1
|
Seplovich G, Bouchi Y, de Rivero Vaccari JP, Pareja JCM, Reisner A, Blackwell L, Mechref Y, Wang KK, Tyndall JA, Tharakan B, Kobeissy F. Inflammasome links traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Neural Regen Res 2025; 20:1644-1664. [PMID: 39104096 PMCID: PMC11688549 DOI: 10.4103/nrr.nrr-d-24-00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 08/07/2024] Open
Abstract
Traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease are three distinct neurological disorders that share common pathophysiological mechanisms involving neuroinflammation. One sequela of neuroinflammation includes the pathologic hyperphosphorylation of tau protein, an endogenous microtubule-associated protein that protects the integrity of neuronal cytoskeletons. Tau hyperphosphorylation results in protein misfolding and subsequent accumulation of tau tangles forming neurotoxic aggregates. These misfolded proteins are characteristic of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease and can lead to downstream neuroinflammatory processes, including assembly and activation of the inflammasome complex. Inflammasomes refer to a family of multimeric protein units that, upon activation, release a cascade of signaling molecules resulting in caspase-induced cell death and inflammation mediated by the release of interleukin-1β cytokine. One specific inflammasome, the NOD-like receptor protein 3, has been proposed to be a key regulator of tau phosphorylation where it has been shown that prolonged NOD-like receptor protein 3 activation acts as a causal factor in pathological tau accumulation and spreading. This review begins by describing the epidemiology and pathophysiology of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Next, we highlight neuroinflammation as an overriding theme and discuss the role of the NOD-like receptor protein 3 inflammasome in the formation of tau deposits and how such tauopathic entities spread throughout the brain. We then propose a novel framework linking traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease as inflammasome-dependent pathologies that exist along a temporal continuum. Finally, we discuss potential therapeutic targets that may intercept this pathway and ultimately minimize long-term neurological decline.
Collapse
Affiliation(s)
| | - Yazan Bouchi
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jennifer C. Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew Reisner
- Department of Pediatrics, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Laura Blackwell
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Morito T, Qi M, Kamano N, Sasaguri H, Bez S, Foiani M, Duff K, Benner S, Endo T, Hama H, Kurokawa H, Miyawaki A, Mizuma H, Sahara N, Shimojo M, Higuchi M, Saido TC, Watamura N. Human MAPT knockin mouse models of frontotemporal dementia for the neurodegenerative research community. CELL REPORTS METHODS 2025; 5:101024. [PMID: 40220760 DOI: 10.1016/j.crmeth.2025.101024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Existing models of frontotemporal dementia (FTD) may not fully recapitulate the pathophysiology of the disease. To generate more pathophysiologically relevant FTD models, we engineered MAPT knockin mouse lines carrying triple mutations, among which the MAPTP301S;Int10+3;S320F line exhibited robust tau pathology starting before 6 months of age. Severe tau accumulation was predominantly observed in the thalamus, hypothalamus, and amygdala with milder involvement of the cortex and hippocampus, leading to synaptic loss, brain atrophy, and FTD-like behavioral abnormalities. Crossbreeding MAPTP301S;Int10+3;S320F mice with App knockin, AppNL-G-F, mice markedly enhanced tau pathology in the cortex and hippocampus, highlighting the interplay between β-amyloid and tau. These findings establish the mutant mice as valuable models for investigating the mechanisms underlying FTD and other tauopathies, providing a relevant platform for in vivo drug screening.
Collapse
Affiliation(s)
- Takahiro Morito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Mohan Qi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan; Dementia Pathophysiology Collaboration Unit, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Sumi Bez
- UK Dementia Research Institute, University College London, London WC1E6BT, UK
| | - Martha Foiani
- UK Dementia Research Institute, University College London, London WC1E6BT, UK
| | - Karen Duff
- UK Dementia Research Institute, University College London, London WC1E6BT, UK
| | - Seico Benner
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Ibaraki 305-8506, Japan
| | | | - Hiroshi Hama
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Hiroshi Kurokawa
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Atushi Miyawaki
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan
| | - Hiroshi Mizuma
- Advanced Neuroimaging Center, National Institutes for Quantum Science and Technology, Inage, Chiba 263-8555, Japan
| | - Naruhiko Sahara
- Advanced Neuroimaging Center, National Institutes for Quantum Science and Technology, Inage, Chiba 263-8555, Japan
| | - Masafumi Shimojo
- Advanced Neuroimaging Center, National Institutes for Quantum Science and Technology, Inage, Chiba 263-8555, Japan
| | - Makoto Higuchi
- Advanced Neuroimaging Center, National Institutes for Quantum Science and Technology, Inage, Chiba 263-8555, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| | - Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan; UK Dementia Research Institute, University College London, London WC1E6BT, UK.
| |
Collapse
|
3
|
Vanderlinden G, Radwan A, Christiaens D, Blommaert J, Sunaert S, Vandenbulcke M, Koole M, Van Laere K. Fibre density and cross-section associate with hallmark pathology in early Alzheimer's disease. Alzheimers Res Ther 2025; 17:73. [PMID: 40188035 PMCID: PMC11971806 DOI: 10.1186/s13195-025-01710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/06/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Tau pathology in Alzheimer's disease (AD) propagates trans-synaptically along structurally connected brain networks and in synergy with amyloid pathology it induces synaptic damage. However, the in vivo relationship of amyloid, tau and synaptic density with white matter (WM) structural changes has been studied rather limitedly. Recent advances in diffusion MRI processing allow quantification of apparent fibre density and fibre cross-section on the fixel level, i.e., individual fibre populations within one voxel. The aim of this study was to investigate the hypothesis of axonal loss due to tau propagation and amyloid pathology and its association with synaptic density in early disease stages. METHODS Twenty-four patients with amnestic mild cognitive impairment (aMCI) and 23 healthy controls (HC) underwent baseline amyloid (11C-PiB/18F-NAV4694), tau (18F-MK-6240) and synaptic density (11C-UCB-J binding to SV2A) PET/MR in combination with diffusion MRI and cognitive assessments. A subset of 14 aMCI patients underwent follow-up visits after 2 years. First, a whole-brain fixel-based analysis was performed to identify differences in fibre density and fibre cross-section between HC and aMCI and longitudinally in the aMCI group. Next, a tract-of-interest analysis was performed, focusing on the temporal-cingulum bundle where most alterations have been shown in early AD. Tau and SV2A PET were quantified in the connected regions, i.e., hippocampus and posterior cingulate/precuneus (PCC-P). Amyloid PET centiloids were measured in the commonly used cortical composite volume-of-interest. RESULTS At baseline, multiple WM tracts showed lower fibre density and lower fibre cross-section in aMCI compared to HC, and these parameters further decreased longitudinally in the aMCI group. In the temporal cingulum bundle, reduced fibre density was significantly associated with reduced hippocampal synaptic density while increased hippocampal and PCC-P tau specifically correlated with reduced fibre cross-section. Increased global amyloid burden was associated with reduced fibre density and fibre cross-section in the temporal cingulum bundle. CONCLUSIONS Our results suggest that WM degeneration already occurs in the aMCI stage of AD and alterations in apparent fibre density and fibre cross-section of the temporal cingulum bundle are associated with AD hallmark pathology.
Collapse
Affiliation(s)
- Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.
| | - Ahmed Radwan
- Translational MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Daan Christiaens
- Translational MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | | | - Stefan Sunaert
- Translational MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Department of Radiology, University Hospitals UZ Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Leuven Brain Institute, Leuven, Belgium
- Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
- Neuropsychiatry, Research Group Psychiatry, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
- Division of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium
| |
Collapse
|
4
|
Thal DR, Poesen K, Vandenberghe R, De Meyer S. Alzheimer's disease neuropathology and its estimation with fluid and imaging biomarkers. Mol Neurodegener 2025; 20:33. [PMID: 40087672 PMCID: PMC11907863 DOI: 10.1186/s13024-025-00819-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) is neuropathologically characterized by the extracellular deposition of the amyloid-β peptide (Aβ) and the intraneuronal accumulation of abnormal phosphorylated tau (τ)-protein (p-τ). Most frequently, these hallmark lesions are accompanied by other co-pathologies in the brain that may contribute to cognitive impairment, such as vascular lesions, intraneuronal accumulation of phosphorylated transactive-response DNA-binding protein 43 (TDP-43), and/or α-synuclein (αSyn) aggregates. To estimate the extent of these AD and co-pathologies in patients, several biomarkers have been developed. Specific tracers target and visualize Aβ plaques, p-τ and αSyn pathology or inflammation by positron emission tomography. In addition to these imaging biomarkers, cerebrospinal fluid, and blood-based biomarker assays reflecting AD-specific or non-specific processes are either already in clinical use or in development. In this review, we will introduce the pathological lesions of the AD brain, the related biomarkers, and discuss to what extent the respective biomarkers estimate the pathology determined at post-mortem histopathological analysis. It became evident that initial stages of Aβ plaque and p-τ pathology are not detected with the currently available biomarkers. Interestingly, p-τ pathology precedes Aβ deposition, especially in the beginning of the disease when biomarkers are unable to detect it. Later, Aβ takes the lead and accelerates p-τ pathology, fitting well with the known evolution of biomarker measures over time. Some co-pathologies still lack clinically established biomarkers today, such as TDP-43 pathology or cortical microinfarcts. In summary, specific biomarkers for AD-related pathologies allow accurate clinical diagnosis of AD based on pathobiological parameters. Although current biomarkers are excellent measures for the respective pathologies, they fail to detect initial stages of the disease for which post-mortem analysis of the brain is still required. Accordingly, neuropathological studies remain essential to understand disease development especially in early stages. Moreover, there is an urgent need for biomarkers reflecting co-pathologies, such as limbic predominant, age-related TDP-43 encephalopathy-related pathology, which is known to modify the disease by interacting with p-τ. Novel biomarker approaches such as extracellular vesicle-based assays and cryptic RNA/peptides may help to better detect these co-pathologies in the future.
Collapse
Affiliation(s)
- Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium.
| | - Koen Poesen
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Steffi De Meyer
- Department of Neurosciences, Laboratory for Molecular Neurobiomarker Research, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Yi D, Byun MS, Jung JH, Jung G, Ahn H, Chang YY, Keum M, Lee J, Lee Y, Kim YK, Kang KM, Sohn C, Risacher SL, Saykin AJ, Lee DY. Locus coeruleus tau is linked to successive cortical tau accumulation. Alzheimers Dement 2025; 21:e14426. [PMID: 39641328 PMCID: PMC11848382 DOI: 10.1002/alz.14426] [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: 08/30/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION We investigated the hypothesis that tau burden in the locus coeruleus (LC) correlates with tau accumulation in cortical regions according to the Braak stages and examined whether the relationships differed according to cortical amyloid beta (Aβ) deposition. METHODS One hundred and seventy well-characterized participants from an ongoing cohort were included. High-resolution T1, tau positron emission tomography (PET), and amyloid PET were obtained. RESULTS LC tau burden was significantly linked to global tau in neocortical regions, as was tau in both early Braak stage (stage I/II) and later Braak stage areas. This relationship was significant only in Aβ-positive individuals. While LC tau did not directly impact memory, it was indirectly associated with delayed memory through mediation or moderation pathways. DISCUSSION The findings from living human brains support the idea that LC tau closely relates to subsequent cortical tau accumulation, particularly among individuals with pathological Aβ accumulation, and identify LC tau burden as a promising indicator of cognitive trajectories of AD. HIGHLIGHTS Tau burden in the LC was significantly associated with cortical tau accumulation. Tau burden in SN or PPN showed no association with cortical tau accumulation. LC tau burden was serially associated with Braak stages. The tau-LC and cortical tau relationship was significant only in the Aβ-positive group. Cortical amyloid's impact on memory worsens with higher tau accumulation in the LC.
Collapse
Affiliation(s)
- Dahyun Yi
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSeoulRepublic of Korea
| | - Min Soo Byun
- Department of NeuropsychiatrySeoul National University HospitalSeoulSeoulRepublic of Korea
- Department of PsychiatrySeoul National University College of MedicineSeoulSeoulRepublic of Korea
| | - Joon Hyung Jung
- Department of PsychiatryChungbuk National University HospitalCheongju‐siRepublic of Korea
| | - Gijung Jung
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSeoulRepublic of Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive ScienceSeoul National University College of HumanitiesSeoulSeoulRepublic of Korea
| | - Yoon Young Chang
- Inje University Sanggye Paik HospitalSeoulSeoulRepublic of Korea
| | - Musung Keum
- Department of NeuropsychiatrySoonchunhyang University HospitalBucheon‐siGyeonggi‐doRepublic of Korea
| | - Jun‐Young Lee
- Department of PsychiatrySeoul National University Boramae Medical CenterSeoulSeoulRepublic of Korea
| | - Yun‐Sang Lee
- Department of Nuclear MedicineSeoul National University College of MedicineSeoulSeoulRepublic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear MedicineSeoul National University Boramae Medical CenterSeoulSeoulRepublic of Korea
| | - Koung Mi Kang
- Department of RadiologySeoul National University HospitalSeoulSeoulRepublic of Korea
- Department of RadiologySeoul National University College of MedicineSeoulSeoulRepublic of Korea
| | - Chul‐Ho Sohn
- Department of RadiologySeoul National University HospitalSeoulSeoulRepublic of Korea
- Department of RadiologySeoul National University College of MedicineSeoulSeoulRepublic of Korea
| | - Shannon L. Risacher
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Andrew J. Saykin
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Network Science InstituteIndiana UniversityBloomingtonIndianaUSA
| | - Dong Young Lee
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSeoulRepublic of Korea
- Department of NeuropsychiatrySeoul National University HospitalSeoulSeoulRepublic of Korea
- Department of PsychiatrySeoul National University College of MedicineSeoulSeoulRepublic of Korea
| | | |
Collapse
|
6
|
Balducci C, Orsini F, Cerovic M, Beeg M, Rocutto B, Dacomo L, Masone A, Busani E, Raimondi I, Lavigna G, Chen PT, Leva S, Colombo L, Zucchelli C, Musco G, Kanaan NM, Gobbi M, Chiesa R, Fioriti L, Forloni G. Tau oligomers impair memory and synaptic plasticity through the cellular prion protein. Acta Neuropathol Commun 2025; 13:17. [PMID: 39871396 PMCID: PMC11773831 DOI: 10.1186/s40478-025-01930-3] [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: 10/23/2024] [Accepted: 01/13/2025] [Indexed: 01/29/2025] Open
Abstract
Deposition of abnormally phosphorylated tau aggregates is a central event leading to neuronal dysfunction and death in Alzheimer's disease (AD) and other tauopathies. Among tau aggregates, oligomers (TauOs) are considered the most toxic. AD brains show significant increase in TauOs compared to healthy controls, their concentration correlating with the severity of cognitive deficits and disease progression. In vitro and in vivo neuronal TauO exposure leads to synaptic and cognitive dysfunction, but their mechanisms of action are unclear. Evidence suggests that the cellular prion protein (PrPC) may act as a mediator of TauO neurotoxicity, as previously proposed for β-amyloid and α-synuclein oligomers. To investigate whether PrPC mediates TauO detrimental activities, we compared their effects on memory and synaptic plasticity in wild type (WT) and PrPC knockout (Prnp0/0) mice. Intracerebroventricular injection of TauOs significantly impaired recognition memory in WT but not in Prnp0/0 mice. Similarly, TauOs inhibited long-term potentiation in acute hippocampal slices from WT but not Prnp0/0 mice. Surface plasmon resonance indicated a high-affinity binding between TauOs and PrPC with a KD of 20-50 nM. Immunofluorescence analysis of naïve and PrPC-overexpressing HEK293 cells exposed to TauOs showed a PrPC dose-dependent association of TauOs with cells over time, and their co-localization with PrPC on the plasma membrane and in intracellular compartments, suggesting PrPC-may play a role in TauO internalization. These findings support the concept that PrPC mediates the detrimental activities of TauOs through a direct interaction, suggesting that targeting this interaction might be a promising therapeutic strategy for AD and other tauopathies.
