1
|
Lavrova A, Pham NTT, Reid RI, Boeve BF, Knopman DS, Petersen RC, Nguyen AT, Ross Reichard R, Dickson DW, Jack CR, Whitwell JL, Josephs KA. Relation of Alzheimer's disease-related TDP-43 proteinopathy to metrics from diffusion tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI). Neurobiol Aging 2025; 150:97-108. [PMID: 40088623 DOI: 10.1016/j.neurobiolaging.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025]
Abstract
Transactive response DNA-binding protein 43 kDa (TDP-43) deposition is linked to regional brain atrophy in Alzheimer's disease (AD), but diffusion changes associated with AD-related TDP-43 proteinopathy remain underexplored. This study evaluates the potential of diffusion tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI) as in vivo markers for detecting TDP-43 proteinopathy in AD. We analyzed DTI and NODDI metrics in 49 cases with AD neuropathologic changes, categorized by postmortem TDP-43 status. Diffusion metrics from the temporal lobe gray and white matter regions and key white matter tracts were compared between TDP-43-positive and negative cases. Group differences were significant in the left hippocampus, amygdala, and uncinate fasciculus after adjusting for age, Braak neurofibrillary tangle (NFT) stage and APOE ε4 status. TDP-43-positive cases showed increased mean diffusivity (MD) and altered neurite density index (NDI) and orientation dispersion index (ODI). Area under the receiver operating characteristic curve (AUROC) analysis revealed high predictive accuracy for amygdala ODI (AUC = 0.809, sensitivity = 0.81, specificity = 0.76), hippocampal MD (AUC = 0.763, sensitivity = 0.81, specificity = 0.67), and uncinate fasciculus MD (AUC = 0.782, sensitivity = 0.88, specificity = 0.61). Combined, DTI/NODDI predictors demonstrated stronger discriminative ability (AUC = 0.856, sensitivity = 0.88, specificity = 0.76). These findings suggest that AD-related TDP-43 proteinopathy is associated with specific diffusion changes in the left temporal lobe. DTI and NODDI metrics, particularly MD, NDI, and ODI, may improve the antemortem detection of TDP-43 pathology in AD.
Collapse
Affiliation(s)
- Anna Lavrova
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Robert I Reid
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | - R Ross Reichard
- Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | | | | | | | | |
Collapse
|
2
|
Sadeghi A, Noorbakhshnia M, Khodashenas S. Protective potential of BM-MSC extracted Exosomes in a rat model of Alzheimer's disease. PLoS One 2025; 20:e0320883. [PMID: 40327601 PMCID: PMC12054907 DOI: 10.1371/journal.pone.0320883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 02/25/2025] [Indexed: 05/08/2025] Open
Abstract
Exosomes are extracellular vesicles, which are released into the extracellular space by all types of cells, especially stem cells. Compared with stem cells, exosomes are safer and can be considered one of the most promising therapeutic strategies for neurodegenerative disease. We examined the effect of exosomes derived from bone marrow mesenchymal stem cells (BM-MSC) on a rat model of Alzheimer's disease (AD). For this purpose, male Wistar rats weighing 220-250 g were used. For the induction of AD, rats received a daily dosage of 100 mg/kg Aluminum chloride (Alcl3) by oral gavage for 60 days. Also, Primary BM-MSC was extracted from the femora of Wistar rats (male, 100-150 g). Extracted exosomes were Characterized and Qualified using TEM Microscope and Zetasizer Nano. Specific markers of exosomes were evaluated by Flow cytometry. MSC-extracted exosomes (150 µg/µl) were injected 2 or 5 times into the animals via tail vein on specific days. Our data revealed that receiving exosomes significantly prevented AlCl3-induced enhancement of hippocampal APP gene expression, beta-amyloid plaque formation, impairment of passive avoidance learning and spatial memory. However, exosome injections in healthy subjects caused some negative effects such as spatial memory impairment. It seems, MSC-derived exosomes can be considered as a candidate to prevent AD progression.
Collapse
Affiliation(s)
- Atefeh Sadeghi
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Maryam Noorbakhshnia
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Shabanali Khodashenas
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Thalassemia Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
3
|
Mondal R, Deb S, Shome G, Sarkar V, Lahiri D, Datta SS, Benito-León J. Molecular dynamics of amyloid-β transport in Alzheimer's disease: Exploring therapeutic plasma exchange with albumin replacement - Current insights and future perspectives. Neurologia 2025; 40:306-328. [PMID: 40280630 DOI: 10.1016/j.nrleng.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 11/07/2023] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION The complex process of amyloid-β (Aβ) transportation across the blood-brain and blood-cerebrospinal fluid barriers is crucial for preventing Aβ accumulation, which linked to dementia and neurodegeneration. This review explores therapeutic plasma exchange with albumin replacement in Alzheimer's disease, based on the dynamics of amyloid-β between the brain, plasma, and cerebrospinal fluid. METHODOLOGY A comprehensive literature review was conducted using PubMed/Medline, Cochrane Library, and open databases (bioRxiv, MedRixv, preprint.org) up to April 30, 2023. The first search utilized the following MeSH terms and keywords: 'Plasma Exchange', 'Plasmapheresis', 'Therapeutic plasma exchange', 'Apheresis', 'Aβ', 'p-tau', 'Total-tau', 'Alzheimer's disease', 'Cognitive dysfunction', 'neurodegenerative diseases', 'centrifugation', 'membranous', and 'filtration' in the Title/Abstract, yielding 146 results. A second search with the keywords: 'Albumin', 'Aβ', 'BBB', 'Alzheimer's dementia', and 'Nerve degeneration' resulted in 125 additional articles for analysis. Finally, a third search using keywords: 'Albumin structural domains', 'Albumin-Aβ interactions', 'Albumin-endothelial interactions', and 'Post-Translational Modification' produced 193 results for further review. RESULTS/DISCUSSION Therapeutic plasma exchange shows potential as a disease-modifying therapy for dementia, specifically for Alzheimer's disease. Additionally, the promising role of albumin supplementation in cognitive improvement has attracted attention. However, clinical evidence supporting therapeutic plasma exchange for dementia remains limited, necessitating further research and development to mitigate potential adverse effects. A deeper understanding of the molecular dynamics of Aβ transportation and the mechanisms of therapeutic plasma exchange is essential. A critical evaluation of existing evidence highlights the importance of balancing potential benefits with associated risks, which will guide the development and application of these treatments in neurodegenerative diseases.
