÈ, Kummer TT, Colonna M, Ulrich JD, Holtzman DM. Impact of TREM2R47H variant on tau pathology-induced gliosis and neurodegeneration. J Clin Invest 2021; 130:4954-4968. [PMID: 32544086 DOI: 10.1172/jci138179] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by plaques containing amyloid-β (Aβ) and neurofibrillary tangles composed of aggregated, hyperphosphorylated tau. Beyond tau and Aβ, evidence suggests that microglia play an important role in AD pathogenesis. Rare variants in the microglia-expressed triggering receptor expressed on myeloid cells 2 (TREM2) gene increase AD risk 2- to 4-fold. It is likely that these TREM2 variants increase AD risk by decreasing the response of microglia to Aβ and its local toxicity. However, neocortical Aβ pathology occurs many years before neocortical tau pathology in AD. Thus, it will be important to understand the role of TREM2 in the context of tauopathy. We investigated the impact of the AD-associated TREM2 variant (R47H) on tau-mediated neuropathology in the PS19 mouse model of tauopathy. We assessed PS19 mice expressing human TREM2CV (common variant) or human TREM2R47H. PS19-TREM2R47H mice had significantly attenuated brain atrophy and synapse loss versus PS19-TREM2CV mice. Gene expression analyses and CD68 immunostaining revealed attenuated microglial reactivity in PS19-TREM2R47H versus PS19-TREM2CV mice. There was also a decrease in phagocytosis of postsynaptic elements by microglia expressing TREM2R47H in the PS19 mice and in human AD brains. These findings suggest that impaired TREM2 signaling reduces microglia-mediated neurodegeneration in the setting of tauopathy.
Collapse
Affiliation(s)
- Maud Gratuze
- Department of Neurology.,Hope Center for Neurological Disorders, and.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Cheryl Eg Leyns
- Department of Neurology.,Hope Center for Neurological Disorders, and.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Marie-Kim St-Pierre
- Axe Neurosciences, Centre de Recherche, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec City, Québec, Canada.,Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Monica Xiong
- Department of Neurology.,Hope Center for Neurological Disorders, and.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nayeon Kim
- Department of Neurology.,Hope Center for Neurological Disorders, and.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Javier Remolina Serrano
- Department of Neurology.,Hope Center for Neurological Disorders, and.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche, Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec City, Québec, Canada.,Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | | | - Marco Colonna
- Hope Center for Neurological Disorders, and.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jason D Ulrich
- Department of Neurology.,Hope Center for Neurological Disorders, and.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David M Holtzman
- Department of Neurology.,Hope Center for Neurological Disorders, and.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
46
|
Shahidehpour RK, Higdon RE, Crawford NG, Neltner JH, Ighodaro ET, Patel E, Price D, Nelson PT, Bachstetter AD. Dystrophic microglia are associated with neurodegenerative disease and not healthy aging in the human brain. Neurobiol Aging 2021; 99:19-27. [PMID: 33422891 PMCID: PMC8293930 DOI: 10.1016/j.neurobiolaging.2020.12.003] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/31/2020] [Accepted: 12/02/2020] [Indexed: 01/26/2023]
Abstract
Loss of physiological microglial function may increase the propagation of neurodegenerative diseases. Cellular senescence is a hallmark of aging; thus, we hypothesized age could be a cause of dystrophic microglia. Stereological counts were performed for total microglia, 2 microglia morphologies (hypertrophic and dystrophic) across the human lifespan. An age-associated increase in the number of dystrophic microglia was found in the hippocampus and frontal cortex. However, the increase in dystrophic microglia was proportional to the age-related increase in the total number of microglia. Thus, aging alone does not explain the presence of dystrophic microglia. We next tested if dystrophic microglia could be a disease-associated microglia morphology. Compared with controls, the number of dystrophic microglia was greater in cases with either Alzheimer's disease, dementia with Lewy bodies, or limbic-predominant age-related TDP-43 encephalopathy. These results demonstrate that microglia dystrophy, and not hypertrophic microglia, are the disease-associated microglia morphology. Finally, we found strong evidence for iron homeostasis changes in dystrophic microglia, providing a possible molecular mechanism driving the degeneration of microglia in neurodegenerative disease.