Collapse
Affiliation(s)
- Claudia Balducci
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Franca Orsini
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Milica Cerovic
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Marten Beeg
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Beatrice Rocutto
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Letizia Dacomo
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Antonio Masone
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Eleonora Busani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Ilaria Raimondi
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Giada Lavigna
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Po-Tao Chen
- Department of Neuroscience, Zuckerman Institute, Columbia University, New York, NY, 10027, USA
| | - Susanna Leva
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Laura Colombo
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Chiara Zucchelli
- Biomolecular NMR Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Giovanna Musco
- Biomolecular NMR Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Nicholas M Kanaan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Marco Gobbi
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Roberto Chiesa
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy.
| | - Luana Fioriti
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy.
- Department of Neuroscience, Zuckerman Institute, Columbia University, New York, NY, 10027, USA.
| | - Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy.
| |
Collapse
|
7
|
Walkiewicz G, Ronisz A, Ospitalieri S, Tsaka G, Tomé SO, Vandenberghe R, von Arnim CAF, Rousseau F, Schymkowitz J, De Groef L, Thal DR. pTau pathology in the retina of TAU58 mice: association with ganglion cell degeneration and implications on seeding and propagation of pTau from human brain lysates. Acta Neuropathol Commun 2024; 12:194. [PMID: 39707519 DOI: 10.1186/s40478-024-01907-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 11/30/2024] [Indexed: 12/23/2024] Open
Abstract
The accumulation of abnormal phosphorylated Tau protein (pTau) in neurons of the brain is a pathological hallmark of Alzheimer's disease (AD). PTau pathology also occurs in the retina of AD cases. Accordingly, questions arise whether retinal pTau can act as a potential seed for inducing cerebral pTau pathology and whether retinal pTau pathology causes degeneration of retinal neurons. To address these questions, we (1) characterized pTau pathology in the retina of TAU58 mice, (2) determined the impact of pTau pathology on retinal ganglion cell density, and (3) used this mouse model to test whether brain lysates from AD and/or non-AD control cases induce seeding in the retina and/or propagation into the brain. TAU58 mice developed retinal pTau pathology at 6 months of age, increasing in severity and extent with age. TAU58 mice showed reduced retinal ganglion cell density compared to wild-type mice, which declined with age and pTau pathology progression. Brain lysates from non-AD Braak neurofibrillary tangle (NFT) stage I controls increased retinal pTau pathology after subretinal injection compared to phosphate-buffered saline (PBS) but did not accelerate pTau pathology in the brain. In contrast, subretinally injected AD brain lysates accelerated pTau pathology in the retina and the contralateral superior colliculus. Subretinal injection of AD brain lysates, but not of non-AD brain, induced in this context a neuroinflammatory response in the retina and in the contralateral primary visual cortex. These results lead to the following conclusions: (1) Brain lysates from AD and non-AD sources can accelerate tauopathy within the retina. (2) The anterograde propagation of pTau pathology from the retina to the brain can be triggered by subretinal injections of AD brain lysates. (3) Such subretinal injections also provoke a neuroinflammatory response in both the retina and the visual cortex. (4) The accumulation of retinal pTau is associated with the degeneration of the involved ganglion cells, indicating that retinal tauopathy might contribute to vision impairment in the elderly and underscore the retina's potential role in spreading tau pathology to the brain.
Collapse
Affiliation(s)
- Grzegorz Walkiewicz
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| | - Alicja Ronisz
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Simona Ospitalieri
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Grigoria Tsaka
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Switch Laboratory, VIB Center for Brain & Disease Research, VIB, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Sandra O Tomé
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurology, University Hospital Leuven, Leuven, Belgium
| | - Christine A F von Arnim
- Department of Neurology, University of Ulm, Ulm, Germany
- Department of Geriatrics, University Medical Center Göttingen, Göttingen, Germany
| | - Frederic Rousseau
- Switch Laboratory, VIB Center for Brain & Disease Research, VIB, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB Center for Brain & Disease Research, VIB, Leuven, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lies De Groef
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
- Department of Pathology, UZ Leuven, Leuven, Belgium.
| |
Collapse
|
8
|
Le NT, Chu N, Joshi G, Higgins NR, Nebie O, Adelakun N, Butts M, Monteiro MJ. Prion protein pathology in Ubiquilin 2 models of ALS. Neurobiol Dis 2024; 201:106674. [PMID: 39299489 DOI: 10.1016/j.nbd.2024.106674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Mutations in UBQLN2 cause ALS and frontotemporal dementia (FTD). The pathological signature in UBQLN2 cases is deposition of highly unusual types of inclusions in the brain and spinal cord that stain positive for UBQLN2. However, what role these inclusions play in pathogenesis remains unclear. Here we show cellular prion protein (PrPC) is found in UBQLN2 inclusions in both mouse and human neuronal induced pluripotent (IPSC) models of UBQLN2 mutations, evidenced by the presence of aggregated forms of PrPC with UBQLN2 inclusions. Turnover studies indicated that the P497H UBQLN2 mutation slows PrPC protein degradation and leads to mislocalization of PrPC in the cytoplasm. Immunoprecipitation studies indicated UBQLN2 and PrPC bind together in a complex. The abnormalities in PrPC caused by UBQLN2 mutations may be relevant in disease pathogenesis.
Collapse
Affiliation(s)
- Nhat T Le
- Department of Cancer Biology and Genetics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Nam Chu
- Department of Cancer Biology and Genetics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Gunjan Joshi
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States of America; Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Nicole R Higgins
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States of America; Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Ouada Nebie
- Department of Cancer Biology and Genetics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Niyi Adelakun
- Department of Cancer Biology and Genetics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Marie Butts
- Department of Cancer Biology and Genetics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Mervyn J Monteiro
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States of America; Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
9
|
Kotarba S, Kozłowska M, Scios M, Saramowicz K, Barczuk J, Granek Z, Siwecka N, Wiese W, Golberg M, Galita G, Sychowski G, Majsterek I, Rozpędek-Kamińska W. Potential Mechanisms of Tunneling Nanotube Formation and Their Role in Pathology Spread in Alzheimer's Disease and Other Proteinopathies. Int J Mol Sci 2024; 25:10797. [PMID: 39409126 PMCID: PMC11477428 DOI: 10.3390/ijms251910797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia worldwide. The etiopathogenesis of this disease remains unknown. Currently, several hypotheses attempt to explain its cause, with the most well-studied being the cholinergic, beta-amyloid (Aβ), and Tau hypotheses. Lately, there has been increasing interest in the role of immunological factors and other proteins such as alpha-synuclein (α-syn) and transactive response DNA-binding protein of 43 kDa (TDP-43). Recent studies emphasize the role of tunneling nanotubes (TNTs) in the spread of pathological proteins within the brains of AD patients. TNTs are small membrane protrusions composed of F-actin that connect non-adjacent cells. Conditions such as pathogen infections, oxidative stress, inflammation, and misfolded protein accumulation lead to the formation of TNTs. These structures have been shown to transport pathological proteins such as Aβ, Tau, α-syn, and TDP-43 between central nervous system (CNS) cells, as confirmed by in vitro studies. Besides their role in spreading pathology, TNTs may also have protective functions. Neurons burdened with α-syn can transfer protein aggregates to glial cells and receive healthy mitochondria, thereby reducing cellular stress associated with α-syn accumulation. Current AD treatments focus on alleviating symptoms, and clinical trials with Aβ-lowering drugs have proven ineffective. Therefore, intensifying research on TNTs could bring scientists closer to a better understanding of AD and the development of effective therapies.
Collapse
Affiliation(s)
- Szymon Kotarba
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Marta Kozłowska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Małgorzata Scios
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Kamil Saramowicz
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Julia Barczuk
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Zuzanna Granek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Wojciech Wiese
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Michał Golberg
- Department of Histology and Embryology, Medical University of Lodz, 90-419 Lodz, Poland;
| | - Grzegorz Galita
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Grzegorz Sychowski
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| |
Collapse
|
10
|
Zhang S, Lu J, Jin Z, Xu H, Zhang D, Chen J, Wang J. Gut microbiota metabolites: potential therapeutic targets for Alzheimer's disease? Front Pharmacol 2024; 15:1459655. [PMID: 39355779 PMCID: PMC11442227 DOI: 10.3389/fphar.2024.1459655] [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: 07/04/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive decline in cognitive function, which significantly increases pain and social burden. However, few therapeutic interventions are effective in preventing or mitigating the progression of AD. An increasing number of recent studies support the hypothesis that the gut microbiome and its metabolites may be associated with upstream regulators of AD pathology. Methods In this review, we comprehensively explore the potential mechanisms and currently available interventions targeting the microbiome for the improvement of AD. Our discussion is structured around modern research advancements in AD, the bidirectional communication between the gut and brain, the multi-target regulatory effects of microbial metabolites on AD, and therapeutic strategies aimed at modulating gut microbiota to manage AD. Results The gut microbiota plays a crucial role in the pathogenesis of AD through continuous bidirectional communication via the microbiota-gut-brain axis. Among these, microbial metabolites such as lipids, amino acids, bile acids and neurotransmitters, especially sphingolipids and phospholipids, may serve as central components of the gut-brain axis, regulating AD-related pathogenic mechanisms including β-amyloid metabolism, Tau protein phosphorylation, and neuroinflammation. Additionally, interventions such as probiotic administration, fecal microbiota transplantation, and antibiotic use have also provided evidence supporting the association between gut microbiota and AD. At the same time, we propose an innovative strategy for treating AD: a healthy lifestyle combined with targeted probiotics and other potential therapeutic interventions, aiming to restore intestinal ecology and microbiota balance. Conclusion Despite previous efforts, the molecular mechanisms by which gut microbes act on AD have yet to be fully described. However, intestinal microorganisms may become an essential target for connecting the gut-brain axis and improving the symptoms of AD. At the same time, it requires joint exploration by multiple centers and multiple disciplines.
Collapse
Affiliation(s)
- Shanshan Zhang
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Ziqi Jin
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Hanying Xu
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Dongmei Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Jianan Chen
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jian Wang
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
11
|
Nabizadeh F. Aβ remotely and locally facilitates Alzheimer's disease tau spreading. Cereb Cortex 2024; 34:bhae386. [PMID: 39329358 DOI: 10.1093/cercor/bhae386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/11/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of amyloid-beta plaques initiated approximately 2 decades before the symptom onset followed by build-up and spreading of neurofibrillary tau aggregates. Although it has been suggested that the amyloid-beta amplifies tau spreading the observed spatial disparity called it into question. Yet, it is unclear how neocortical amyloid-beta remotely affects early pathological tau, triggering it to leave the early formation area, and how amyloid-beta facilitates tau aggregate spreading throughout cortical regions. I aimed to investigate how amyloid-beta can facilitate tau spreading through neuronal connections in the Alzheimer's disease pathological process by combining functional magnetic resonance imaging normative connectomes and longitudinal in vivo molecular imaging data. In total, the imaging data of 317 participants, including 173 amyloid-beta-negative non-demented and 144 amyloid-beta -positive non-demented participants, have entered the study from Alzheimer's Disease Neuroimaging Initiative. Furthermore, normative resting-state functional magnetic resonance imaging connectomes were used to model tau spreading through functional connections. It was observed that the amyloid-beta in regions with the highest deposition (amyloid-beta epicenter) is remotely associated with connectivity-based spreading of tau pathology. Moreover, amyloid-beta in regions that exhibit the highest tau pathology (tau epicenter) is associated with increased connectivity-based tau spreading to non-epicenter regions. The findings provide a further explanation for a long-standing question of how amyloid-beta can affect tau aggregate spreading through neuronal connections despite spatial incongruity. The results suggest that amyloid-beta pathology can remotely and locally facilitate connectivity-based spreading of tau aggregates.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Shahid Hemmat Highway, Tehran 14496-14535, Iran
| |
Collapse
|
12
|
So RWL, Amano G, Stuart E, Ebrahim Amini A, Aguzzi A, Collingridge GL, Watts JC. α-Synuclein strain propagation is independent of cellular prion protein expression in a transgenic synucleinopathy mouse model. PLoS Pathog 2024; 20:e1012517. [PMID: 39264912 PMCID: PMC11392418 DOI: 10.1371/journal.ppat.1012517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/20/2024] [Indexed: 09/14/2024] Open
Abstract
The cellular prion protein, PrPC, has been postulated to function as a receptor for α-synuclein, potentially facilitating cell-to-cell spreading and/or toxicity of α-synuclein aggregates in neurodegenerative disorders such as Parkinson's disease. Previously, we generated the "Salt (S)" and "No Salt (NS)" strains of α-synuclein aggregates that cause distinct pathological phenotypes in M83 transgenic mice overexpressing A53T-mutant human α-synuclein. To test the hypothesis that PrPC facilitates the propagation of α-synuclein aggregates, we produced M83 mice that either express or do not express PrPC. Following intracerebral inoculation with the S or NS strain, the absence of PrPC in M83 mice did not prevent disease development and had minimal influence on α-synuclein strain-specified attributes such as the extent of cerebral α-synuclein deposition, selective targeting of specific brain regions and cell types, the morphology of induced α-synuclein deposits, and the structural fingerprints of protease-resistant α-synuclein aggregates. Likewise, there were no appreciable differences in disease manifestation between PrPC-expressing and PrPC-lacking M83 mice following intraperitoneal inoculation of the S strain. Interestingly, intraperitoneal inoculation with the NS strain resulted in two distinct disease phenotypes, indicative of α-synuclein strain evolution, but this was also independent of PrPC expression. Overall, these results suggest that PrPC plays at most a minor role in the propagation, neuroinvasion, and evolution of α-synuclein strains in mice that express A53T-mutant human α-synuclein. Thus, other putative receptors or cell-to-cell propagation mechanisms may have a larger effect on the spread of α-synuclein aggregates during disease.