Collapse
Affiliation(s)
- R Mondal
- Department of Clinical Pharmacology and Therapeutic Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India
| | - S Deb
- Department of Neuroscience, SN Pradhan Center for Neuroscience, University of Calcutta, Kolkata 700019, India
| | - G Shome
- Department of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | - V Sarkar
- Department of Neuroscience, SN Pradhan Center for Neuroscience, University of Calcutta, Kolkata 700019, India
| | - D Lahiri
- Baycrest Academy of Research and Education, Toronto, Canada; Rotman Research Institute, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Canada; Department of Neurology, Institute of Neurosciences, Kolkata, India
| | - S S Datta
- Department of Transfusion Medicine, Tata Medical Center, Kolkata 700160, India
| | - J Benito-León
- Department of Neurology, University Hospital "12 de Octubre", Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Department of Medicine, Faculty of Medicine, Complutense University, Madrid, Spain.
| |
Collapse
|
4
|
Youssef H, Gatto RG, Pham NTT, Jones D, Petersen RC, Machulda MM, Whitwell JL, Josephs KA. Multiple Neuropathologies Underly Hippocampal Subfield Atrophy in a Case With a Slowly Progressive Amnestic Syndrome: Challenging the Notion of Pure LATE-NC. Neuropathology 2025. [PMID: 39973236 DOI: 10.1111/neup.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/21/2025]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly, marked by abnormal protein buildup (beta-amyloid and tau) resulting in neuronal loss, especially in the medial temporal lobe and other limbic regions. The presence of transactive response DNA binding protein 43 (TDP-43) immunoreactive inclusions in medial temporal lobe regions has also been associated with neuroimaging changes in limbic regions. It has been proposed that hypometabolism in limbic regions on [18F] fluorodeoxyglucose positron emission tomography (FDG-PET) in a patient with a slowly evolving amnestic syndrome may be a signature of the presence of TDP-43. In this context, we observed an 86-year-old Caucasian female with dementia characterized by a slowly evolving amnestic syndrome, along with focal medial temporal atrophy evident on MRI and hypometabolism in limbic regions on FDG-PET. The patient subsequently died and underwent an autopsy. We performed detailed neuroimaging and digital neuropathological analyses of the hippocampal subfields to better understand the relationship between clinico-imaging findings and histopathology. In addition to TDP-43, we identified three other pathological processes in the medial temporal lobe: sequestosome-1/p62, argyrophilic grain disease (AGD), and primary age-related tauopathy (PART). Hippocampal subfield volumes and rates of atrophy were no different from those of matched healthy controls, except for the atrophy rate in cornu ammonis 1 (CA1). Digital histopathology revealed the relative highest burden of pathology for p62, followed by TDP-43, AGD, and PART in CA1. Multiple pathological processes appear to have contributed to the hippocampal atrophy and hypometabolism in our patient with a slowly progressive amnestic syndrome.
Collapse
Affiliation(s)
- Hossam Youssef
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rodolfo G Gatto
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - David Jones
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Mary M Machulda
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Carlos AF, Weigand SD, Pham NTT, Petersen RC, Jack CR, Dickson DW, Whitwell JL, Josephs KA. White matter hyperintensities and TDP-43 pathology in Alzheimer's disease. Alzheimers Dement 2025; 21:ealz14516. [PMID: 39821594 PMCID: PMC11851154 DOI: 10.1002/alz.14516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/06/2024] [Accepted: 12/10/2024] [Indexed: 01/19/2025]
Abstract
INTRODUCTION Greater white matter hyperintensities (WMHs) on magnetic resonance imaging (MRI) are seen with transactive response DNA-binding protein 43 (TDP-43) pathology in frontotemporal lobar degeneration (FTLD-TDP). WMH associations with TDP-43 pathology in Alzheimer's disease (AD-TDP) remain unclear. METHODS A total of 157 participants from Mayo Clinic Rochester with autopsy-confirmed AD, known TDP-43 status, and antemortem fluid-attenuated inversion recovery (FLAIR) MRI were included. Vascular risk factors were assessed. A semi-automated WMH segmentation-quantification process produced total and regional WMH volumes. Penalized linear regression models adjusting for age at MRI analyzed TDP-43 associations (status and typing) with WMHs. RESULTS TDP-43-positive status was not associated with WMH burden overall because opposite effects were seen based on AD-TDP typing. Despite similar antemortem vascular risk factors and postmortem vascular pathologies, AD-TDP type-α showed greater total and regional WMH burden (particularly in subcortical frontotemporal and basal ganglia regions) than TDP-43 negatives and AD-TDP type-β. DISCUSSION AD-TDP types may have different WMH pathomechanisms, with type-α having associations more like FTLD-TDP than AD. HIGHLIGHTS In transactive response DNA-binding protein 43 (TDP-43) pathology in Alzheimer's disease (AD-TDP), TDP-43 status alone is not associated with total or regional WMH burden AD-TDP type-α shows greater total, frontotemporal subcortical, and basal ganglia white matter hyperintensities (WMHs) AD-TDP type-β shows less total and subcortical occipital WMHs AD-TDP type-α effect on WMH burden closely mimics the effects of frontotemporal lobar degeneration with TDP-43 (FTLD-TDP) rather than AD Different relationships of AD-TDP types with WMHs suggest different pathomechanisms.
Collapse
Affiliation(s)
| | - Stephen D. Weigand
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | | | | | | | - Dennis W. Dickson
- Department of Neuroscience (Neuropathology)Mayo ClinicJacksonvilleFloridaUSA
| | | | | |
Collapse
|
6
|
Wisse LEM, Wuestefeld A, Murray ME, Jagust W, La Joie R. Role of tau versus TDP-43 pathology on medial temporal lobe atrophy in aging and Alzheimer's disease. Alzheimers Dement 2025; 21:e14582. [PMID: 39985478 PMCID: PMC11846482 DOI: 10.1002/alz.14582] [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: 10/01/2024] [Revised: 12/16/2024] [Accepted: 01/12/2025] [Indexed: 02/24/2025]
Abstract
Hippocampal atrophy on magnetic resonance imaging is an important biomarker in Alzheimer's disease (AD). While hippocampal atrophy was thought to result from tau tangles in AD, different neuropathologies can lead to hippocampal atrophy, especially TAR DNA-binding protein 43 (TDP-43) pathology. In this narrative review, we evaluate existing studies on the relative contribution of tau and TDP-43 pathology to medial temporal lobe (MTL) atrophy. We report a clear association of both tau and TDP-43 neuropathology with MTL atrophy, even after correcting for other neuropathologies. Next, we discuss a potential synergism between tau and TDP-43 and the relative timing of the effects of both neuropathologies. Finally, avenues for future research will be discussed. A better understanding of the interplay between tau and TDP-43 neuropathologies and their effect on atrophy will help with the development of more specific biomarkers for limbic-predominant age-related TDP-43 encephalopathy and pinpointing of the optimal timing for testing anti-tau and anti-TDP-43 treatments in trials. HIGHLIGHTS: Both tau and TAR DNA-binding protein 43 (TDP-43) pathology contribute to medial temporal lobe atrophy. There is a positive association between tau and TDP-43 and potentially a synergism. It is unclear if tau and TDP-43 have an additive or synergistic effect on atrophy. The relative timing of the tau and TDP-43 effects on atrophy remains unclear. Clarifying the interplay between tau and TDP-43 will help improve magnetic resonance imaging biomarkers.