Collapse
Affiliation(s)
- Ryan K Shahidehpour
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Rebecca E Higdon
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Nicole G Crawford
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA; Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Janna H Neltner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Pathology and Laboratory Medicine, Division of Neuropathology, University of Kentucky, Lexington, KY, USA
| | - Eseosa T Ighodaro
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Ela Patel
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Douglas Price
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Pathology and Laboratory Medicine, Division of Neuropathology, University of Kentucky, Lexington, KY, USA
| | - Adam D Bachstetter
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA; Department of Neuroscience, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
47
|
Morshed N, Ralvenius WT, Nott A, Watson LA, Rodriguez FH, Akay LA, Joughin BA, Pao P, Penney J, LaRocque L, Mastroeni D, Tsai L, White FM. Phosphoproteomics identifies microglial Siglec-F inflammatory response during neurodegeneration. Mol Syst Biol 2020; 16:e9819. [PMID: 33289969 PMCID: PMC7722784 DOI: 10.15252/msb.20209819] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the appearance of amyloid-β plaques, neurofibrillary tangles, and inflammation in brain regions involved in memory. Using mass spectrometry, we have quantified the phosphoproteome of the CK-p25, 5XFAD, and Tau P301S mouse models of neurodegeneration. We identified a shared response involving Siglec-F which was upregulated on a subset of reactive microglia. The human paralog Siglec-8 was also upregulated on microglia in AD. Siglec-F and Siglec-8 were upregulated following microglial activation with interferon gamma (IFNγ) in BV-2 cell line and human stem cell-derived microglia models. Siglec-F overexpression activates an endocytic and pyroptotic inflammatory response in BV-2 cells, dependent on its sialic acid substrates and immunoreceptor tyrosine-based inhibition motif (ITIM) phosphorylation sites. Related human Siglecs induced a similar response in BV-2 cells. Collectively, our results point to an important role for mouse Siglec-F and human Siglec-8 in regulating microglial activation during neurodegeneration.
Collapse
Affiliation(s)
- Nader Morshed
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
| | - William T Ralvenius
- Picower Institute for Learning and MemoryMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Brain and Cognitive SciencesMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Alexi Nott
- Picower Institute for Learning and MemoryMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Brain and Cognitive SciencesMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Brain SciencesImperial College LondonUK
- UK Dementia Research Institute at Imperial College LondonLondonUK
| | - L Ashley Watson
- Picower Institute for Learning and MemoryMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Brain and Cognitive SciencesMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Felicia H Rodriguez
- Department of Chemical and Materials EngineeringNew Mexico State UniversityLas CrucesNMUSA
| | - Leyla A Akay
- Picower Institute for Learning and MemoryMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Brain and Cognitive SciencesMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Brian A Joughin
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Ping‐Chieh Pao
- Picower Institute for Learning and MemoryMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Brain and Cognitive SciencesMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Jay Penney
- Picower Institute for Learning and MemoryMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Brain and Cognitive SciencesMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Lauren LaRocque
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Diego Mastroeni
- ASU‐Banner Neurodegenerative Disease Research CenterTempeAZUSA
| | - Li‐Huei Tsai
- Picower Institute for Learning and MemoryMassachusetts Institute of TechnologyCambridgeMAUSA
- Department of Brain and Cognitive SciencesMassachusetts Institute of TechnologyCambridgeMAUSA
- Broad Institute of MIT and HarvardCambridgeMAUSA
| | - Forest M White
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMAUSA
- Center for Precision Cancer MedicineMassachusetts Institute of TechnologyCambridgeMAUSA
| |
Collapse
|
48
|
Perea JR, Bolós M, Avila J. Microglia in Alzheimer's Disease in the Context of Tau Pathology. Biomolecules 2020; 10:biom10101439. [PMID: 33066368 PMCID: PMC7602223 DOI: 10.3390/biom10101439] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/13/2022] Open
Abstract
Microglia are the cells that comprise the innate immune system in the brain. First described more than a century ago, these cells were initially assigned a secondary role in the central nervous system (CNS) with respect to the protagonists, neurons. However, the latest advances have revealed the complexity and importance of microglia in neurodegenerative conditions such as Alzheimer’s disease (AD), the most common form of dementia associated with aging. This pathology is characterized by the accumulation of amyloid-β peptide (Aβ), which forms senile plaques in the neocortex, as well as by the aggregation of hyperphosphorylated tau protein, a process that leads to the development of neurofibrillary tangles (NFTs). Over the past few years, efforts have been focused on studying the interaction between Aβ and microglia, together with the ability of the latter to decrease the levels of this peptide. Given that most clinical trials following this strategy have failed, current endeavors focus on deciphering the molecular mechanisms that trigger the tau-induced inflammatory response of microglia. In this review, we summarize the most recent studies on the physiological and pathological functions of tau protein and microglia. In addition, we analyze the impact of microglial AD-risk genes (APOE, TREM2, and CD33) in tau pathology, and we discuss the role of extracellular soluble tau in neuroinflammation.
Collapse
Affiliation(s)
- Juan Ramón Perea
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 1 Nicolás Cabrera, 28049 Madrid, Spain; (J.R.P.); (M.B.)
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), 5 Valderrebollo, 28031 Madrid, Spain
| | - Marta Bolós
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 1 Nicolás Cabrera, 28049 Madrid, Spain; (J.R.P.); (M.B.)
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), 5 Valderrebollo, 28031 Madrid, Spain
| | - Jesús Avila
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 1 Nicolás Cabrera, 28049 Madrid, Spain; (J.R.P.); (M.B.)