Collapse
Affiliation(s)
- Raphaella W L So
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Genki Amano
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Erica Stuart
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Aeen Ebrahim Amini
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Graham L Collingridge
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Song F, Kovac V, Mohammadi B, Littau JL, Scharfenberg F, Matamoros Angles A, Vanni I, Shafiq M, Orge L, Galliciotti G, Djakkani S, Linsenmeier L, Černilec M, Hartman K, Jung S, Tatzelt J, Neumann JE, Damme M, Tschirner SK, Lichtenthaler SF, Ricklefs FL, Sauvigny T, Schmitz M, Zerr I, Puig B, Tolosa E, Ferrer I, Magnus T, Rupnik MS, Sepulveda-Falla D, Matschke J, Šmid LM, Bresjanac M, Andreoletti O, Krasemann S, Foliaki ST, Nonno R, Becker-Pauly C, Monzo C, Crozet C, Haigh CL, Glatzel M, Curin Serbec V, Altmeppen HC. Cleavage site-directed antibodies reveal the prion protein in humans is shed by ADAM10 at Y226 and associates with misfolded protein deposits in neurodegenerative diseases. Acta Neuropathol 2024; 148:2. [PMID: 38980441 PMCID: PMC11233397 DOI: 10.1007/s00401-024-02763-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
Proteolytic cell surface release ('shedding') of the prion protein (PrP), a broadly expressed GPI-anchored glycoprotein, by the metalloprotease ADAM10 impacts on neurodegenerative and other diseases in animal and in vitro models. Recent studies employing the latter also suggest shed PrP (sPrP) to be a ligand in intercellular communication and critically involved in PrP-associated physiological tasks. Although expectedly an evolutionary conserved event, and while soluble forms of PrP are present in human tissues and body fluids, for the human body neither proteolytic PrP shedding and its cleavage site nor involvement of ADAM10 or the biological relevance of this process have been demonstrated thus far. In this study, cleavage site prediction and generation (plus detailed characterization) of sPrP-specific antibodies enabled us to identify PrP cleaved at tyrosin 226 as the physiological and apparently strictly ADAM10-dependent shed form in humans. Using cell lines, neural stem cells and brain organoids, we show that shedding of human PrP can be stimulated by PrP-binding ligands without targeting the protease, which may open novel therapeutic perspectives. Site-specific antibodies directed against human sPrP also detect the shed form in brains of cattle, sheep and deer, hence in all most relevant species naturally affected by fatal and transmissible prion diseases. In human and animal prion diseases, but also in patients with Alzheimer`s disease, sPrP relocalizes from a physiological diffuse tissue pattern to intimately associate with extracellular aggregated deposits of misfolded proteins characteristic for the respective pathological condition. Findings and research tools presented here will accelerate novel insight into the roles of PrP shedding (as a process) and sPrP (as a released factor) in neurodegeneration and beyond.
Collapse
Affiliation(s)
- Feizhi Song
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Valerija Kovac
- Centre for Immunology and Development, Blood Transfusion Centre of Slovenia (BTCS), Ljubljana, Slovenia
| | - Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jessica L Littau
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | | - Andreu Matamoros Angles
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ilaria Vanni
- Department of Food Safety and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Leonor Orge
- National Institute for Agricultural and Veterinary Research (INIAV), Oeiras, Portugal
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Giovanna Galliciotti
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Salma Djakkani
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Luise Linsenmeier
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Maja Černilec
- Centre for Immunology and Development, Blood Transfusion Centre of Slovenia (BTCS), Ljubljana, Slovenia
| | - Katrina Hartman
- Centre for Immunology and Development, Blood Transfusion Centre of Slovenia (BTCS), Ljubljana, Slovenia
| | - Sebastian Jung
- Department of Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Jörg Tatzelt
- Department of Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
- Cluster of Excellence RESOLV, Ruhr University Bochum, Bochum, Germany
| | - Julia E Neumann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- Center for Molecular Neurobiology Hamburg (ZMNH), UKE, Hamburg, Germany
| | - Markus Damme
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| | - Sarah K Tschirner
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University Munich, 81675, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University Munich, 81675, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Thomas Sauvigny
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Matthias Schmitz
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI), UKE, Hamburg, Germany
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, IDIBELL, Hospitalet de Llobregat, Spain
| | - Tim Magnus
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI), UKE, Hamburg, Germany
| | - Marjan S Rupnik
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Diego Sepulveda-Falla
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jakob Matschke
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Lojze M Šmid
- LNPR, Institute of Pathophysiology and Prion Laboratory, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mara Bresjanac
- LNPR, Institute of Pathophysiology and Prion Laboratory, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Olivier Andreoletti
- UMR INRAE ENVT 1225, Interactions Hôtes-Agents Pathogènes, École Nationale Vétérinaire de Toulouse, Toulouse, France
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Simote T Foliaki
- Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, MT, USA
| | - Romolo Nonno
- Department of Food Safety and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | | | - Cecile Monzo
- Institute for Regenerative Medicine and Biotherapies (IRMB), Neural Stem Cell, MSC and Neurodegenerative Diseases, INSERM, Montpellier, France
| | - Carole Crozet
- Institute for Regenerative Medicine and Biotherapies (IRMB), Neural Stem Cell, MSC and Neurodegenerative Diseases, INSERM, Montpellier, France
| | - Cathryn L Haigh
- Laboratory of Persistent Viral Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, MT, USA
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Vladka Curin Serbec
- Centre for Immunology and Development, Blood Transfusion Centre of Slovenia (BTCS), Ljubljana, Slovenia.
| | - Hermann C Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| |
Collapse
|
14
|
Tomé SO, Gawor K, Thal DR. LATE-NC in Alzheimer's disease: Molecular aspects and synergies. Brain Pathol 2024; 34:e13213. [PMID: 37793659 PMCID: PMC11189776 DOI: 10.1111/bpa.13213] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 10/06/2023] Open
Abstract
Alzheimer's disease (AD) is classically characterized by senile plaques and neurofibrillary tangles (NFTs). However, multiple copathologies can be observed in the AD brain and contribute to the development of cognitive decline. Limbic-predominant age-related TDP-43 encephalopathy neuropathological changes (LATE-NC) accumulates in the majority of AD cases and leads to more severe cognitive decline compared with AD pathology alone. In this review, we focus on the synergistic relationship between LATE-NC and tau in AD, highlighting the aggravating role of TDP-43 aggregates on tau pathogenesis and its impact on the clinical picture and therapeutic strategies. Additionally, we discuss to what extent the molecular patterns of LATE-NC in AD differ from frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP) neuropathological changes. Thus, we highlight the importance of tau and TDP-43 synergies for subtyping AD patients, which may respond differently to therapeutic interventions depending on the presence of comorbid LATE-NC.
Collapse
Grants
- 10810 Alzheimer Forschung Initiative (Germany)
- 13803 Alzheimer Forschung Initiative (Germany)
- 22-AAIIA-963171 Alzheimer's Association (USA)
- A2022019F BrightFocus Foundation (USA)
- TH-624-4-1 Deutsche Forschungsgemeinschaft (DFG, Germany)
- 4-2 Deutsche Forschungsgemeinschaft (DFG, Germany)
- 6-1 Deutsche Forschungsgemeinschaft (DFG, Germany)
- G065721N Fonds Wetenschappelijk Onderzoek (FWO, Belgium)
- G0F8516N Fonds Wetenschappelijk Onderzoek (FWO, Belgium)
- 2020/017 Stichting Alzheimer Onderzoek (SAO/FRA, Belgium)
- C3/20/057 Onderzoeksraad, KU Leuven (Belgium)
- PDMT2/21/069 Onderzoeksraad, KU Leuven (Belgium)
- IWT 135043 Vlaamse Impulsfinanciering voor Netwerken voor Dementie-onderzoek (Belgium)
- Alzheimer Forschung Initiative (Germany)
- Alzheimer's Association (USA)
- BrightFocus Foundation (USA)
- Deutsche Forschungsgemeinschaft (DFG, Germany)
- Fonds Wetenschappelijk Onderzoek (FWO, Belgium)
- Onderzoeksraad, KU Leuven (Belgium)
Collapse
Affiliation(s)
- Sandra O. Tomé
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain InstituteKU LeuvenLeuvenBelgium
| | - Klara Gawor
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain InstituteKU LeuvenLeuvenBelgium
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain InstituteKU LeuvenLeuvenBelgium
- Department of PathologyUniversity Hospitals of LeuvenLeuvenBelgium
| |
Collapse
|
15
|
Kim DK, Suh K, Park J, Lee SE, Han J, Chang S, Kim Y, Mook-Jung I. FGFR3 drives Aβ-induced tau uptake. Exp Mol Med 2024; 56:1631-1642. [PMID: 38951140 PMCID: PMC11297141 DOI: 10.1038/s12276-024-01274-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 07/03/2024] Open
Abstract
The amyloid cascade hypothesis suggests that amyloid beta (Aβ) contributes to initiating subsequent tau pathology in Alzheimer's disease (AD). However, the underlying mechanisms through which Aβ contributes to tau uptake and propagation remain poorly understood. Here, we show that preexisting amyloid pathology accelerates the uptake of extracellular tau into neurons. Using quantitative proteomic analysis of endocytic vesicles, we reveal that Aβ induces the internalization of fibroblast growth factor receptor 3 (FGFR3). Extracellular tau binds to the extracellular domain of FGFR3 and is internalized by the FGFR3 ligand, fibroblast growth factor 2 (FGF2). Aβ accelerates FGF2 secretion from neurons, thereby inducing the internalization of tau-attached FGFR3. Knockdown of FGFR3 in the hippocampus reduces tau aggregation by decreasing tau uptake and improving memory function in AD model mice. These data suggest FGFR3 in neurons as a novel tau receptor and a key mediator of Aβ-induced tau uptake in AD.
Collapse
Affiliation(s)
- Dong Kyu Kim
- Department of Biomedical Science, College of Medicine, Seoul National University, Seoul, Korea
- Convergence Research Center for Dementia, Medical Research Center, Seoul National University, Seoul, Korea
| | - Kyujin Suh
- Department of Biomedical Science, College of Medicine, Seoul National University, Seoul, Korea
- Convergence Research Center for Dementia, Medical Research Center, Seoul National University, Seoul, Korea
| | - Junho Park
- Department of Medical Science, CHA University School of Medicine, Seongnam, Republic of Korea
- Advanced Omics Center, Future Medicine Research Institute, CHA Bundang Medical Center, Seongnam, Republic of Korea
| | - Sang-Eun Lee
- Department of Biomedical Science, College of Medicine, Seoul National University, Seoul, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jihui Han
- Department of Biomedical Science, College of Medicine, Seoul National University, Seoul, Korea
- Convergence Research Center for Dementia, Medical Research Center, Seoul National University, Seoul, Korea
| | - Sunghoe Chang
- Department of Biomedical Science, College of Medicine, Seoul National University, Seoul, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Youngsoo Kim
- Department of Medical Science, CHA University School of Medicine, Seongnam, Republic of Korea
- Advanced Omics Center, Future Medicine Research Institute, CHA Bundang Medical Center, Seongnam, Republic of Korea
| | - Inhee Mook-Jung
- Department of Biomedical Science, College of Medicine, Seoul National University, Seoul, Korea.
- Convergence Research Center for Dementia, Medical Research Center, Seoul National University, Seoul, Korea.
| |
Collapse
|
16
|
Zhai J, Hao C, Wang X, Cao Y, Pan Y, Zhou M, Sun J, Li C. Design, synthesis, and evaluation of dual-target inhibitors for the treatment of Alzheimer's disease. Arch Pharm (Weinheim) 2024; 357:e2300693. [PMID: 38332316 DOI: 10.1002/ardp.202300693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 02/10/2024]
Abstract
Aβ1-42 and acetylcholinesterase (AChE) are two key therapeutic targets for Alzheimer's disease (AD). The purpose of this study is to develop a dual-target inhibitor that inhibits both of these targets by fusing the chemical structure of baicalein and donepezil. Among them, we modified the structure of baicalein to arylcoumarin, synthesized three kinds of structural compounds, and evaluated their biological activities. The results showed that compound 3b had the strongest inhibitory effect on AChE (IC50 = 0.05 ± 0.02 µM), which was better than those of donepezil and baicalein. In addition, compound 3b has a strong ability to inhibit the aggregation of Aβ1-42 and protect nerve cells, and it can also penetrate the blood-brain barrier well. Using a zebrafish behavioral analyzer test, it was found that compound 3b can alleviate the behavioral effects of AlCl3-induced zebrafish larval movement retardation, which has a certain guiding significance for simulating the movement disorders of AD patients. In summary, compound 3b is expected to become a multifunctional agent for treating and alleviating the symptoms of AD patients.