Collapse
Affiliation(s)
| | - Anika Wuestefeld
- Clinical Memory Research Unit, Department of Clinical Sciences MalmöLund UniversityLundSweden
| | - Melissa E. Murray
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
- Department of Laboratory Medicine and PathologyMayo Clinic FloridaJacksonvilleFloridaUSA
| | - William Jagust
- Department of NeuroscienceUniversity of California BerkeleyBerkeleyCaliforniaUSA
- Molecular Biophysics and Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
7
|
Almeida FC, Santos A, Jesus T, Coelho A, Quintas‐Neves M, Gauthreaux K, Mock CN, Kukull WA, Crary JF, Oliveira TG. Lewy body co-pathology in Alzheimer's disease and primary age-related tauopathy contributes to differential neuropathological, cognitive, and brain atrophy patterns. Alzheimers Dement 2025; 21:e14191. [PMID: 39711133 PMCID: PMC11772724 DOI: 10.1002/alz.14191] [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: 03/27/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) co-pathology with Lewy bodies (LB) is frequent and influences clinical manifestations and outcomes. Its significance in primary age-related tauopathy (PART) is unknown. We investigated the influence of LB on cognition and brain atrophy in AD and PART. METHODS We performed a retrospective cohort study in a large sample of autopsied participants with AD neuropathological change (ADNC) with and without LB and PART with and without LB, with corresponding ante mortem magnetic resonance imaging (MRI) data from the National Alzheimer's Coordinating Center dataset. RESULTS LB co-pathology worsened cognitive impairment in both PART and ADNC. On longitudinal follow-up, LB impacted cognitive decline in multiple domains. Additionally, LB influenced brain atrophy on MRI across groups and LB regional staging was different in PART and ADNC, accompanying tauopathy progression. DISCUSSION These results suggest that LB co-pathology is associated with divergent patterns of cognitive impairment, brain atrophy, and regional pathological distribution in PART and AD. HIGHLIGHTS Lewy body (LB) co-pathology is frequent in Alzheimer's disease (AD) with important clinical implications. LB co-pathology is also present in primary age-related tauopathy (PART), but its significance is still understudied. In PART and AD, LB leads to higher cognitive impairment and brain regional atrophy. In PART and AD, LB tends to accompany neurofibrillary tangle progression, suggesting amyloid pathology might be a trigger for regional pathology progression.
Collapse
Affiliation(s)
- Francisco C. Almeida
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of Minho, Campus GualtarBragaPortugal
- ICVS/3B's—PT Government Associate LaboratoryBragaPortugal
- Department of NeuroradiologyCentro Hospitalar Universitário do PortoPortoPortugal
| | - Alexandra Santos
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of Minho, Campus GualtarBragaPortugal
- ICVS/3B's—PT Government Associate LaboratoryBragaPortugal
| | - Tiago Jesus
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of Minho, Campus GualtarBragaPortugal
- ICVS/3B's—PT Government Associate LaboratoryBragaPortugal
- Center AlgoritmiLASI, University of Minho, Campus GualtarBragaPortugal
| | - Ana Coelho
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of Minho, Campus GualtarBragaPortugal
- ICVS/3B's—PT Government Associate LaboratoryBragaPortugal
| | - Miguel Quintas‐Neves
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of Minho, Campus GualtarBragaPortugal
- ICVS/3B's—PT Government Associate LaboratoryBragaPortugal
- Department of NeuroradiologyHospital de Braga, ULS BragaBragaPortugal
| | - Kathryn Gauthreaux
- Department of EpidemiologyNational Alzheimer's Coordinating CenterUniversity of WashingtonSeattleWashingtonUSA
| | - Charles N. Mock
- Department of EpidemiologyNational Alzheimer's Coordinating CenterUniversity of WashingtonSeattleWashingtonUSA
| | - Walter A. Kukull
- Department of EpidemiologyNational Alzheimer's Coordinating CenterUniversity of WashingtonSeattleWashingtonUSA
| | - John F. Crary
- Neuropathology Brain Bank & Research Core, Department of Pathology, Nash Family Department of Neuroscience, Department of Artificial Intelligence & Human Health, Friedman Brain InstituteRonald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of Minho, Campus GualtarBragaPortugal
- ICVS/3B's—PT Government Associate LaboratoryBragaPortugal
- Department of NeuroradiologyHospital de Braga, ULS BragaBragaPortugal
| |
Collapse
|
8
|
Park HH, Kim BH, Leem SH, Park YH, Chung H, Yoo DH, Park I, Nam Y, Kim S, Shin SJ, Moon M. Therapeutic Potential of Traditional Oriental Medicines in Targeting Tau Pathology: Insights from Cell-free and Cell-based Screening. Curr Med Chem 2025; 32:1830-1845. [PMID: 38486385 DOI: 10.2174/0109298673295901240311072440] [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: 12/29/2023] [Revised: 02/08/2024] [Accepted: 02/27/2024] [Indexed: 05/13/2025]
Abstract
BACKGROUND Traditional Oriental Medicines (TOMs) formulated using a variety of medicinal plants have a low risk of side effects. In previous studies, five TOMs, namely Dangguijakyaksan, Hwanglyeonhaedoktang, Ukgansan, Palmijihwanghwan, and Jowiseungchungtang have been commonly used to treat patients with Alzheimer's disease (AD). However, only a few studies have investigated the effects of these five TOMs on tau pathology. OBJECTIVE This study aimed to examine the effect of five TOMs on various tau pathologies, including post-translational modifications, aggregation and deposition, tau-induced neurotoxicity, and tau-induced neuroinflammation. METHODS Immunocytochemistry was used to investigate the hyperphosphorylation of tau induced by okadaic acid. In addition, the thioflavin T assay was used to assess the effects of the TOMs on the inhibition of tau K18 aggregation and the dissociation of tau K18 aggregates. Moreover, a water-soluble tetrazolium-1 assay and a quantitative reverse transcription polymerase chain reaction were used to evaluate the effects of the TOMs on tau-induced neurotoxicity and inflammatory cytokines in HT22 and BV2 cells, respectively. RESULTS The five TOMs investigated in this study significantly reduced okadaic acid-induced tau hyperphosphorylation. Hwanglyeonhaedoktang inhibited the aggregation of tau and promoted the dissociation of tau aggregates. Dangguijakyaksan and Hwanglyeonhaedoktang attenuated tau-induced neurotoxicity in HT22 cells. In addition, Dangguijakyaksan, Hwanglyeonhaedoktang, Ukgansan, and Palmijihwanghwan reduced tauinduced pro-inflammatory cytokine levels in BV2 cells. CONCLUSION Our results suggest that five TOMs are potential therapeutic candidates for tau pathology. In particular, Hwanglyeonhaedoktang showed the greatest efficacy among the five TOMs in cell-free and cell-based screening approaches. These findings suggest that Hwanglyeonhaedoktang is suitable for treating AD patients with tau pathology.