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), 5 Valderrebollo, 28031 Madrid, Spain
- Correspondence: ; Tel.:+34-196-4564
| |
Collapse
|
49
|
Kim E, Otgontenger U, Jamsranjav A, Kim SS. Deleterious Alteration of Glia in the Brain of Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21186676. [PMID: 32932623 PMCID: PMC7555758 DOI: 10.3390/ijms21186676] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 02/07/2023] Open
Abstract
The deterioration of neurons in Alzheimer’s disease (AD) arises from genetic, immunologic, and cellular factors inside the cortex. The traditional consensus of the amyloid-beta (Aβ) paradigm as a singular cause of AD has been under revision, with the accumulation of exploding neurobiological evidence. Among the multifaceted casualties of AD, the involvement of glia gains significance for its dynamic contribution to neurons, either in a neuroprotective or neurotoxic fashion. Basically, microglia and astrocytes contribute to neuronal sustainability by releasing neuroprotective cytokines, maintaining an adequate amount of glutamate in the synapse, and pruning excessive synaptic terminals. Such beneficial effects divert to the other detrimental cascade in chronic neuroinflammatory conditions. In this change, there are new discoveries of specific cytokines, microRNAs, and complementary factors. Previously unknown mechanisms of ion channels such as Kv1.3, Kir2.1, and HCN are also elucidated in the activation of microglia. The activation of glia is responsible for the excitotoxicity through the overflow of glutamate transmitter via mGluRs expressed on the membrane, which can lead to synaptic malfunction and engulfment. The communication between microglia and astrocytes is mediated through exosomes as well as cytokines, where numerous pieces of genetic information are transferred in the form of microRNAs. The new findings tell us that the neuronal environment in the AD condition is a far more complicated and dynamically interacting space. The identification of each molecule in the milieu and cellular communication would contribute to a better understanding of AD in the neurobiological perspective, consequently suggesting a possible therapeutic clue.
Collapse
Affiliation(s)
| | | | | | - Sang Seong Kim
- Correspondence: ; Tel.: +82-31-400-5812; Fax: +82-31-400-5958
| |
Collapse
|
50
|
Yang T, Liu H, Tran KC, Leng A, Massa SM, Longo FM. Small-molecule modulation of the p75 neurotrophin receptor inhibits a wide range of tau molecular pathologies and their sequelae in P301S tauopathy mice. Acta Neuropathol Commun 2020; 8:156. [PMID: 32891185 PMCID: PMC7487850 DOI: 10.1186/s40478-020-01034-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/29/2020] [Indexed: 12/16/2022] Open
Abstract
In tauopathies, phosphorylation, acetylation, cleavage and other modifications of tau drive intracellular generation of diverse forms of toxic tau aggregates and associated seeding activity, which have been implicated in subsequent synaptic failure and neurodegeneration. Suppression of this wide range of pathogenic species, seeding and toxicity mechanisms, while preserving the physiological roles of tau, presents a key therapeutic goal. Identification and targeting of signaling networks that influence a broad spectrum of tau pathogenic mechanisms might prevent or reverse synaptic degeneration and modify disease outcomes. The p75 neurotrophin receptor (p75NTR) modulates such networks, including activation of multiple tau kinases, calpain and rhoA-cofilin activity. The orally bioavailable small-molecule p75NTR modulator, LM11A-31, was administered to tauP301S mice for 3 months starting at 6 months of age, when tau pathology was well established. LM11A-31 was found to reduce: excess activation of hippocampal cdk5 and JNK kinases and calpain; excess cofilin phosphorylation, tau phosphorylation, acetylation and cleavage; accumulation of multiple forms of insoluble tau aggregates and filaments; and, microglial activation. Hippocampal extracts from treated mice had substantially reduced tau seeding activity. LM11A-31 treatment also led to a reversal of pyramidal neuron dendritic spine loss, decreased loss of dendritic complexity and improvement in performance of hippocampal behaviors. These studies identify a therapeutically tractable upstream signaling module regulating a wide spectrum of basic mechanisms underlying tauopathies.
Collapse
Affiliation(s)
- Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 300 Pasteur Drive, Room H3160, Stanford, CA, 94305, USA
| | - Harry Liu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 300 Pasteur Drive, Room H3160, Stanford, CA, 94305, USA
| | - Kevin C Tran
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 300 Pasteur Drive, Room H3160, Stanford, CA, 94305, USA
| | - Albert Leng
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 300 Pasteur Drive, Room H3160, Stanford, CA, 94305, USA
| | - Stephen M Massa
- Department of Neurology, San Francisco Veterans Affairs Health Care System and University of California, San Francisco, 4150 Clement St., San Francisco, CA, 94121, USA.
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 300 Pasteur Drive, Room H3160, Stanford, CA, 94305, USA.
| |
Collapse
|