Collapse
Affiliation(s)
- Jingfang Zhai
- National Key Laboratory of Advanced Drug Delivery System, Key Lab for Rare & Uncommon Diseases of Shandong Province, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Canhua Hao
- National Key Laboratory of Advanced Drug Delivery System, Key Lab for Rare & Uncommon Diseases of Shandong Province, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaojing Wang
- National Key Laboratory of Advanced Drug Delivery System, Key Lab for Rare & Uncommon Diseases of Shandong Province, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuexing Cao
- Jinan Shangcheng Pharmatech. Co. Ltd., Jinan, China
| | - Yinbo Pan
- National Key Laboratory of Advanced Drug Delivery System, Key Lab for Rare & Uncommon Diseases of Shandong Province, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Min Zhou
- National Key Laboratory of Advanced Drug Delivery System, Key Lab for Rare & Uncommon Diseases of Shandong Province, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Sun
- National Key Laboratory of Advanced Drug Delivery System, Key Lab for Rare & Uncommon Diseases of Shandong Province, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Chuanyou Li
- Jiangsu Second Chinese Medicine Hospital & The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
17
|
Xu Y, Zhang J, Li X. Erjingwan and Alzheimer's disease: research based on network pharmacology and experimental confirmation. Front Pharmacol 2024; 15:1328334. [PMID: 38741585 PMCID: PMC11089143 DOI: 10.3389/fphar.2024.1328334] [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: 10/26/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Background Alzheimer's disease (AD), a challenging neurodegenerative condition, has emerged as a significant global public health concern. The Chinese medicine decoction Erjingwan (EJW) has shown promising efficacy in AD treatment, though its mechanism remains unclear. Objective This study aims to elucidate the mechanism by which EJW treats AD through network pharmacology analysis and in vivo experiments. Methods We identified EJW's components using the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database and determined AD-related targets from various databases. A network comprising herbs-compounds-targets was established, and EJW's core targets were ascertained through protein-protein interaction (PPI) analysis. This study assessed the cognitive abilities of APP/PS1 mice using Morris water mazes and Y mazes, in addition to analyzing blood samples for triglyceride (TG), total cholesterol (TC), low-density lipoprotein (LDL), and high-density lipoprotein (HDL) levels. Brain tissues were examined histologically with HE staining, Nissl staining, and immunohistochemistry (IHC) for amyloid β-protein (Aβ) detection. Superoxide dismutase (SOD), reactive oxygen species (ROS), Interleukin-1β (IL-1β), and Interleukin-6 (IL-6) levels in the hippocampal region were measured by ELISA. mRNA expression of apolipoprotein A-I (APOA-I), apolipoprotein B (APOB), apolipoprotein E4 (APOE4), advanced glycation end products (AGE), the receptor for AGE (RAGE), and nuclear factor kappa-B (NF-κB) was evaluated by quantitative PCR (q-PCR). Western blotting was used to detect the expression of AGE, RAGE, NF-κB, and Tau protein. Results Screening identified 57 chemical components and 222 potential targets of EJW. Ten core targets for AD treatment were identified, with enrichment analysis suggesting EJW's effects are related to lipid metabolism and AGEs/RAGE pathways. EJW enhanced learning and memory in APP/PS1 mice, protected neuronal structure in the hippocampal region, reduced Aβ deposition, and altered levels of TG, TC, LDL, IL-1β, and IL-6, and the expression of APOE4, AGEs, RAGE, NF-κB, and Tau protein, while increasing SOD, APOA-I, and APOB mRNA expression. Conclusion The study identified four core components of EJW-iosgenin, baicalein, beta-sitosterol, quercetin-and ten core targets including AKT1, IL6, VEGFA, TP53, CASP3, for treating AD. Experimental results demonstrate EJW's capacity to modulate lipid profiles, reduce pathological markers such as Aβ1-42, Tau, IL-6, IL-1β, reactive oxygen species, SOD, and enhance cognitive functions in APP/PS1 mice, potentially through inhibiting the AGEs/RAGE/NF-κB pathway.
Collapse
Affiliation(s)
- Yuya Xu
- Department of Neurology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jian Zhang
- School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xuling Li
- Department of Neurology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
18
|
Kang T, Han Z, Zhu L, Cao B. TFR1 knockdown alleviates iron overload and mitochondrial dysfunction during neural differentiation of Alzheimer's disease-derived induced pluripotent stem cells by interacting with GSK3B. Eur J Med Res 2024; 29:101. [PMID: 38321571 PMCID: PMC10845644 DOI: 10.1186/s40001-024-01677-y] [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: 10/12/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
Iron metabolism disorders are implicated in the pathogenesis of Alzheimer's disease (AD). It was previously reported that transferrin receptor (TFR1) expression was upregulated in AD mouse model. However, the precise biological functions of TFR1 in AD progression remains unclear. Herein, we observed a gradual increase in TFR1 protein expression during the differentiation of AD patient-derived induced pluripotent stem cells (AD-iPS). TFR1 knockdown inhibited the protein expression of ferritin and ferritin heavy chain 1 (FTH1), enhanced the expression of ferroportin 1 (FPN1), and decreased intracellular levels of total iron, labile iron, and reactive oxygen species (ROS). Moreover, TFR1 knockdown improved mitochondrial membrane potential (MMP), increased adenosine triphosphate (ATP) content, downregulated mitochondrial fission proteins, and upregulated mitochondrial fusion proteins. TFR1 knockdown alleviated iron overload and mitochondrial dysfunction in neural cells differentiated from AD-iPS, while TFR1 overexpression showed the opposite results. Additionally, TFR1interacted with glycogen synthase kinase 3 beta (GSK3B) and promoted GSK3B expression. GSK3B overexpression reversed the inhibitory effects of TFR1 knockdown on iron overload and mitochondrial dysfunction in AD-iPS differentiated neural cells. In conclusion, TFR1 knockdown alleviated iron overload and mitochondrial dysfunction in neural cells differentiated from AD-iPS by promoting GSK3B expression. Our findings provide a potential therapeutic target for the treatment of AD.
Collapse
Affiliation(s)
- Tao Kang
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Zheng Han
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Lijuan Zhu
- Department of Anesthesia, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Bingqing Cao
- Department of Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China.
| |
Collapse
|
19
|
Cimiotti CGV, Paganetti P, Rossi S, Soldini E, Sacco L. Correlation between Blood Monocytes and CSF Tau in Alzheimer's Disease: The Effect of Gender and Cognitive Decline. NEUROSCI 2023; 4:319-330. [PMID: 39484180 PMCID: PMC11523745 DOI: 10.3390/neurosci4040026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 11/03/2024] Open
Abstract
Neuroinflammation is one of the main mechanisms contributing to the pathogenesis of Alzheimer's disease (AD), although its key role and the immune cells involved have not yet been identified. Blood monocytes appear to play a role in the clearance of AD-related amyloid-β (Aβ) and tau protein. This retrospective study evaluated a possible correlation between blood monocytes; the concentrations of Aβ, total tau (t-Tau), and phosphorylated tau (p-Tau) in the cerebrospinal fluid (CSF); and cognitive decline assessed according to the Montreal Cognitive Assessment (MoCA). We collected data from 33 patients with AD or mild cognitive impairment (MCI) due to AD (15 men and 18 women) and found, along with a significant reduction in the concentration of blood monocytes in women (p-value = 0.083),significant correlations between the number of blood monocytes and the concentration of t-Tau in CSF (p-value = 0.045) and between blood monocytes and MoCA score (p-value = 0.037). These results confirm the role of blood monocytes in the pathogenesis of AD, provide further evidence of a gender difference in the neuroinflammatory process underlying AD, and show that blood monocyte count may reflect the cognitive impairment of AD patients.
Collapse
Affiliation(s)
- Carlotta Ginevra Valentina Cimiotti
- Neuropsychological and Speech Therapy Unit, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, CH-6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, CH-6900 Lugano, Switzerland
| | - Paolo Paganetti
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, CH-6900 Lugano, Switzerland
- Laboratory for Aging Disorders, LRT, Ente Ospedaliero Cantonale, CH-6500 Bellinzona, Switzerland
| | - Stefania Rossi
- Neuropsychological and Speech Therapy Unit, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, CH-6900 Lugano, Switzerland
| | - Emiliano Soldini
- Competence Centre for Healthcare Practices and Policies, Department of Business Economics, Health, and Social Care (DEASS), University of Applied Sciences and Arts of Southern Switzerland, CH-6928 Manno, Switzerland;
| | - Leonardo Sacco
- Neuropsychological and Speech Therapy Unit, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, CH-6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, CH-6900 Lugano, Switzerland
| |
Collapse
|
20
|
Tsering W, Hery GP, Phillips JL, Lolo K, Bathe T, Villareal JA, Ruan IY, Prokop S. Transformation of non-neuritic into neuritic plaques during AD progression drives cortical spread of tau pathology via regenerative failure. Acta Neuropathol Commun 2023; 11:190. [PMID: 38037144 PMCID: PMC10691154 DOI: 10.1186/s40478-023-01688-6] [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: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023] Open
Abstract
Extracellular amyloid-β (Aβ) plaques and intracellular aggregates of tau protein in form of neurofibrillary tangles (NFT) are pathological hallmarks of Alzheimer's disease (AD). The exact mechanism how these two protein aggregates interact in AD is still a matter of debate. Neuritic plaques (NP), a subset of Aβ plaques containing dystrophic neurites (DN), are suggested to be unique to AD and might play a role in the interaction of Aβ and tau. Quantifying NP and non-NP in postmortem brain specimens from patients with increasing severity of AD neuropathological changes (ADNC), we demonstrate that the total number of Aβ plaques and NP increase, while the number of non-NP stagnates. Furthermore, investigating the correlation between NP and NFT, we identified unexpected brain region-specific differences when comparing cases with increasingly more severe ADNC. In neocortical regions NFT counts increase in parallel with NP counts during the progression of ADNC, while this correlation is not observed in hippocampus. These data support the notion that non-NP are transformed into NP during the progression of ADNC and indicate that NP might drive cortical NFT formation. Next, using spatial transcriptomics, we analyzed the gene expression profile of the microenvironment around non-NP and NP. We identified an upregulation of neuronal systems and Ca-dependent event pathways around NP compared to non-NP. We speculate that the upregulation of these transcripts may hint at a compensatory mechanism underlying NP formation. Our studies suggest that the transformation of non-NP to NP is a key event in ADNC progression and points to regenerative failure as a potential driving force of this process.
Collapse
Affiliation(s)
- Wangchen Tsering
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- College of Medicine, Mcknight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Gabriela P Hery
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jennifer L Phillips
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Kiara Lolo
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Tim Bathe
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA
- College of Medicine, Mcknight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jonathan A Villareal
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Isabelle Y Ruan
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA.
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL, USA.
- College of Medicine, Mcknight Brain Institute, University of Florida, Gainesville, FL, USA.
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
21
|
Van Dam D, Valkenburg F, Van Kolen K, Pintelon I, Timmermans JP, De Deyn PP. Behavioral and Neuropathological Phenotyping of the Tau58/2 and Tau58/4 Transgenic Mouse Models for FTDP-17. Life (Basel) 2023; 13:2088. [PMID: 37895469 PMCID: PMC10608666 DOI: 10.3390/life13102088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The Tau58/2 and Tau58/4 mouse lines expressing 0N4R tau with a P301S mutation mimic aspects of frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). In a side-by-side comparison, we report the age-dependent development of cognitive, motor, and behavioral deficits in comparison with the spatial-temporal evolution of cellular tau pathology in both models. METHODS We applied the SHIRPA primary screen and specific neuromotor, behavioral, and cognitive paradigms. The spatiotemporal development of tau pathology was investigated immunohistochemically. Levels of sarkosyl-insoluble paired helical filaments were determined via a MesoScale Discovery biomarker assay. RESULTS Neuromotor impairments developed from age 3 months in both models. On electron microscopy, spinal cord neurofibrillary pathology was visible in mice aged 3 months; however, AT8 immunoreactivity was not yet observed in Tau58/4 mice. Behavioral abnormalities and memory deficits occurred at a later stage (>9 months) when tau pathology was fully disseminated throughout the brain. Spatiotemporally, tau pathology spread from the spinal cord via the midbrain to the frontal cortex, while the hippocampus was relatively spared, thus explaining the late onset of cognitive deficits. CONCLUSIONS Our findings indicate the face and construct validity of both Tau58 models, which may provide new, valuable insights into the pathologic effects of tau species in vivo and may consequently facilitate the development of new therapeutic targets to delay or halt neurodegenerative processes occurring in tauopathies.
Collapse
Affiliation(s)
- Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium;
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Femke Valkenburg
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium;
| | - Kristof Van Kolen
- Neuroscience Department, Janssen Research and Development, 2340 Beerse, Belgium;
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (I.P.); (J.-P.T.)
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (I.P.); (J.-P.T.)
| | - Peter Paul De Deyn
- Laboratory of Neurochemistry and Behavior, Experimental Neurobiology Unit, University of Antwerp, Wilrijk, 2610 Antwerp, Belgium;
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
22
|
Kepp KP, Robakis NK, Høilund-Carlsen PF, Sensi SL, Vissel B. The amyloid cascade hypothesis: an updated critical review. Brain 2023; 146:3969-3990. [PMID: 37183523 DOI: 10.1093/brain/awad159] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/16/2023] Open
Abstract
Results from recent clinical trials of antibodies that target amyloid-β (Aβ) for Alzheimer's disease have created excitement and have been heralded as corroboration of the amyloid cascade hypothesis. However, while Aβ may contribute to disease, genetic, clinical, imaging and biochemical data suggest a more complex aetiology. Here we review the history and weaknesses of the amyloid cascade hypothesis in view of the new evidence obtained from clinical trials of anti-amyloid antibodies. These trials indicate that the treatments have either no or uncertain clinical effect on cognition. Despite the importance of amyloid in the definition of Alzheimer's disease, we argue that the data point to Aβ playing a minor aetiological role. We also discuss data suggesting that the concerted activity of many pathogenic factors contribute to Alzheimer's disease and propose that evolving multi-factor disease models will better underpin the search for more effective strategies to treat the disease.