Collapse
Grants
- NRF-2018R1D1A3B07041059, RS-2023-00240010, NRF-2022R1A6A3A13053190, RS-2023-00212388, RS-2023-00273557, NRF-2022R1I1A3052949 Basic Science Research Program of the National Research Foundation of Korea (NRF), by the Ministry of Science, ICT & Future Planning
- HF21C0021, HI23C1263 Korea Health Technology R&D Project through, Korea Health Industry Development Institute (KHIDI), by the Ministry of Health & Welfare, Republic of Korea
Collapse
Affiliation(s)
- Hyun Ha Park
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Byeong-Hyeon Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Seol Hwa Leem
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Hyunju Chung
- Department of Core Research Laboratory, Medical Science Research Institute, Kyung Hee University Hospital Gangdong, Seoul, 05278, Republic of Korea
| | - Doo-Han Yoo
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro Seo-gu, Daejeon, 35365, Republic of Korea
- Department of Occupational Therapy, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Insu Park
- Department of Biomedical Engineering, Konyang University, 158, Gwanjeodong- ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro Seo-gu, Daejeon, 35365, Republic of Korea
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro Seo-gu, Daejeon, 35365, Republic of Korea
| |
Collapse
|
9
|
Nakayama Y, Chambers JK, Takaichi Y, Uchida K. Cytoplasmic aggregation of TDP43 and topographic correlation with tau and α-synuclein accumulation in the rTg4510 mouse model of tauopathy. J Neuropathol Exp Neurol 2024; 83:833-842. [PMID: 38879441 DOI: 10.1093/jnen/nlae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024] Open
Abstract
In patients with TDP43 proteinopathy, phosphorylated TDP43 (p-TDP43) accumulates in the cytoplasm of neurons. The accumulation of p-TDP43 has also been reported in patients with tauopathy and α-synucleinopathy. We investigated spatiotemporal changes in p-TDP43 accumulation in the brains of rTg4510 mice that overexpressed human mutant tau (P301L) and exhibited hyperphosphorylated tau (hp-tau) and phosphorylated αSyn (p-αSyn) accumulation. Immunohistochemically, p-TDP43 aggregates were observed in the cytoplasm of neurons, which increased with age. A significant positive correlation was observed between the number of cells with p-TDP43 aggregates and hp-tau and p-αSyn aggregates. Suppression of the human mutant tau (P301L) expression by doxycycline treatment reduces the accumulation of p-TDP43, hp-tau, and p-αSyn. Proteinase K-resistant p-TDP43 aggregates were found in regions with high hp-tau, and p-αSyn accumulation. Western blotting of the sarkosyl-insoluble fraction revealed bands of monomeric TDP43 and p-TDP43. These results indicate that the accumulation of mouse p-TDP43 is associated with the accumulation of human mutant tau (P301L) in rTg4510 mouse brains. The accumulation of hp-tau and p-αSyn may promote sarkosyl-insoluble p-TDP43 aggregates that are resistant to proteinase K. The synergistic effects of tau, TDP43, and αSyn may be involved in the pathology of proteinopathies, leading to the accumulation of multiple abnormal proteins.
Collapse
Affiliation(s)
- Yutaro Nakayama
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| | - James K Chambers
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| | - Yuta Takaichi
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki Uchida
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
10
|
Josephs KA, Tosakulwong N, Weigand SD, Graff-Radford J, Schwarz CG, Senjem ML, Machulda MM, Kantarci K, Knopman DS, Nguyen A, Reichard RR, Dickson DW, Petersen RC, Lowe VJ, Jack CR, Whitwell JL. Flortaucipir PET uncovers relationships between tau and amyloid-β in primary age-related tauopathy and Alzheimer's disease. Sci Transl Med 2024; 16:eado8076. [PMID: 39047115 PMCID: PMC11423951 DOI: 10.1126/scitranslmed.ado8076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024]
Abstract
[18F]-Flortaucipir positron emission tomography (PET) is considered a good biomarker of Alzheimer's disease. However, it is unknown how flortaucipir is associated with the distribution of tau across brain regions and how these associations are influenced by amyloid-β. It is also unclear whether flortaucipir can detect tau in definite primary age-related tauopathy (PART). We identified 248 individuals at Mayo Clinic who had undergone [18F]-flortaucipir PET during life, had died, and had undergone an autopsy, 239 cases of which also had amyloid-β PET. We assessed nonlinear relationships between flortaucipir uptake in nine medial temporal and cortical regions, Braak tau stage, and Thal amyloid-β phase using generalized additive models. We found that flortaucipir uptake was greater with increasing tau stage in all regions. Increased uptake at low tau stages in medial temporal regions was only observed in cases with a high amyloid-β phase. Flortaucipir uptake linearly increased with the amyloid-β phase in medial temporal and cortical regions. The highest flortaucipir uptake occurred with high Alzheimer's disease neuropathologic change (ADNC) scores, followed by low-intermediate ADNC scores, then PART, with the entorhinal cortex providing the best differentiation between groups. Flortaucipir PET had limited ability to detect PART, and imaging-defined PART did not correspond with pathologically defined PART. In summary, spatial patterns of flortaucipir mirrored the histopathological tau distribution, were influenced by the amyloid-β phase, and were useful for distinguishing different ADNC scores and PART.
Collapse
Affiliation(s)
- Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Nirubol Tosakulwong
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Stephen D Weigand
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Aivi Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - R Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
11
|
Corriveau-Lecavalier N, Botha H, Graff-Radford J, Switzer AR, Przybelski SA, Wiste HJ, Murray ME, Reichard RR, Dickson DW, Nguyen AT, Ramanan VK, McCarter SJ, Boeve BF, Machulda MM, Fields JA, Stricker NH, Nelson PT, Grothe MJ, Knopman DS, Lowe VJ, Petersen RC, Jack CR, Jones DT. Clinical criteria for a limbic-predominant amnestic neurodegenerative syndrome. Brain Commun 2024; 6:fcae183. [PMID: 39021510 PMCID: PMC11251771 DOI: 10.1093/braincomms/fcae183] [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: 03/07/2024] [Revised: 04/16/2024] [Accepted: 05/22/2024] [Indexed: 07/20/2024] Open
Abstract
Predominant limbic degeneration has been associated with various underlying aetiologies and an older age, predominant impairment of episodic memory and slow clinical progression. However, the neurological syndrome associated with predominant limbic degeneration is not defined. This endeavour is critical to distinguish such a syndrome from those originating from neocortical degeneration, which may differ in underlying aetiology, disease course and therapeutic needs. We propose a set of clinical criteria for a limbic-predominant amnestic neurodegenerative syndrome that is highly associated with limbic-predominant age-related TDP-43 encephalopathy but also other pathologic entities. The criteria incorporate core, standard and advanced features, including older age at evaluation, mild clinical syndrome, disproportionate hippocampal atrophy, impaired semantic memory, limbic hypometabolism, absence of neocortical degeneration and low likelihood of neocortical tau, with degrees of certainty (highest, high, moderate and low). We operationalized this set of criteria using clinical, imaging and biomarker data to validate its associations with clinical and pathologic outcomes. We screened autopsied patients from Mayo Clinic and Alzheimer's Disease Neuroimaging Initiative cohorts and applied the criteria to those with an antemortem predominant amnestic syndrome (Mayo, n = 165; Alzheimer's Disease Neuroimaging Initiative, n = 53) and who had Alzheimer's disease neuropathological change, limbic-predominant age-related TDP-43 encephalopathy or both pathologies at autopsy. These neuropathology-defined groups accounted for 35, 37 and 4% of cases in the Mayo cohort, respectively, and 30, 22 and 9% of cases in the Alzheimer's Disease Neuroimaging Initiative cohort, respectively. The criteria effectively categorized these cases, with Alzheimer's disease having the lowest likelihoods, limbic-predominant age-related TDP-43 encephalopathy patients having the highest likelihoods and patients with both pathologies having intermediate likelihoods. A logistic regression using the criteria features as predictors of TDP-43 achieved a balanced accuracy of 74.6% in the Mayo cohort, and out-of-sample predictions in an external cohort achieved a balanced accuracy of 73.3%. Patients with high likelihoods had a milder and slower clinical course and more severe temporo-limbic degeneration compared to those with low likelihoods. Stratifying patients with both Alzheimer's disease neuropathological change and limbic-predominant age-related TDP-43 encephalopathy from the Mayo cohort according to their likelihoods revealed that those with higher likelihoods had more temporo-limbic degeneration and a slower rate of decline and those with lower likelihoods had more lateral temporo-parietal degeneration and a faster rate of decline. The implementation of criteria for a limbic-predominant amnestic neurodegenerative syndrome has implications to disambiguate the different aetiologies of progressive amnestic presentations in older age and guide diagnosis, prognosis, treatment and clinical trials.
Collapse
Affiliation(s)
- Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Aaron R Switzer
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Heather J Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 3224, USA
| | - Robert Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 3224, USA
| | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Vijay K Ramanan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Julie A Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Nikki H Stricker
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Peter T Nelson
- Department of Pathology, University of Kentucky, Lexington, KY 40506, USA
| | - Michel J Grothe
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center, Madrid, Spain
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
12
|
Lavrova A, Pham NTT, Vernon CJ, Carlos AF, Petersen RC, Dickson DW, Lowe VJ, Jack CR, Whitwell JL, Josephs KA. A multimodal clinical diagnostic approach using MRI and 18F-FDG-PET for antemortem diagnosis of TDP-43 in cases with low-intermediate Alzheimer's disease neuropathologic changes and primary age-related tauopathy. J Neurol 2024; 271:4105-4118. [PMID: 38578498 PMCID: PMC11233234 DOI: 10.1007/s00415-024-12312-5] [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: 01/08/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 04/06/2024]
Abstract
OBJECTIVE To evaluate the utility of clinical assessment scales for MRI and 18F-FDG-PET as potential in vivo predictive diagnostic tools for TAR DNA-binding protein of 43 kDa (TDP-43) proteinopathy in cases with low-intermediate Alzheimer's disease neuropathologic changes (ADNC) and primary age-related tauopathy (PART). METHODS We conducted a cross-sectional analysis on patients with antemortem MRI and 18F-FDG-PET scans and postmortem diagnosis of low-intermediate ADNC or PART (Braak stage ≤ III; Thal β-amyloid phase 0-5). We employed visual imaging scales to grade structural changes on MRI and metabolic changes on 18F-FDG-PET and statistically compared demographic and clinicopathological characteristics between TDP-43 positive and negative cases. Independent regression analyses were performed to assess further influences of pathological characteristics on imaging outcomes. Within-reader repeatability and inter-reader reliability were calculated (CI = 0.95). Additional quantitative region-of-interest analyses of MRI gray matter volumes and PET ligand uptake were performed. RESULTS Of the 64 cases in the study, 20 (31%) were TDP-43 ( +), of which 12 (60%) were female. TDP-43 ( +) cases were more likely to have hippocampal sclerosis (HS) (p = 0.014) and moderate-severe medial temporal lobe atrophy on MRI (p = 0.048). TDP-43( +) cases also showed a trend for less parietal atrophy on MRI (p = 0.086) and more medial temporal lobe hypometabolism on 18F-FDG-PET (p = 0.087) than TDP-43( - ) cases. Regression analysis showed an association between medial temporal hypometabolism and HS (p = 0.0113). ICC values for MRI and PET within one reader were 0.75 and 0.91; across two readers were 0.79 and 0.82. The region-of-interest-based analysis confirmed a significant difference between TDP-43( +) and TDP-43( - ) cases for medial temporal lobe gray matter volume on MRI (p = 0.014) and medial temporal metabolism on PET (p = 0.011). CONCLUSION Visual inspection of the medial temporal lobe on MRI and FDG-PET may help to predict TDP-43 status in the context of low-intermediate ADNC and PART.
Collapse
Affiliation(s)
- Anna Lavrova
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Arenn F Carlos
- Department of Neurology, College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA
| | - Ronald C Petersen
- Department of Neurology, College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA
| | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Keith A Josephs
- Department of Neurology, College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
13
|
Pina‐Escudero SD, La Joie R, Spina S, Hwang J, Miller ZA, Huang EJ, Grant H, Mundada NS, Boxer AL, Gorno‐Tempini ML, Rosen HJ, Kramer JH, Miller BL, Seeley WW, Rabinovici GD, Grinberg LT. Comorbid neuropathology and atypical presentation of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12602. [PMID: 39040464 PMCID: PMC11262028 DOI: 10.1002/dad2.12602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/07/2023] [Accepted: 01/19/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) neuropathological changes present with amnestic and nonamnestic (atypical) syndromes. The contribution of comorbid neuropathology as a substratum of atypical expression of AD remains under investigated. METHODS We examined whether atypical AD exhibited increased comorbid neuropathology compared to typical AD and if such neuropathologies contributed to the accelerated clinical decline in atypical AD. RESULTS We examined 60 atypical and 101 typical AD clinicopathological cases. The number of comorbid pathologies was similar between the groups (p = 0.09). Argyrophilic grain disease was associated with atypical presentation (p = 0.008) after accounting for sex, age of onset, and disease duration. Vascular brain injury was more common in typical AD (p = 0.022). Atypical cases had a steeper Mini-Mental Status Examination (MMSE) decline over time (p = 0.033). DISCUSSION Comorbid neuropathological changes are unlikely to contribute to atypical AD presentation and the steeper cognitive decline seen in this cohort. Highlights Autopsy cohort of 60 atypical and 101 typical AD; does comorbid pathology explain atypical presentation?Atypical versus Typical AD: No significant differences in comorbid neuropathologies were found (p = 0.09).Argyrophilic Grain Disease Association: significantly correlates with atypical AD presentations, suggesting a unique neuropathological pattern (p = 0.008).Vascular Brain Injury Prevalence: Vascular brain injury is more common in typical AD than in atypical AD (p = 0.022).Cognitive Decline in Atypical AD: Atypical AD patients experience a steeper cognitive decline measured by MMSE than those with typical AD despite lacking more comorbid neuropathology, highlighting the severity of atypical AD pathogenesis (p = 0.033).