Collapse
Affiliation(s)
- Kasper P Kepp
- Section of Biophysical and Biomedicinal chemistry, DTU Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Nikolaos K Robakis
- Icahn School of Medicine at Mount Sinai Medical Center, New York, NY 10029, USA
| | - Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Stefano L Sensi
- Center for Advanced Studies and Technology-CAST, and Institute for Advanced Biotechnology (ITAB), University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
| | - Bryce Vissel
- St Vincent's Hospital Centre for Applied Medical Research, St Vincent's Hospital, Sydney, 2010, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, Sydney, NSW 2052, Australia
| |
Collapse
|
23
|
Flavin WP, Hosseini H, Ruberti JW, Kavehpour HP, Giza CC, Prins ML. Traumatic brain injury and the pathways to cerebral tau accumulation. Front Neurol 2023; 14:1239653. [PMID: 37638180 PMCID: PMC10450935 DOI: 10.3389/fneur.2023.1239653] [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: 06/13/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Tau is a protein that has received national mainstream recognition for its potential negative impact to the brain. This review succinctly provides information on the structure of tau and its normal physiological functions, including in hibernation and changes throughout the estrus cycle. There are many pathways involved in phosphorylating tau including diabetes, stroke, Alzheimer's disease (AD), brain injury, aging, and drug use. The common mechanisms for these processes are put into context with changes observed in mild and repetitive mild traumatic brain injury (TBI). The phosphorylation of tau is a part of the progression to pathology, but the ability for tau to aggregate and propagate is also addressed. Summarizing both the functional and dysfunctional roles of tau can help advance our understanding of this complex protein, improve our care for individuals with a history of TBI, and lead to development of therapeutic interventions to prevent or reverse tau-mediated neurodegeneration.
Collapse
Affiliation(s)
- William P. Flavin
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Helia Hosseini
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
| | - Jeffrey W. Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - H. Pirouz Kavehpour
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Mechanical and Aerospace Engineering, UCLA, Los Angeles, CA, United States
| | - Christopher C. Giza
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Mayumi L. Prins
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| |
Collapse
|
24
|
Gomez-Sequeda N, Mendivil-Perez M, Jimenez-Del-Rio M, Lopera F, Velez-Pardo C. Cholinergic-like neurons and cerebral spheroids bearing the PSEN1 p.Ile416Thr variant mirror Alzheimer's disease neuropathology. Sci Rep 2023; 13:12833. [PMID: 37553376 PMCID: PMC10409854 DOI: 10.1038/s41598-023-39630-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
Familial Alzheimer's disease (FAD) is a complex neurodegenerative disorder for which there are no therapeutics to date. Several mutations in presenilin 1 (PSEN 1), which is the catalytic component of γ-secretase complex, are causal of FAD. Recently, the p.Ile416Thr (I416T) PSEN 1 mutation has been reported in large kindred in Colombia. However, cell and molecular information from I416T mutation is scarce. Here, we demonstrate that menstrual stromal cells (MenSCs)-derived planar (2D) PSEN 1 I416T cholinergic-like cells (ChLNS) and (3D) cerebral spheroids (CSs) reproduce the typical neuropathological markers of FAD in 4 post-transdifferentiating or 11 days of transdifferentiating, respectively. The models produce intracellular aggregation of APPβ fragments (at day 4 and 11) and phosphorylated protein TAU at residue Ser202/Thr205 (at day 11) suggesting that iAPPβ fragments precede p-TAU. Mutant ChLNs and CSs displayed DJ-1 Cys106-SO3 (sulfonic acid), failure of mitochondria membrane potential (ΔΨm), and activation of transcription factor c-JUN and p53, expression of pro-apoptotic protein PUMA, and activation of executer protein caspase 3 (CASP3), all markers of cell death by apoptosis. Moreover, we found that both mutant ChLNs and CSs produced high amounts of extracellular eAβ42. The I416T ChLNs and CSs were irresponsive to acetylcholine induced Ca2+ influx compared to WT. The I416T PSEN 1 mutation might work as dominant-negative PSEN1 mutation. These findings might help to understanding the recurring failures of clinical trials of anti-eAβ42, and support the view that FAD is triggered by the accumulation of other intracellular AβPP metabolites, rather than eAβ42.
Collapse
Affiliation(s)
- Nicolas Gomez-Sequeda
- Grupo de Neurociencias de Antioquia, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratorio 412, Medellín, Colombia
| | - Miguel Mendivil-Perez
- Grupo de Neurociencias de Antioquia, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratorio 412, Medellín, Colombia
| | - Marlene Jimenez-Del-Rio
- Grupo de Neurociencias de Antioquia, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratorio 412, Medellín, Colombia
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratorio 412, Medellín, Colombia
| | - Carlos Velez-Pardo
- Grupo de Neurociencias de Antioquia, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59, Torre 1, Laboratorio 412, Medellín, Colombia.
| |
Collapse
|
25
|
Griffiths J, Grant SGN. Synapse pathology in Alzheimer's disease. Semin Cell Dev Biol 2023; 139:13-23. [PMID: 35690535 DOI: 10.1016/j.semcdb.2022.05.028] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/12/2022] [Accepted: 05/27/2022] [Indexed: 12/31/2022]
Abstract
Synapse loss and damage are central features of Alzheimer's disease (AD) and contribute to the onset and progression of its behavioural and physiological features. Here we review the literature describing synapse pathology in AD, from what we have learned from microscopy in terms of its impacts on synapse architecture, to the mechanistic role of Aβ, tau and glial cells, mitochondrial dysfunction, and the link with AD risk genes. We consider the emerging view that synapse pathology may operate at a further level, that of synapse diversity, and discuss the prospects for leveraging new synaptome mapping methods to comprehensively understand the molecular properties of vulnerable and resilient synapses. Uncovering AD impacts on brain synapse diversity should inform therapeutic approaches targeted at preserving or replenishing lost and damaged synapses and aid the interpretation of clinical imaging approaches that aim to measure synapse damage.
Collapse
Affiliation(s)
- Jessica Griffiths
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Dementia Research Institute at Imperial College, Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Seth G N Grant
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK.
| |
Collapse
|
26
|
Neurotransmitters in Prevention and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24043841. [PMID: 36835251 PMCID: PMC9966535 DOI: 10.3390/ijms24043841] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Alzheimer's disease (AD) is the most frequent cause of cognitive impairment in middle-aged and older populations. There is a lack of drugs that demonstrate significant efficacy in AD, so the study of the pathogenesis of AD is of great importance. More efficacious interventions are needed, as reflected by our population's fast aging. Synaptic plasticity is the capacity of neurons to adjust their connections, and it is strongly tied to learning and memory, cognitive function, and brain injury recovery. Changes in synaptic strength, such as long-term potentiation (LTP) or inhibition (LTD), are thought to represent the biological foundation of the early stages of learning and memory. The results of numerous studies confirm that neurotransmitters and their receptors play an important role in the regulation of synaptic plasticity. However, so far, there is no definite correlation between the function of neurotransmitters in aberrant neural oscillation and AD-related cognitive impairment. We summarized the AD process to understand the impact of neurotransmitters in the progression and pathogenesis of AD, including the current status of neurotransmitter target drugs, and the latest evidence of neurotransmitters' function and changes in the AD process.
Collapse
|
27
|
Moonen S, Koper MJ, Van Schoor E, Schaeverbeke JM, Vandenberghe R, von Arnim CAF, Tousseyn T, De Strooper B, Thal DR. Pyroptosis in Alzheimer's disease: cell type-specific activation in microglia, astrocytes and neurons. Acta Neuropathol 2023; 145:175-195. [PMID: 36481964 DOI: 10.1007/s00401-022-02528-y] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022]
Abstract
The major neuropathological hallmarks of Alzheimer's disease (AD) are amyloid β (Aβ) plaques and neurofibrillary tangles (NFT), accompanied by neuroinflammation and neuronal loss. Increasing evidence is emerging for the activation of the canonical NOD-, LRR- and pyrin domain-containing 3 (NLRP3) inflammasome in AD. However, the mechanisms leading to neuronal loss in AD and the involvement of glial cells in these processes are still not clear. The aim of this study was to investigate the contribution of pyroptosis, a pro-inflammatory mechanism of cell death downstream of the inflammasome, to neurodegeneration in AD. Immunohistochemistry and biochemical analysis of protein levels were performed on human post-mortem brain tissue. We investigated the presence of cleaved gasdermin D (GSDMD), the pyroptosis effector protein, as well as the NLRP3 inflammasome-forming proteins, in the medial temporal lobe of 23 symptomatic AD, 25 pathologically defined preclinical AD (p-preAD) and 21 non-demented control cases. Cleaved GSDMD was detected in microglia, but also in astrocytes and in few pyramidal neurons in the first sector of the cornu ammonis (CA1) of the hippocampus and the temporal cortex of Brodmann area 36. Only microglia expressed all NLRP3 inflammasome-forming proteins (i.e., ASC, NLRP3, caspase-1). Cleaved GSDMD-positive astrocytes and neurons exhibited caspase-8 and non-canonical inflammasome protein caspase-4, respectively, potentially indicating alternative pathways for GSDMD cleavage. Brains of AD patients exhibited increased numbers of cleaved GSDMD-positive cells. Cleaved GSDMD-positive microglia and astrocytes were found in close proximity to Aβ plaques, while cleaved GSDMD-positive neurons were devoid of NFTs. In CA1, NLRP3-positive microglia and cleaved GSDMD-positive neurons were associated with local neuronal loss, indicating a possible contribution of NLRP3 inflammasome and pyroptosis activation to AD-related neurodegeneration. Taken together, our results suggest cell type-specific activation of pyroptosis in AD and extend the current knowledge about the contribution of neuroinflammation to the neurodegenerative process in AD via a direct link to neuron death by pyroptosis.
Collapse
Affiliation(s)
- Sebastiaan Moonen
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), O&N IV Herestraat 49, Bus 1032, 3000, Leuven, Belgium.
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium.
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, VIB, Leuven, Belgium.
| | - Marta J Koper
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), O&N IV Herestraat 49, Bus 1032, 3000, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Evelien Van Schoor
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), O&N IV Herestraat 49, Bus 1032, 3000, Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, VIB, Leuven, Belgium
- Laboratory for Neurobiology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Jolien M Schaeverbeke
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), O&N IV Herestraat 49, Bus 1032, 3000, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
- Department of Neurology, University Hospital Leuven, Leuven, Belgium
| | - Christine A F von Arnim
- Department of Neurology, University of Ulm, Ulm, Germany
- Department of Geriatrics, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas Tousseyn
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Bart De Strooper
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, VIB, Leuven, Belgium
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), O&N IV Herestraat 49, Bus 1032, 3000, Leuven, Belgium.
- Department of Pathology, University Hospital Leuven, Leuven, Belgium.
| |
Collapse
|
28
|
Amyloid-β in Alzheimer's disease - front and centre after all? Neuronal Signal 2023; 7:NS20220086. [PMID: 36687366 PMCID: PMC9829960 DOI: 10.1042/ns20220086] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022] Open
Abstract
The amyloid hypothesis, which proposes that accumulation of the peptide amyloid-β at synapses is the key driver of Alzheimer's disease (AD) pathogenesis, has been the dominant idea in the field of Alzheimer's research for nearly 30 years. Recently, however, serious doubts about its validity have emerged, largely motivated by disappointing results from anti-amyloid therapeutics in clinical trials. As a result, much of the AD research effort has shifted to understanding the roles of a variety of other entities implicated in pathogenesis, such as microglia, astrocytes, apolipoprotein E and several others. All undoubtedly play an important role, but the nature of this has in many cases remained unclear, partly due to their pleiotropic functions. Here, we propose that all of these AD-related entities share at least one overlapping function, which is the local regulation of amyloid-β levels, and that this may be critical to their role in AD pathogenesis. We also review what is currently known of the actions of amyloid-β at the synapse in health and disease, and consider in particular how it might interact with the key AD-associated protein tau in the disease setting. There is much compelling evidence in support of the amyloid hypothesis; rather than detract from this, the implication of many disparate AD-associated cell types, molecules and processes in the regulation of amyloid-β levels may lend further support.
Collapse
|
29
|
Ghalayini J, Boulianne GL. Deciphering mechanisms of action of ACE inhibitors in neurodegeneration using Drosophila models of Alzheimer's disease. Front Neurosci 2023; 17:1166973. [PMID: 37113150 PMCID: PMC10126366 DOI: 10.3389/fnins.2023.1166973] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder for which there is no cure. Recently, several studies have reported a significant reduction in the incidence and progression of dementia among some patients receiving antihypertensive medications such as angiotensin-converting enzyme inhibitors (ACE-Is) and angiotensin receptor blockers (ARBs). Why these drugs are beneficial in some AD patients and not others is unclear although it has been shown to be independent of their role in regulating blood pressure. Given the enormous and immediate potential of ACE-Is and ARBs for AD therapeutics it is imperative that we understand how they function. Recently, studies have shown that ACE-Is and ARBs, which target the renin angiotensin system in mammals, are also effective in suppressing neuronal cell death and memory defects in Drosophila models of AD despite the fact that this pathway is not conserved in flies. This suggests that the beneficial effects of these drugs may be mediated by distinct and as yet, identified mechanisms. Here, we discuss how the short lifespan and ease of genetic manipulations available in Drosophila provide us with a unique and unparalleled opportunity to rapidly identify the targets of ACE-Is and ARBs and evaluate their therapeutic effectiveness in robust models of AD.
Collapse
Affiliation(s)
- Judy Ghalayini
- Program in Developmental and Stem Cell Biology, Peter Gilgin Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Gabrielle L. Boulianne
- Program in Developmental and Stem Cell Biology, Peter Gilgin Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- *Correspondence: Gabrielle L. Boulianne,
| |
Collapse
|
30
|
The central role of tau in Alzheimer’s disease: From neurofibrillary tangle maturation to the induction of cell death. Brain Res Bull 2022; 190:204-217. [DOI: 10.1016/j.brainresbull.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022]
|
31
|
Li X, Ospitalieri S, Robberechts T, Hofmann L, Schmid C, Rijal Upadhaya A, Koper MJ, von Arnim CAF, Kumar S, Willem M, Gnoth K, Ramakers M, Schymkowitz J, Rousseau F, Walter J, Ronisz A, Balakrishnan K, Thal DR. Seeding, maturation and propagation of amyloid β-peptide aggregates in Alzheimer’s disease. Brain 2022; 145:3558-3570. [PMID: 36270003 DOI: 10.1093/brain/awac202] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Alzheimer’s disease is neuropathologically characterized by the deposition of the amyloid β-peptide (Aβ) as amyloid plaques. Aβ plaque pathology starts in the neocortex before it propagates into further brain regions. Moreover, Aβ aggregates undergo maturation indicated by the occurrence of post-translational modifications. Here, we show that propagation of Aβ plaques is led by presumably non-modified Aβ followed by Aβ aggregate maturation. This sequence was seen neuropathologically in human brains and in amyloid precursor protein transgenic mice receiving intracerebral injections of human brain homogenates from cases varying in Aβ phase, Aβ load and Aβ maturation stage. The speed of propagation after seeding in mice was best related to the Aβ phase of the donor, the progression speed of maturation to the stage of Aβ aggregate maturation. Thus, different forms of Aβ can trigger propagation/maturation of Aβ aggregates, which may explain the lack of success when therapeutically targeting only specific forms of Aβ.
Collapse
Affiliation(s)
- Xiaohang Li
- Department of Imaging and Pathology, Laboratory of Neuropathology, Leuven Brain Institute, KU-Leuven , Leuven , Belgium
| | - Simona Ospitalieri
- Department of Imaging and Pathology, Laboratory of Neuropathology, Leuven Brain Institute, KU-Leuven , Leuven , Belgium
| | - Tessa Robberechts
- Department of Imaging and Pathology, Laboratory of Neuropathology, Leuven Brain Institute, KU-Leuven , Leuven , Belgium
| | - Linda Hofmann
- Institute of Pathology, Laboratory of Neuropathology, Ulm University , Ulm , Germany
| | - Christina Schmid
- Institute of Pathology, Laboratory of Neuropathology, Ulm University , Ulm , Germany
| | - Ajeet Rijal Upadhaya
- Institute of Pathology, Laboratory of Neuropathology, Ulm University , Ulm , Germany
| | - Marta J Koper
- Department of Imaging and Pathology, Laboratory of Neuropathology, Leuven Brain Institute, KU-Leuven , Leuven , Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU-Leuven (University of Leuven), Leuven Brain Institute , Leuven , Belgium
- Center for Brain and Disease Research, VIB , Leuven , Belgium
| | - Christine A F von Arnim
- Department of Neurology, Ulm University , Ulm , Germany
- Division of Geriatrics, University Medical Center Göttingen , Göttingen , Germany
| | - Sathish Kumar
- Department of Neurology, University of Bonn , Bonn , Germany
| | - Michael Willem
- Chair of Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-University Munich , Munich , Germany
| | - Kathrin Gnoth
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology , Halle , Germany
| | - Meine Ramakers
- Center for Brain and Disease Research, VIB , Leuven , Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU-Leuven , Leuven , Belgium
| | - Joost Schymkowitz
- Center for Brain and Disease Research, VIB , Leuven , Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU-Leuven , Leuven , Belgium
| | - Frederic Rousseau
- Center for Brain and Disease Research, VIB , Leuven , Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU-Leuven , Leuven , Belgium
| | - Jochen Walter
- Department of Neurology, University of Bonn , Bonn , Germany
| | - Alicja Ronisz
- Department of Imaging and Pathology, Laboratory of Neuropathology, Leuven Brain Institute, KU-Leuven , Leuven , Belgium
| | - Karthikeyan Balakrishnan
- Institute of Pathology, Laboratory of Neuropathology, Ulm University , Ulm , Germany
- Department of Gene Therapy, Ulm University , Ulm , Germany
| | - Dietmar Rudolf Thal
- Department of Imaging and Pathology, Laboratory of Neuropathology, Leuven Brain Institute, KU-Leuven , Leuven , Belgium
- Institute of Pathology, Laboratory of Neuropathology, Ulm University , Ulm , Germany
- Department of Pathology, UZ-Leuven , Leuven , Belgium
| |
Collapse
|
32
|
Vanderlinden G, Ceccarini J, Vande Casteele T, Michiels L, Lemmens R, Triau E, Serdons K, Tournoy J, Koole M, Vandenbulcke M, Van Laere K. Spatial decrease of synaptic density in amnestic mild cognitive impairment follows the tau build-up pattern. Mol Psychiatry 2022; 27:4244-4251. [PMID: 35794185 DOI: 10.1038/s41380-022-01672-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/03/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023]
Abstract
Next to amyloid and tau, synaptic loss is a key pathological hallmark in Alzheimer's disease, closely related to cognitive dysfunction and neurodegeneration. Tau is thought to cause synaptic loss, but this has not been experimentally verified in vivo. In a 2-year follow-up study, dual tracer PET-MR was performed in 12 amnestic MCI patients using 18F-MK-6240 for tau and 11C-UCB-J for SV2A as a proxy for synaptic density. Tau already accumulated in the neocortex at baseline with progression in Braak V/VI at follow-up. While synaptic loss was limited to limbic regions at baseline, it followed the specific tau pattern to stage IV/V regions two years later, indicating that tau spread might drive synaptic vulnerability. Moreover, synaptic density changes correlated to changes in cognitive function. This study shows for the first time in vivo that synaptic loss regionally follows tau accumulation after two years, providing a disease-modifying window of opportunity for (combined) tau-targeting therapies.
Collapse
Affiliation(s)
- Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Imaging Pathology, KU Leuven, Leuven, Belgium.
| | - Jenny Ceccarini
- Nuclear Medicine and Molecular Imaging, Imaging Pathology, KU Leuven, Leuven, Belgium
| | - Thomas Vande Casteele
- Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Laura Michiels
- Department of Neurosciences, KU Leuven, Leuven, Belgium.,Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Robin Lemmens
- Department of Neurosciences, KU Leuven, Leuven, Belgium.,Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium.,VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Eric Triau
- Private Practice Neurology, Leuven, Belgium
| | - Kim Serdons
- Department of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium
| | - Jos Tournoy
- Department of Geriatric Medicine, University Hospitals UZ Leuven, Leuven, Belgium.,Department of Public Health and Primary Care, Gerontology and Geriatrics, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Imaging Pathology, KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.,Department of Old-Age Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Imaging Pathology, KU Leuven, Leuven, Belgium.,Department of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Koper MJ, Tomé SO, Gawor K, Belet A, Van Schoor E, Schaeverbeke J, Vandenberghe R, Vandenbulcke M, Ghebremedhin E, Otto M, von Arnim CAF, Balusu S, Blaschko MB, De Strooper B, Thal DR. LATE-NC aggravates GVD-mediated necroptosis in Alzheimer's disease. Acta Neuropathol Commun 2022; 10:128. [PMID: 36057624 PMCID: PMC9441100 DOI: 10.1186/s40478-022-01432-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/15/2022] [Indexed: 12/26/2022] Open
Abstract
It has become evident that Alzheimer's Disease (AD) is not only linked to its hallmark lesions-amyloid plaques and neurofibrillary tangles (NFTs)-but also to other co-occurring pathologies. This may lead to synergistic effects of the respective cellular and molecular players, resulting in neuronal death. One of these co-pathologies is the accumulation of phosphorylated transactive-response DNA binding protein 43 (pTDP-43) as neuronal cytoplasmic inclusions, currently considered to represent limbic-predominant age-related TDP-43 encephalopathy neuropathological changes (LATE-NC), in up to 70% of symptomatic AD cases. Granulovacuolar degeneration (GVD) is another AD co-pathology, which also contains TDP-43 and other AD-related proteins. Recently, we found that all proteins required for necroptosis execution, a previously defined programmed form of neuronal cell death, are present in GVD, such as the phosphorylated necroptosis executioner mixed-lineage kinase domain-like protein (pMLKL). Accordingly, this protein is a reliable marker for GVD lesions, similar to other known GVD proteins. Importantly, it is not yet known whether the presence of LATE-NC in symptomatic AD cases is associated with necroptosis pathway activation, presumably contributing to neuron loss by cell death execution. In this study, we investigated the impact of LATE-NC on the severity of necroptosis-associated GVD lesions, phosphorylated tau (pTau) pathology and neuronal density. First, we used 230 human post-mortem cases, including 82 controls without AD neuropathological changes (non-ADNC), 81 non-demented cases with ADNC, i.e.: pathologically-defined preclinical AD (p-preAD) and 67 demented cases with ADNC. We found that Braak NFT stage and LATE-NC stage were good predictors for GVD expansion and neuronal loss in the hippocampal CA1 region. Further, we compared the impact of TDP-43 accumulation on hippocampal expression of pMLKL-positive GVD, pTau as well as on neuronal density in a subset of nine non-ADNC controls, ten symptomatic AD cases with (ADTDP+) and eight without LATE-NC (ADTDP-). Here, we observed increased levels of pMLKL-positive, GVD-exhibiting neurons in ADTDP+ cases, compared to ADTDP- and controls, which was accompanied by augmented pTau pathology. Neuronal loss in the CA1 region was increased in ADTDP+ compared to ADTDP- cases. These data suggest that co-morbid LATE-NC in AD impacts not only pTau pathology but also GVD-mediated necroptosis pathway activation, which results in an accelerated neuronal demise. This further highlights the cumulative and synergistic effects of comorbid pathologies leading to neuronal loss in AD. Accordingly, protection against necroptotic neuronal death appears to be a promising therapeutic option for AD and LATE.
Collapse
Affiliation(s)
- Marta J Koper
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Sandra O Tomé
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Klara Gawor
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Annelies Belet
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Evelien Van Schoor
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
- Laboratory for Neurobiology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Jolien Schaeverbeke
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Laboratory for Translational Neuropsychiatry, Department of Neuroscience, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Laboratory for Translational Neuropsychiatry, Department of Neuroscience, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Department of Geriatric Psychiatry, UZ Leuven, Leuven, Belgium
| | - Estifanos Ghebremedhin
- Institute of Anatomy - Anatomy I, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
| | - Markus Otto
- Department of Neurology, Ulm University, Ulm, Germany
- Department of Neurology, University of Halle, Halle, Germany
| | - Christine A F von Arnim
- Department of Neurology, Ulm University, Ulm, Germany
- Department of Geriatrics, Göttingen University, Göttingen, Germany
| | - Sriram Balusu
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Matthew B Blaschko
- Department of Electronics, Center for Processing Speech and Images, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
- Department of Pathology, UZ Leuven, Leuven, Belgium.
| |
Collapse
|
34
|
Grimaldi I, Leser FS, Janeiro JM, da Rosa BG, Campanelli AC, Romão L, Lima FRS. The multiple functions of PrP C in physiological, cancer, and neurodegenerative contexts. J Mol Med (Berl) 2022; 100:1405-1425. [PMID: 36056255 DOI: 10.1007/s00109-022-02245-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022]
Abstract
Cellular prion protein (PrPC) is a highly conserved glycoprotein, present both anchored in the cell membrane and soluble in the extracellular medium. It has a diversity of ligands and is variably expressed in numerous tissues and cell subtypes, most notably in the central nervous system (CNS). Its importance has been brought to light over the years both under physiological conditions, such as embryogenesis and immune system homeostasis, and in pathologies, such as cancer and neurodegenerative diseases. During development, PrPC plays an important role in CNS, participating in axonal growth and guidance and differentiation of glial cells, but also in other organs such as the heart, lung, and digestive system. In diseases, PrPC has been related to several types of tumors, modulating cancer stem cells, enhancing malignant properties, and inducing drug resistance. Also, in non-neoplastic diseases, such as Alzheimer's and Parkinson's diseases, PrPC seems to alter the dynamics of neurotoxic aggregate formation and, consequently, the progression of the disease. In this review, we explore in detail the multiple functions of this protein, which proved to be relevant for understanding the dynamics of organism homeostasis, as well as a promising target in the treatment of both neoplastic and degenerative diseases.
Collapse
Affiliation(s)
- Izabella Grimaldi
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Felipe Saceanu Leser
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - José Marcos Janeiro
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bárbara Gomes da Rosa
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Clara Campanelli
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luciana Romão
- Cell Morphogenesis Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Flavia Regina Souza Lima
- Glial Cell Biology Laboratory, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
35
|
Said HM, Kaya D, Yavuz I, Dost FS, Altun ZS, Isik AT. A Comparison of Cerebrospinal Fluid Beta-Amyloid and Tau in Idiopathic Normal Pressure Hydrocephalus and Neurodegenerative Dementias. Clin Interv Aging 2022; 17:467-477. [PMID: 35431542 PMCID: PMC9012339 DOI: 10.2147/cia.s360736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/02/2022] [Indexed: 01/17/2023] Open
Abstract
PURPOSE Idiopathic normal pressure hydrocephalus (iNPH) is the leading reversible cause of cognitive impairment and gait disturbance that has similar clinical manifestations and accompanies to major neurodegenerative disorders in older adults. We aimed to investigate whether cerebrospinal fluid (CSF) biomarker for Alzheimer's disease (AD) may be useful in the differential diagnosis of iNPH. PATIENTS AND METHODS Amyloid-beta (Aß) 42 and 40, total tau (t-tau), phosphorylated tau (p-tau) were measured via ELISA in 192 consecutive CSF samples of patients with iNPH (n=80), AD (n=48), frontotemporal dementia (FTD) (n=34), Lewy body diseases (LBDs) (n=30) consisting of Parkinson's disease dementia and dementia with Lewy bodies. RESULTS The mean age of the study population was 75.6±7.7 years, and 54.2% were female. CSF Aβ42 levels were significantly higher, and p-tau and t-tau levels were lower in iNPH patients than in those with AD and LBDs patients. Additionally, iNPH patients had significantly higher levels of t-tau than those with FTD. Age and sex-adjusted multi-nominal regression analysis revealed that the odds of having AD relative to iNPH decreased by 37% when the Aβ42 level increased by one standard deviation (SD), and the odds of having LBDs relative to iNPH decreased by 47%. The odds of having LBDs relative to iNPH increased 76% when the p-tau level increased 1SD. It is 2.5 times more likely for a patient to have LBD relative to NPH and 2.1 times more likely to have AD relative to iNPH when the t-tau value increased 1SD. CONCLUSION Our results suggest that levels of CSF Aβ42, p-tau, and t-tau, in particularly decreased t-tau, are of potential value in differentiating iNPH from LBDs and also confirm previous studies reporting t-tau level is lower and Aβ42 level is higher in iNPH than in AD.