Collapse
Affiliation(s)
- Stefanie D. Pina‐Escudero
- Global Brain Health InstituteUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Renaud La Joie
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Salvatore Spina
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Ji‐Hye Hwang
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Zachary A. Miller
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Eric J. Huang
- Department of PathologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Harli Grant
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Nidhi S. Mundada
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Adam L. Boxer
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Maria Luisa Gorno‐Tempini
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Howard J. Rosen
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Joel H. Kramer
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Bruce L. Miller
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - William W. Seeley
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Gil D. Rabinovici
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lea Tenenholz Grinberg
- Memory and Aging CenterDepartment of NeurologyWeill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of PathologyUniversity of Sao PauloSao PauloSao PauloBrazil
| |
Collapse
|
14
|
Jadhav VS, Stair JG, Eck RJ, Smukowski SN, Currey HN, Toscano LG, Hincks JC, Latimer CS, Valdmanis PN, Kraemer BC, Liachko NF. Transcriptomic evaluation of tau and TDP-43 synergism shows tauopathy predominance and reveals potential modulating targets. Neurobiol Dis 2024; 193:106441. [PMID: 38378122 PMCID: PMC11059213 DOI: 10.1016/j.nbd.2024.106441] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
Alzheimer's disease (AD), the most common aging-associated neurodegenerative dementia disorder, is defined by the presence of amyloid beta (Aβ) and tau aggregates in the brain. However, more than half of patients also exhibit aggregates of the protein TDP-43 as a secondary pathology. The presence of TDP-43 pathology in AD is associated with increased tau neuropathology and worsened clinical outcomes in AD patients. Using C. elegans models of mixed pathology in AD, we have previously shown that TDP-43 specifically synergizes with tau but not Aβ, resulting in enhanced neuronal dysfunction, selective neurodegeneration, and increased accumulation of pathological tau. However, cellular responses to co-morbid tau and TDP-43 preceding neurodegeneration have not been characterized. In this study, we evaluate transcriptomic changes at time-points preceding frank neuronal loss using a C. elegans model of tau and TDP-43 co-expression (tau-TDP-43 Tg). We find significant differential expression and exon usage in genes enriched in multiple pathways including lipid metabolism and lysosomal degradation. We note that early changes in tau-TDP-43 Tg resemble changes with tau alone, but a unique expression signature emerges during aging. We test loss-of-function mutations in a subset of tau and TDP-43 responsive genes, identifying new modifiers of neurotoxicity. Characterizing early cellular responses to tau and TDP-43 co-pathology is critical for understanding protective and pathogenic responses to mixed proteinopathies, and an important step in developing therapeutic strategies protecting against pathological tau and TDP-43 in AD.
Collapse
Affiliation(s)
- Vaishnavi S Jadhav
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA; Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Jade G Stair
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Randall J Eck
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA; Neuroscience Graduate Program, University of Washington, Seattle, WA 98195, USA
| | - Samuel N Smukowski
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Heather N Currey
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Laura Garcia Toscano
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA; Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Joshua C Hincks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Caitlin S Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Mental Illness Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Paul N Valdmanis
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Brian C Kraemer
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA; Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
| | - Nicole F Liachko
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA; Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.
| |
Collapse
|
15
|
Whitwell JL. Atypical clinical variants of Alzheimer's disease: are they really atypical? Front Neurosci 2024; 18:1352822. [PMID: 38482142 PMCID: PMC10933030 DOI: 10.3389/fnins.2024.1352822] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/15/2024] [Indexed: 02/12/2025] Open
Abstract
Alzheimer's disease (AD) is a neuropathological disorder defined by the deposition of the proteins, tau and β-amyloid. Alzheimer's disease is commonly thought of as a disease of the elderly that is associated with episodic memory loss. However, the very first patient described with AD was in her 50's with impairments in multiple cognitive domains. It is now clear that AD can present with multiple different non-amnestic clinical variants which have been labeled as atypical variants of AD. Instead of these variants of AD being considered "atypical," I propose that they provide an excellent disease model of AD and reflect the true clinical heterogeneity of AD. The atypical variants of AD usually have a relatively young age at onset, and they show striking cortical tau deposition on molecular PET imaging which relates strongly with patterns of neurodegeneration and clinical outcomes. In contrast, elderly patients with AD show less tau deposition on PET, and neuroimaging and clinical outcomes are confounded by other age-related pathologies, including TDP-43 and vascular pathology. There is also considerable clinical and anatomical heterogeneity across atypical and young-onset amnestic variants of AD which reflects the fact that AD is a disease that causes impairments in multiple cognitive domains. Future studies should focus on careful characterization of cognitive impairment in AD and consider the full clinical spectrum of AD, including atypical AD, in the design of research studies investigating disease mechanisms in AD and clinical treatment trials, particularly with therapeutics targeting tau.
Collapse
|
16
|
Planche V, Mansencal B, Manjon JV, Meissner WG, Tourdias T, Coupé P. Staging of progressive supranuclear palsy-Richardson syndrome using MRI brain charts for the human lifespan. Brain Commun 2024; 6:fcae055. [PMID: 38444913 PMCID: PMC10914441 DOI: 10.1093/braincomms/fcae055] [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: 11/13/2023] [Revised: 12/22/2023] [Accepted: 02/19/2024] [Indexed: 03/07/2024] Open
Abstract
Brain charts for the human lifespan have been recently proposed to build dynamic models of brain anatomy in normal aging and various neurological conditions. They offer new possibilities to quantify neuroanatomical changes from preclinical stages to death, where longitudinal MRI data are not available. In this study, we used brain charts to model the progression of brain atrophy in progressive supranuclear palsy-Richardson syndrome. We combined multiple datasets (n = 8170 quality controlled MRI of healthy subjects from 22 cohorts covering the entire lifespan, and n = 62 MRI of progressive supranuclear palsy-Richardson syndrome patients from the Four Repeat Tauopathy Neuroimaging Initiative (4RTNI)) to extrapolate lifetime volumetric models of healthy and progressive supranuclear palsy-Richardson syndrome brain structures. We then mapped in time and space the sequential divergence between healthy and progressive supranuclear palsy-Richardson syndrome charts. We found six major consecutive stages of atrophy progression: (i) ventral diencephalon (including subthalamic nuclei, substantia nigra, and red nuclei), (ii) pallidum, (iii) brainstem, striatum and amygdala, (iv) thalamus, (v) frontal lobe, and (vi) occipital lobe. The three structures with the most severe atrophy over time were the thalamus, followed by the pallidum and the brainstem. These results match the neuropathological staging of tauopathy progression in progressive supranuclear palsy-Richardson syndrome, where the pathology is supposed to start in the pallido-nigro-luysian system and spreads rostrally via the striatum and the amygdala to the cerebral cortex, and caudally to the brainstem. This study supports the use of brain charts for the human lifespan to study the progression of neurodegenerative diseases, especially in the absence of specific biomarkers as in PSP.
Collapse
Affiliation(s)
- Vincent Planche
- Institut des Maladies Neurodégénératives, Univ. Bordeaux, CNRS, UMR 5293, F-33000 Bordeaux, France
- Centre Mémoire Ressources Recherches, Service de Neurologie des Maladies Neurodégénératives, Pôle de Neurosciences Cliniques, CHU de Bordeaux, F-33000 Bordeaux, France
| | - Boris Mansencal
- CNRS, Univ. Bordeaux, Bordeaux INP, Laboratoire Bordelais de Recherche en Informatique (LABRI), UMR5800, F-33400 Talence, France
| | - Jose V Manjon
- Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA), Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - Wassilios G Meissner
- Institut des Maladies Neurodégénératives, Univ. Bordeaux, CNRS, UMR 5293, F-33000 Bordeaux, France
- Service de Neurologie des Maladies Neurodégénératives, Réseau NS-Park/FCRIN, CHU Bordeaux, F-33000, Bordeaux, France
- Department of Medicine, Christchurch, and New Zealand Brain Research Institute, Christchurch, 8011, New Zealand
| | - Thomas Tourdias
- Inserm U1215—Neurocentre Magendie, Bordeaux F-33000, France
- Service de Neuroimagerie diagnostique et thérapeutique, CHU de Bordeaux, F-33000 Bordeaux, France
| | - Pierrick Coupé
- CNRS, Univ. Bordeaux, Bordeaux INP, Laboratoire Bordelais de Recherche en Informatique (LABRI), UMR5800, F-33400 Talence, France
| |
Collapse
|
17
|
Phillips JS, Robinson JL, Cousins KAQ, Wolk DA, Lee EB, McMillan CT, Trojanowski JQ, Grossman M, Irwin DJ. Polypathologic Associations with Gray Matter Atrophy in Neurodegenerative Disease. J Neurosci 2024; 44:e0808232023. [PMID: 38050082 PMCID: PMC10860605 DOI: 10.1523/jneurosci.0808-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/01/2023] [Accepted: 10/26/2023] [Indexed: 12/06/2023] Open
Abstract
Mixed pathologies are common in neurodegenerative disease; however, antemortem imaging rarely captures copathologic effects on brain atrophy due to a lack of validated biomarkers for non-Alzheimer's pathologies. We leveraged a dataset comprising antemortem MRI and postmortem histopathology to assess polypathologic associations with atrophy in a clinically heterogeneous sample of 125 human dementia patients (41 female, 84 male) with T1-weighted MRI ≤ 5 years before death and postmortem ordinal ratings of amyloid-[Formula: see text], tau, TDP-43, and [Formula: see text]-synuclein. Regional volumes were related to pathology using linear mixed-effects models; approximately 25% of data were held out for testing. We contrasted a polypathologic model comprising independent factors for each proteinopathy with two alternatives: a model that attributed atrophy entirely to the protein(s) associated with the patient's primary diagnosis and a protein-agnostic model based on the sum of ordinal scores for all pathology types. Model fits were evaluated using log-likelihood and correlations between observed and fitted volume scores. Additionally, we performed exploratory analyses relating atrophy to gliosis, neuronal loss, and angiopathy. The polypathologic model provided superior fits in the training and testing datasets. Tau, TDP-43, and [Formula: see text]-synuclein burden were inversely associated with regional volumes, but amyloid-[Formula: see text] was not. Gliosis and neuronal loss explained residual variance in and mediated the effects of tau, TDP-43, and [Formula: see text]-synuclein on atrophy. Regional brain atrophy reflects not only the primary molecular pathology but also co-occurring proteinopathies; inflammatory immune responses may independently contribute to degeneration. Our findings underscore the importance of antemortem biomarkers for detecting mixed pathology.
Collapse
Affiliation(s)
- Jeffrey S Phillips
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - John L Robinson
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Katheryn A Q Cousins
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - David A Wolk
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Edward B Lee
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Corey T McMillan
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - John Q Trojanowski
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Murray Grossman
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - David J Irwin
- Departments of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
18
|
Guo X, Yan L, Zhang D, Zhao Y. Passive immunotherapy for Alzheimer's disease. Ageing Res Rev 2024; 94:102192. [PMID: 38219962 DOI: 10.1016/j.arr.2024.102192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/03/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease characterized by cognitive impairment with few therapeutic options. Despite many failures in developing AD treatment during the past 20 years, significant advances have been achieved in passive immunotherapy of AD very recently. Here, we review characteristics, clinical trial data, and mechanisms of action for monoclonal antibodies (mAbs) targeting key players in AD pathogenesis, including amyloid-β (Aβ), tau and neuroinflammation modulators. We emphasized the efficacy of lecanemab and donanemab on cognition and amyloid clearance in AD patients in phase III clinical trials and discussed factors that may contribute to the efficacy and side effects of anti-Aβ mAbs. In addition, we provided important information on mAbs targeting tau or inflammatory regulators in clinical trials, and indicated that mAbs against the mid-region of tau or pathogenic tau have therapeutic potential for AD. In conclusion, passive immunotherapy targeting key players in AD pathogenesis offers a promising strategy for effective AD treatment.
Collapse
Affiliation(s)
- Xiaoyi Guo
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Li Yan
- School of Traditional Chinese Medicine, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong 510632, China
| | - Denghong Zhang
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Yingjun Zhao
- Center for Brain Sciences, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China.
| |
Collapse
|
19
|
Agra Almeida Quadros AR, Li Z, Wang X, Ndayambaje IS, Aryal S, Ramesh N, Nolan M, Jayakumar R, Han Y, Stillman H, Aguilar C, Wheeler HJ, Connors T, Lopez-Erauskin J, Baughn MW, Melamed Z, Beccari MS, Olmedo Martínez L, Canori M, Lee CZ, Moran L, Draper I, Kopin AS, Oakley DH, Dickson DW, Cleveland DW, Hyman BT, Das S, Ertekin-Taner N, Lagier-Tourenne C. Cryptic splicing of stathmin-2 and UNC13A mRNAs is a pathological hallmark of TDP-43-associated Alzheimer's disease. Acta Neuropathol 2024; 147:9. [PMID: 38175301 PMCID: PMC10766724 DOI: 10.1007/s00401-023-02655-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024]
Abstract
Nuclear clearance and cytoplasmic accumulations of the RNA-binding protein TDP-43 are pathological hallmarks in almost all patients with amyotrophic lateral sclerosis (ALS) and up to 50% of patients with frontotemporal dementia (FTD) and Alzheimer's disease. In Alzheimer's disease, TDP-43 pathology is predominantly observed in the limbic system and correlates with cognitive decline and reduced hippocampal volume. Disruption of nuclear TDP-43 function leads to abnormal RNA splicing and incorporation of erroneous cryptic exons in numerous transcripts including Stathmin-2 (STMN2, also known as SCG10) and UNC13A, recently reported in tissues from patients with ALS and FTD. Here, we identify both STMN2 and UNC13A cryptic exons in Alzheimer's disease patients, that correlate with TDP-43 pathology burden, but not with amyloid-β or tau deposits. We also demonstrate that processing of the STMN2 pre-mRNA is more sensitive to TDP-43 loss of function than UNC13A. In addition, full-length RNAs encoding STMN2 and UNC13A are suppressed in large RNA-seq datasets generated from Alzheimer's disease post-mortem brain tissue. Collectively, these results open exciting new avenues to use STMN2 and UNC13A as potential therapeutic targets in a broad range of neurodegenerative conditions with TDP-43 proteinopathy including Alzheimer's disease.
Collapse
Affiliation(s)
- Ana Rita Agra Almeida Quadros
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Zhaozhi Li
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xue Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - I Sandra Ndayambaje
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sandeep Aryal
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Nandini Ramesh
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Matthew Nolan
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Rojashree Jayakumar
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yi Han
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hannah Stillman
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Corey Aguilar
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hayden J Wheeler
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Theresa Connors
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jone Lopez-Erauskin
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Michael W Baughn
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Ze'ev Melamed
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Melinda S Beccari
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Laura Olmedo Martínez
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Canori
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Chao-Zong Lee
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura Moran
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Derek H Oakley
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Don W Cleveland
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Bradley T Hyman
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sudeshna Das
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA.
| | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA.
| |
Collapse
|
20
|
Lalive HM, Griffa A, Carlier S, Nasuti M, Di Noto T, Maréchal B, Rouaud O, Allali G. Amnestic Syndrome in Memory Clinics: Similar Morphological Brain Patterns in Older Adults with and without Alzheimer's Disease. J Alzheimers Dis 2024; 100:333-343. [PMID: 38875037 DOI: 10.3233/jad-240026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Background Amnestic syndrome of the hippocampal type (ASHT) in Memory Clinics is a presentation common to Alzheimer's disease (AD). However, ASHT can be found in other neurodegenerative disorders. Objective To compare brain morphometry including hippocampal volumes between amnestic older adults with and without AD pathology and investigate their relationship with memory performance and cerebrospinal fluid (CSF) biomarkers. Methods Brain morphometry of 92 consecutive patients (72.5±6.8 years old; 39% female) with Free and Cued Selective Recall Reminding Test (FCSRT) total recall < 40/48 was assessed with an automated algorithm and compared between AD and non-AD patients, as defined by CSF biomarkers. Results AD and non-AD patients presented comparable brain morphology. Total recall was associated to hippocampal volume irrespectively from AD pathology. Conclusions Brain morphometry, including hippocampal volumes, is similar between AD and non-AD older adults with ASHT evaluated in a Memory Clinic, underlying the importance of using molecular biomarkers for the diagnosis of AD.