Collapse
Affiliation(s)
- Harun Muayad Said
- Department of Molecular Medicine, Graduate School of Health Sciences, Dokuz Eylul University, Izmir, Turkey
| | - Derya Kaya
- Unit for Brain Aging and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
- Geriatric Science Association, Izmir, Turkey
| | - Idil Yavuz
- Department of Statistics, Dokuz Eylul University, Faculty of Science, Izmir, Turkey
| | - Fatma Sena Dost
- Unit for Brain Aging and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
- Geriatric Science Association, Izmir, Turkey
| | - Zekiye Sultan Altun
- Department of Basic Oncology, Oncology Institute, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Ahmet Turan Isik
- Unit for Brain Aging and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
- Geriatric Science Association, Izmir, Turkey
| |
Collapse
|
36
|
Zou X, Yuan Y, Liao Y, Jiang C, Zhao F, Ding D, Gu Y, Chen L, Chu Y, Hsu Y, Liebig PA, Xu B, Mao Y. Moyamoya disease: A human model for chronic hypoperfusion and intervention in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12285. [PMID: 35415209 PMCID: PMC8985488 DOI: 10.1002/trc2.12285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/10/2022] [Accepted: 03/03/2022] [Indexed: 11/07/2022]
Abstract
Introduction Chronic cerebral hypoperfusion has been considered the etiology for sporadic Alzheimer's disease (AD). However, no valid clinical evidence exists due to the similar risk factors between cerebrovascular disease and AD. Methods We used moyamoya disease (MMD) as a model of chronic hypoperfusion and cognitive impairment, without other etiology interference. Results Based on the previous reports and preliminary findings, we hypothesized that chronic cerebral hypoperfusion could be an independent upstream crucial variable, resulting in AD, and induce pathological hallmarks such as amyloid beta peptide and hyperphosphorylated tau accumulation. Discussion Timely intervention with revascularisation would help reverse the brain damage with AD hallmarks and lead to cognitive improvement.
Collapse
Affiliation(s)
- Xiang Zou
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Yifan Yuan
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Yujun Liao
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Conglin Jiang
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Fan Zhao
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Ding Ding
- Huashan HospitalInstitute of NeurologyFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Yuxiang Gu
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Liang Chen
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
- Tianqiao and Chrissy Chen International Institute for Brain DiseasesShanghaiChina
| | - Ying‐Hua Chu
- MR CollaborationSiemens Healthineers Ltd.ShanghaiChina
| | - Yi‐Cheng Hsu
- MR CollaborationSiemens Healthineers Ltd.ShanghaiChina
| | | | - Bin Xu
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
| | - Ying Mao
- Department of NeurosurgeryHuashan HospitalFudan UniversityShanghaiChina
- Neurosurgical Institute of Fudan UniversityShanghaiChina
- Shanghai Clinical Medical Center of NeurosurgeryShanghaiChina
- Shanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationShanghaiChina
- Huashan HospitalInstitute of NeurologyFudan UniversityShanghaiChina
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceSchool of Basic Medical Sciences and Institutes of Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
37
|
Mohammadi B, Song F, Matamoros-Angles A, Shafiq M, Damme M, Puig B, Glatzel M, Altmeppen HC. Anchorless risk or released benefit? An updated view on the ADAM10-mediated shedding of the prion protein. Cell Tissue Res 2022; 392:215-234. [PMID: 35084572 PMCID: PMC10113312 DOI: 10.1007/s00441-022-03582-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/12/2022] [Indexed: 11/24/2022]
Abstract
The prion protein (PrP) is a broadly expressed glycoprotein linked with a multitude of (suggested) biological and pathological implications. Some of these roles seem to be due to constitutively generated proteolytic fragments of the protein. Among them is a soluble PrP form, which is released from the surface of neurons and other cell types by action of the metalloprotease ADAM10 in a process termed 'shedding'. The latter aspect is the focus of this review, which aims to provide a comprehensive overview on (i) the relevance of proteolytic processing in regulating cellular PrP functions, (ii) currently described involvement of shed PrP in neurodegenerative diseases (including prion diseases and Alzheimer's disease), (iii) shed PrP's expected roles in intercellular communication in many more (patho)physiological conditions (such as stroke, cancer or immune responses), (iv) and the need for improved research tools in respective (future) studies. Deeper mechanistic insight into roles played by PrP shedding and its resulting fragment may pave the way for improved diagnostics and future therapeutic approaches in diseases of the brain and beyond.
Collapse
Affiliation(s)
- Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- Working Group for Interdisciplinary Neurobiology and Immunology (INI Research), Hamburg, Germany
| | - Feizhi Song
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Andreu Matamoros-Angles
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Damme
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | |
Collapse
|
38
|
Kovač V, Čurin Šerbec V. Prion Protein: The Molecule of Many Forms and Faces. Int J Mol Sci 2022; 23:ijms23031232. [PMID: 35163156 PMCID: PMC8835406 DOI: 10.3390/ijms23031232] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/10/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Cellular prion protein (PrPC) is a glycosylphosphatidylinositol (GPI)-anchored protein most abundantly found in the outer membrane of neurons. Due to structural characteristics (a flexible tail and structured core), PrPC interacts with a wide range of partners. Although PrPC has been proposed to be involved in many physiological functions, only peripheral nerve myelination homeostasis has been confirmed as a bona fide function thus far. PrPC misfolding causes prion diseases and PrPC has been shown to mediate β-rich oligomer-induced neurotoxicity in Alzheimer’s and Parkinson’s disease as well as neuroprotection in ischemia. Upon proteolytic cleavage, PrPC is transformed into released and attached forms of PrP that can, depending on the contained structural characteristics of PrPC, display protective or toxic properties. In this review, we will outline prion protein and prion protein fragment properties as well as overview their involvement with interacting partners and signal pathways in myelination, neuroprotection and neurodegenerative diseases.
Collapse
|
39
|
Roda AR, Serra-Mir G, Montoliu-Gaya L, Tiessler L, Villegas S. Amyloid-beta peptide and tau protein crosstalk in Alzheimer's disease. Neural Regen Res 2022; 17:1666-1674. [PMID: 35017413 PMCID: PMC8820696 DOI: 10.4103/1673-5374.332127] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease is a neurodegenerative disease that accounts for most of the 50-million dementia cases worldwide in 2018. A large amount of evidence supports the amyloid cascade hypothesis, which states that amyloid-beta accumulation triggers tau hyperphosphorylation and aggregation in form of neurofibrillary tangles, and these aggregates lead to inflammation, synaptic impairment, neuronal loss, and thus to cognitive decline and behavioral abnormalities. The poor correlation found between cognitive decline and amyloid plaques, have led the scientific community to question whether amyloid-beta accumulation is actually triggering neurodegeneration in Alzheimer’s disease. The occurrence of tau neurofibrillary tangles better correlates to neuronal loss and clinical symptoms and, although amyloid-beta may initiate the cascade of events, tau impairment is likely the effector molecule of neurodegeneration. Recently, it has been shown that amyloid-beta and tau cooperatively work to impair transcription of genes involved in synaptic function and, more importantly, that downregulation of tau partially reverses transcriptional perturbations. Despite mounting evidence points to an interplay between amyloid-beta and tau, some factors could independently affect both pathologies. Thus, the dual pathway hypothesis, which states that there are common upstream triggers causing both amyloid-beta and tau abnormalities has been proposed. Among others, the immune system seems to be strongly involved in amyloid-beta and tau pathologies. Other factors, as the apolipoprotein E ε4 isoform has been suggested to act as a link between amyloid-beta and tau hyperphosphorylation. Interestingly, amyloid-beta-immunotherapy reduces not only amyloid-beta but also tau levels in animal models and in clinical trials. Likewise, it has been shown that tau-immunotherapy also reduces amyloid-beta levels. Thus, even though amyloid-beta immunotherapy is more advanced than tau-immunotherapy, combined amyloid-beta and tau-directed therapies at early stages of the disease have recently been proposed as a strategy to stop the progression of Alzheimer’s disease.
Collapse
Affiliation(s)
- Alejandro R Roda
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Gabriel Serra-Mir
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Lidia Tiessler
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Sandra Villegas
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
40
|
Zhang T, Xia Y, Hu L, Chen D, Gan CL, Wang L, Mei Y, Lan G, Shui X, Tian Y, Li R, Zhang M, Lee TH. Death-associated protein kinase 1 mediates Aβ42 aggregation-induced neuronal apoptosis and tau dysregulation in Alzheimer's disease. Int J Biol Sci 2022; 18:693-706. [PMID: 35002518 PMCID: PMC8741852 DOI: 10.7150/ijbs.66760] [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: 09/02/2021] [Accepted: 11/27/2021] [Indexed: 12/22/2022] Open
Abstract
The aggregation of amyloid-β (Aβ) peptides into oligomers and fibrils is a key pathological feature of Alzheimer's disease (AD). An increasing amount of evidence suggests that oligomeric Aβ might be the major culprit responsible for various neuropathological changes in AD. Death-associated protein kinase 1 (DAPK1) is abnormally elevated in brains of AD patients and plays an important role in modulating tau homeostasis by regulating prolyl isomerase Pin1 phosphorylation. However, it remains elusive whether and how Aβ species influence the function of DAPK1, and whether this may further affect the function and phosphorylation of tau in neurons. Herein, we demonstrated that Aβ aggregates (both oligomers and fibrils) prepared from synthetic Aβ42 peptides were able to upregulate DAPK1 protein levels and thereby its function through heat shock protein 90 (HSP90)-mediated protein stabilization. DAPK1 activation not only caused neuronal apoptosis, but also phosphorylated Pin1 at the Ser71 residue, leading to tau accumulation and phosphorylation at multiple AD-related sites in primary neurons. Both DAPK1 knockout (KO) and the application of a specific DAPK1 inhibitor could effectively protect primary neurons against Aβ aggregate-induced cell death and tau dysregulation, corroborating the critical role of DAPK1 in mediating Aβ aggregation-induced neuronal damage. Our study suggests a mechanistic link between Aβ oligomerization and tau hyperphosphorylation mediated by DAPK1, and supports the role of DAPK1 as a promising target for early intervention in AD.
Collapse
Affiliation(s)
- Tao Zhang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Yongfang Xia
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Li Hu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Dongmei Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Chen-Ling Gan
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Long Wang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Yingxue Mei
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Guihua Lan
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Xindong Shui
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Yuan Tian
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Ruomeng Li
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Mi Zhang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Tae Ho Lee
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| |
Collapse
|
41
|
Frisoni GB, Altomare D, Thal DR, Ribaldi F, van der Kant R, Ossenkoppele R, Blennow K, Cummings J, van Duijn C, Nilsson PM, Dietrich PY, Scheltens P, Dubois B. The probabilistic model of Alzheimer disease: the amyloid hypothesis revised. Nat Rev Neurosci 2022; 23:53-66. [PMID: 34815562 PMCID: PMC8840505 DOI: 10.1038/s41583-021-00533-w] [Citation(s) in RCA: 242] [Impact Index Per Article: 80.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 01/03/2023]
Abstract
The current conceptualization of Alzheimer disease (AD) is driven by the amyloid hypothesis, in which a deterministic chain of events leads from amyloid deposition and then tau deposition to neurodegeneration and progressive cognitive impairment. This model fits autosomal dominant AD but is less applicable to sporadic AD. Owing to emerging information regarding the complex biology of AD and the challenges of developing amyloid-targeting drugs, the amyloid hypothesis needs to be reconsidered. Here we propose a probabilistic model of AD in which three variants of AD (autosomal dominant AD, APOE ε4-related sporadic AD and APOE ε4-unrelated sporadic AD) feature decreasing penetrance and decreasing weight of the amyloid pathophysiological cascade, and increasing weight of stochastic factors (environmental exposures and lower-risk genes). Together, these variants account for a large share of the neuropathological and clinical variability observed in people with AD. The implementation of this model in research might lead to a better understanding of disease pathophysiology, a revision of the current clinical taxonomy and accelerated development of strategies to prevent and treat AD.
Collapse
Affiliation(s)
- Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland.
| | - Daniele Altomare
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, and Leuven Brain Institute, University of Leuven, Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Centro S. Giovanni di Dio Fatebenefratelli, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Rik van der Kant
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Kaj Blennow
- Cinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences; University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Cornelia van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | | | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Life Science Partners, Amsterdam, Netherlands
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d'Alzheimer, IM2A, Groupe Hospitalier Pitié-Salpêtrière, Sorbonne Université, Paris, France
- Institut du Cerveau et de la Moelle Épinière, UMR-S975, INSERM, Paris, France
| |
Collapse
|
42
|
Libard S, Giedraitis V, Kilander L, Ingelsson M, Alafuzoff I. Mixed Pathologies in a Subject with a Novel PSEN1 G206R Mutation. J Alzheimers Dis 2022; 90:1601-1614. [PMID: 36314207 PMCID: PMC9789486 DOI: 10.3233/jad-220655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND There are more than 300 presenilin-1 (PSEN1) mutations identified but a thorough postmortem neuropathological assessment of the mutation carriers is seldom performed. OBJECTIVE To assess neuropathological changes (NC) in a 73-year-old subject with the novel PSEN1 G206R mutation suffering from cognitive decline in over 20 years. To compare these findings with an age- and gender-matched subject with sporadic Alzheimer's disease (sAD). METHODS The brains were assessed macro- and microscopically and the proteinopathies were staged according to current recommendations. RESULTS The AD neuropathological change (ADNC) was more extensive in the mutation carrier, although both individuals reached a high level of ADNC. The transactive DNA binding protein 43 pathology was at the end-stage in the index subject, a finding not previously described in familial AD. This pathology was moderate in the sAD subject. The PSEN1 G206R subject displayed full-blown alpha-synuclein pathology, while this proteinopathy was absent in the sAD case. Additionally, the mutation carrier displayed pronounced neuroinflammation, not previously described in association with PSEN1 mutations. CONCLUSION Our findings are exceptional, as the PSEN1 G206R subject displayed an end-stage pathology of every common proteinopathy. It is unclear whether the observed alterations are caused by the mutation or are related to a cross-seeding mechanisms. The pronounced neuroinflammation in the index patient can be reactive to the extensive NC or a contributing factor to the proteinopathies. Thorough postmortem neuropathological and genetic assessment of subjects with familial AD is warranted, for further understanding of a dementing illness.
Collapse
Affiliation(s)
- Sylwia Libard
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Surgical Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Lena Kilander
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Department of Medicine and Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Irina Alafuzoff
- Department of Surgical Pathology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
43
|
Jellinger KA. Recent update on the heterogeneity of the Alzheimer’s disease spectrum. J Neural Transm (Vienna) 2021; 129:1-24. [DOI: 10.1007/s00702-021-02449-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/25/2021] [Indexed: 02/03/2023]
|
44
|
Vatsa P, Negi R, Ansari UA, Khanna VK, Pant AB. Insights of Extracellular Vesicles of Mesenchymal Stem Cells: a Prospective Cell-Free Regenerative Medicine for Neurodegenerative Disorders. Mol Neurobiol 2021; 59:459-474. [PMID: 34714469 DOI: 10.1007/s12035-021-02603-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent, adult stem cells which are found in numerous tissues like the umbilical cord, Wharton's jelly, bone marrow, and adipose tissue. They possess the capacity of self-renewal by dividing and differentiating into various cellular lineages. Their characteristic therapeutic potential exploited so far has made them a desirable candidate in regenerative medicine. Neurodegenerative diseases (NDs) like Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), and ischemic stroke have been treated with MSCs and MSC-derived products. Over the past few decades, we have witnessed significant contributions in discovering the etiology of various NDs and their possible therapeutic solutions. One of the MSC-based therapeutics is extracellular vesicles (EVs), which contain multiple biologically active molecules like nucleic acids and proteins. The contents of EVs are ferried between cells for intercellular communication which then leads to regulation of the homeostasis of recipient cells. EVs serve as a considerable means of cell-free therapies like for tissue repair or regeneration as EVs can maintain therapeutically effective cargo of parent cells and are free of various ethical issues in cell-based therapies. Due to paucity of standard protocols in extraction procedures of EVs and their pharmacological properties and mechanisms, the development of new EV dependent therapies is challenging. With this review, an attempt has been made to annotate these mechanisms, which can help advance the novel therapeutic approaches towards the treat and define a more narrowed down approach for each ND to devise effective MSC-based therapies to cure and avert these diseases.
Collapse
Affiliation(s)
- P Vatsa
- System Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow, Uttar Pradesh, 226001, India
- CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - R Negi
- System Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow, Uttar Pradesh, 226001, India
- CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - U A Ansari
- System Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow, Uttar Pradesh, 226001, India
- CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - V K Khanna
- System Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow, Uttar Pradesh, 226001, India
- CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - A B Pant
- System Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow, Uttar Pradesh, 226001, India.
- CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
45
|
Jellinger KA, Wenning GK, Stefanova N. Is Multiple System Atrophy a Prion-like Disorder? Int J Mol Sci 2021; 22:10093. [PMID: 34576255 PMCID: PMC8472631 DOI: 10.3390/ijms221810093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023] Open
Abstract
Multiple system atrophy (MSA) is a rapidly progressive, fatal neurodegenerative disease of uncertain aetiology that belongs to the family of α-synucleinopathies. It clinically presents with parkinsonism, cerebellar, autonomic, and motor impairment in variable combinations. Pathological hallmarks are fibrillary α-synuclein (αSyn)-rich glial cytoplasmic inclusions (GCIs) mainly involving oligodendroglia and to a lesser extent neurons, inducing a multisystem neurodegeneration, glial activation, and widespread demyelinization. The neuronal αSyn pathology of MSA has molecular properties different from Lewy bodies in Parkinson's disease (PD), both of which could serve as a pool of αSyn (prion) seeds that could initiate and drive the pathogenesis of synucleinopathies. The molecular cascade leading to the "prion-like" transfer of "strains" of aggregated αSyn contributing to the progression of the disease is poorly understood, while some presented evidence that MSA is a prion disease. However, this hypothesis is difficult to reconcile with postmortem analysis of human brains and the fact that MSA-like pathology was induced by intracerebral inoculation of human MSA brain homogenates only in homozygous mutant 53T mice, without production of disease-specific GCIs, or with replication of MSA prions in primary astrocyte cultures from transgenic mice expressing human αSyn. Whereas recent intrastriatal injection of Lewy body-derived or synthetic human αSyn fibrils induced PD-like pathology including neuronal αSyn aggregates in macaques, no such transmission of αSyn pathology in non-human primates by MSA brain lysate has been reported until now. Given the similarities between αSyn and prions, there is a considerable debate whether they should be referred to as "prions", "prion-like", "prionoids", or something else. Here, the findings supporting the proposed nature of αSyn as a prion and its self-propagation through seeding as well as the transmissibility of neurodegenerative disorders are discussed. The proof of disease causation rests on the concordance of scientific evidence, none of which has provided convincing evidence for the classification of MSA as a prion disease or its human transmission until now.
Collapse
Affiliation(s)
| | - Gregor K. Wenning
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (G.K.W.); (N.S.)
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (G.K.W.); (N.S.)
| |
Collapse
|
46
|
Van Kolen K, Malia TJ, Theunis C, Nanjunda R, Teplyakov A, Ernst R, Wu SJ, Luo J, Borgers M, Vandermeeren M, Bottelbergs A, Wintmolders C, Lacy E, Maurin H, Larsen P, Willems R, Van De Casteele T, Triana-Baltzer G, Slemmon R, Galpern W, Trojanowski JQ, Sun H, Mercken MH. Discovery and Functional Characterization of hPT3, a Humanized Anti-Phospho Tau Selective Monoclonal Antibody. J Alzheimers Dis 2021; 77:1397-1416. [PMID: 32894244 DOI: 10.3233/jad-200544] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND As a consequence of the discovery of an extracellular component responsible for the progression of tau pathology, tau immunotherapy is being extensively explored in both preclinical and clinical studies as a disease modifying strategy for the treatment of Alzheimer's disease. OBJECTIVE Describe the characteristics of the anti-phospho (T212/T217) tau selective antibody PT3 and its humanized variant hPT3. METHODS By performing different immunization campaigns, a large collection of antibodies has been generated and prioritized. In depth, in vitro characterization using surface plasmon resonance, phospho-epitope mapping, and X-ray crystallography experiments were performed. Further characterization involved immunohistochemical staining on mouse- and human postmortem tissue and neutralization of tau seeding by immunodepletion assays. RESULTS AND CONCLUSION Various in vitro experiments demonstrated a high intrinsic affinity for PT3 and hPT3 for AD brain-derived paired helical filaments but also to non-aggregated phospho (T212/T217) tau. Further functional analyses in cellular and in vivo models of tau seeding demonstrated almost complete depletion of tau seeds in an AD brain homogenate. Ongoing trials will provide the clinical evaluation of the tau spreading hypothesis in Alzheimer's disease.
Collapse
Affiliation(s)
- Kristof Van Kolen
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Thomas J Malia
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Clara Theunis
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Rupesh Nanjunda
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Alexey Teplyakov
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Robin Ernst
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Sheng-Jiun Wu
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Jinquan Luo
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Marianne Borgers
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Marc Vandermeeren
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Astrid Bottelbergs
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Cindy Wintmolders
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Eilyn Lacy
- Biologics Research, Janssen Research and Development, Spring House, PA, USA
| | - Hervé Maurin
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Peter Larsen
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Roland Willems
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| | - Tom Van De Casteele
- Translational Medicine and Early Development Statistics Janssen Research & Development, Beerse, Belgium
| | | | - Randy Slemmon
- Neuroscience biomarkers, Janssen Research & Development, La Jolla, CA, USA
| | - Wendy Galpern
- Neuroscience Experimental medicine, Janssen Research & Development, Titusville, NJ, USA
| | | | - Hong Sun
- Neuroscience Clinical Development, Janssen Research & Development, Titusville, NJ, USA
| | - Marc H Mercken
- Neuroscience Department, Janssen Research and Development, Beerse, Belgium
| |
Collapse
|
47
|
Wang P, Ye Y. Astrocytes in Neurodegenerative Diseases: A Perspective from Tauopathy and α-Synucleinopathy. Life (Basel) 2021; 11:life11090938. [PMID: 34575087 PMCID: PMC8471224 DOI: 10.3390/life11090938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022] Open
Abstract
Neurodegenerative diseases are aging-associated chronic pathological conditions affecting primarily neurons in humans. Inclusion bodies containing misfolded proteins have emerged as a common pathologic feature for these diseases. In many cases, misfolded proteins produced by a neuron can be transmitted to another neuron or a non-neuronal cell, leading to the propagation of disease-associated pathology. While undergoing intercellular transmission, misfolded proteins released from donor cells can often change the physiological state of recipient cells. Accumulating evidence suggests that astrocytes are highly sensitive to neuron-originated proteotoxic insults, which convert them into an active inflammatory state. Conversely, activated astrocytes can release a plethora of factors to impact neuronal functions. This review summarizes our current understanding of the complex molecular interplays between astrocyte and neuron, emphasizing on Tau and α-synuclein (α-syn), the disease-driving proteins for Alzheimer's and Parkinson's diseases, respectively.
Collapse
Affiliation(s)
| | - Yihong Ye
- Correspondence: ; Tel.: +1-301-594-0845; Fax: +1-301-496-0201
| |
Collapse
|
48
|
Dexter E, Kong Q. Neuroprotective effect and potential of cellular prion protein and its cleavage products for treatment of neurodegenerative disorders part I. a literature review. Expert Rev Neurother 2021; 21:969-982. [PMID: 34470561 DOI: 10.1080/14737175.2021.1965881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The cellular prion protein (PrPC) is well known for its pathogenic roles in prion diseases, several other neurodegenerative diseases (such as Alzheimer's disease), and multiple types of cancer, but the beneficial aspects of PrPC and its cleavage products received much less attention. AREAS COVERED Here the authors will systematically review the literatures on the negative as well as protective aspects of PrPC and its derivatives (especially PrP N-terminal N1 peptide and shed PrP). The authors will dissect the current findings on N1 and shed PrP, including evidence for their neuroprotective effects, the categories of PrPC cleavage, and numerous cleavage enzymes involved. The authors will also discuss the protective effects and therapeutic potentials of PrPC-rich exosomes. The cited articles were obtained from extensive PubMed searches of recent literature, including peer-reviewed original articles and review articles. EXPERT OPINION PrP and its N-terminal fragments have strong neuroprotective activities that should be explored for therapeutics and prophylactics development against prion disease, Alzheimer's disease and a few other neurodegenerative diseases. The strategies to develop PrP-based therapeutics and prophylactics for these neurodegenerative diseases will be discussed in a companion article (Part II).
Collapse
Affiliation(s)
- Emily Dexter
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, USA
| | - Qingzhong Kong
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, USA.,Department of Neurology, School of Medicine, Case Western Reserve University, Cleveland, USA
| |
Collapse
|
49
|
Abstract
Prion diseases are neurodegenerative disorders caused by conformational conversion of the cellular prion protein (PrPC) into scrapie prion protein (PrPSc). As the main component of prion, PrPSc acts as an infectious template that recruits and converts normal cellular PrPC into its pathogenic, misfolded isoform. Intriguingly, the phenomenon of prionoid, or prion-like, spread has also been observed in many other disease-associated proteins, such as amyloid β (Aβ), tau and α-synuclein. This Cell Science at a Glance and the accompanying poster highlight recently described physiological roles of prion protein and the advanced understanding of pathogenesis of prion disease they have afforded. Importantly, prion protein may also be involved in the pathogenesis of other neurodegenerative disorders such as Alzheimer's and Parkinson's disease. Therapeutic studies of prion disease have also exploited novel strategies to combat these devastating diseases. Future studies on prion protein and prion disease will deepen our understanding of the pathogenesis of a broad spectrum of neurodegenerative conditions.
Collapse
Affiliation(s)
- Caihong Zhu
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zürich, Zürich, CH-8091, Switzerland
| |
Collapse
|
50
|
Niu F, Sharma A, Wang Z, Feng L, Muresanu DF, Sahib S, Tian ZR, Lafuente JV, Buzoianu AD, Castellani RJ, Nozari A, Menon PK, Patnaik R, Wiklund L, Sharma HS. Nanodelivery of oxiracetam enhances memory, functional recovery and induces neuroprotection following concussive head injury. PROGRESS IN BRAIN RESEARCH 2021; 265:139-230. [PMID: 34560921 DOI: 10.1016/bs.pbr.2021.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Military personnel are the most susceptible to concussive head injury (CHI) caused by explosion, blast or missile or blunt head trauma. Mild to moderate CHI could induce lifetime functional and cognitive disturbances causing significant decrease in quality of life. Severe CHI leads to instant death and lifetime paralysis. Thus, further exploration of novel therapeutic agents or new features of known pharmacological agents are needed to enhance quality of life of CHI victims. Previous reports from our laboratory showed that mild CHI induced by weight drop technique causing an impact of 0.224N results in profound progressive functional deficit, memory impairment and brain pathology from 5h after trauma that continued over several weeks of injury. In this investigation we report that TiO2 nanowired delivery of oxiracetam (50mg/kg, i.p.) daily for 5 days after CHI resulted in significant improvement of functional deficit on the 8th day. This was observed using Rota Rod treadmill, memory improvement assessed by the time spent in finding hidden platform under water. The motor function improvement is seen in oxiracetam treated CHI group by placing forepaw on an inclined mesh walking and foot print analysis for stride length and distance between hind feet. TiO2-nanowired oxiracetam also induced marked improvements in the cerebral blood flow, reduction in the BBB breakdown and edema formation as well as neuroprotection of neuronal, glial and myelin damages caused by CHI at light and electron microscopy on the 7th day after 5 days TiO2 oxiracetam treatment. Adverse biochemical events such as upregulation of CSF nitrite and nitrate, IL-6, TNF-a and p-Tau are also reduced significantly in oxiracetam treated CHI group. On the other hand post treatment of 100mg/kg dose of normal oxiracetam in identical conditions after CHI is needed to show slight but significant neuroprotection together with mild recovery of memory function and functional deficits on the 8th day. These observations are the first to point out that nanowired delivery of oxiracetam has superior neuroprotective ability in CHI. These results indicate a promising clinical future of TiO2 oxiracetam in treating CHI patients for better quality of life and neurorehabilitation, not reported earlier.
Collapse
Affiliation(s)
- Feng Niu
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Zhenguo Wang
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|