Collapse
Affiliation(s)
- Hadrien M Lalive
- Leenaards Memory Centre, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Alessandra Griffa
- Leenaards Memory Centre, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Medical Image Processing Laboratory, Neuro-X Institute, École Polytechnique Fédérale De Lausanne - EPFL, Geneva, Switzerland
| | - Sabrina Carlier
- Leenaards Memory Centre, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Mirco Nasuti
- Leenaards Memory Centre, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Tommaso Di Noto
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Switzerland
- Signal Processing Laboratory - LTS5, École Polytechnique Fédérale de Lausanne - EPFL, Lausanne, Switzerland
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Bénédicte Maréchal
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Switzerland
- Signal Processing Laboratory - LTS5, École Polytechnique Fédérale de Lausanne - EPFL, Lausanne, Switzerland
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Olivier Rouaud
- Leenaards Memory Centre, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Gilles Allali
- Leenaards Memory Centre, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
21
|
Quattrini G, Pini L, Boscolo Galazzo I, Jelescu IO, Jovicich J, Manenti R, Frisoni GB, Marizzoni M, Pizzini FB, Pievani M. Microstructural alterations in the locus coeruleus-entorhinal cortex pathway in Alzheimer's disease and frontotemporal dementia. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12513. [PMID: 38213948 PMCID: PMC10781651 DOI: 10.1002/dad2.12513] [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: 08/25/2023] [Revised: 11/04/2023] [Accepted: 11/20/2023] [Indexed: 01/13/2024]
Abstract
INTRODUCTION We investigated in vivo the microstructural integrity of the pathway connecting the locus coeruleus to the transentorhinal cortex (LC-TEC) in patients with Alzheimer's disease (AD) and frontotemporal dementia (FTD). METHODS Diffusion-weighted MRI scans were collected for 21 AD, 20 behavioral variants of FTD (bvFTD), and 20 controls. Fractional anisotropy (FA), mean, axial, and radial diffusivities (MD, AxD, RD) were computed in the LC-TEC pathway using a normative atlas. Atrophy was assessed using cortical thickness and correlated with microstructural measures. RESULTS We found (i) higher RD in AD than controls; (ii) higher MD, RD, and AxD, and lower FA in bvFTD than controls and AD; and (iii) a negative association between LC-TEC MD, RD, and AxD, and entorhinal cortex (EC) thickness in bvFTD (all p < 0.050). DISCUSSION LC-TEC microstructural alterations are more pronounced in bvFTD than AD, possibly reflecting neurodegeneration secondary to EC atrophy. Highlights Microstructural integrity of LC-TEC pathway is understudied in AD and bvFTD.LC-TEC microstructural alterations are present in both AD and bvFTD.Greater LC-TEC microstructural alterations in bvFTD than AD.LC-TEC microstructural alterations in bvFTD are associated to EC neurodegeneration.
Collapse
Affiliation(s)
- Giulia Quattrini
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE)IRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
| | - Lorenzo Pini
- Padova Neuroscience CenterUniversity of PadovaPadovaItaly
| | | | - Ileana O. Jelescu
- Department of RadiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Jorge Jovicich
- Center of Mind/Brain SciencesUniversity of TrentoRoveretoItaly
| | - Rosa Manenti
- Neuropsychology UnitIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Giovanni B. Frisoni
- Memory Center and LANVIE ‐ Laboratory of Neuroimaging of AgingUniversity Hospitals and University of GenevaGenevaSwitzerland
| | - Moira Marizzoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE)IRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Laboratory of Biological PsychiatryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Francesca B. Pizzini
- Department of Engineering for Innovation MedicineUniversity of VeronaVeronaItaly
| | - Michela Pievani
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE)IRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| |
Collapse
|
22
|
Youssef H, Gatto RG, Pham NTT, Petersen RC, Machulda MM, Reichard RR, Dickson DW, Jack CR, Whitwell JL, Josephs KA. TDP-43 Is Associated with Subiculum and Cornu Ammonis 1 Hippocampal Subfield Atrophy in Primary Age-Related Tauopathy. J Alzheimers Dis 2024; 99:1023-1032. [PMID: 38728190 PMCID: PMC11265226 DOI: 10.3233/jad-240136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
Background TAR DNA binding protein 43 (TDP-43) has been shown to be associated with whole hippocampal atrophy in primary age-related tauopathy (PART). It is currently unknown which subregions of the hippocampus are contributing to TDP-43 associated whole hippocampal atrophy in PART. Objective To identify which specific hippocampal subfield regions are contributing to TDP-43-associated whole hippocampal atrophy in PART. Methods A total of 115 autopsied cases from the Mayo Clinic Alzheimer Disease Research Center, Neurodegenerative Research Group, and the Mayo Clinic Study of Aging were analyzed. All cases underwent antemortem brain volumetric MRI, neuropathological assessment of the distribution of Aβ (Thal phase), and neurofibrillary tangle (Braak stage) to diagnose PART, as well as assessment of TDP-43 presence/absence in the amygdala, hippocampus and beyond. Hippocampal subfield segmentation was performed using FreeSurfer version 7.4.1. Statistical analyses using logistic regression were performed to assess for associations between TDP-43 and hippocampal subfield volumes, accounting for potential confounders. Results TDP-43 positive patients (n = 37, 32%), of which 15/15 were type-α, had significantly smaller whole hippocampal volumes, and smaller volumes of the body and tail of the hippocampus compared to TDP-43 negative patients. Subfield analyses revealed an association between TDP-43 and the molecular layer of hippocampal body and the body of cornu ammonis 1 (CA1), subiculum, and presubiculum regions. There was no association between TDP-43 stage and subfield volumes. Conclusions Whole hippocampal volume loss linked to TDP-43 in PART is mainly due to volume loss occurring in the molecular layer, CA1, subiculum and presubiculum of the hippocampal body.
Collapse
Affiliation(s)
- Hossam Youssef
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Mary M. Machulda
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - R. Ross Reichard
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Dennis W. Dickson
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL, USA
| | | | | | | |
Collapse
|
23
|
Hanseeuw BJ, Jacobs HIL, Schultz AP, Buckley RF, Farrell ME, Guehl NJ, Becker JA, Properzi M, Sanchez JS, Quiroz YT, Vannini P, Sepulcre J, Yang HS, Chhatwal JP, Gatchel J, Marshall GA, Amariglio R, Papp K, Rentz DM, Normandin M, Price JC, Healy BC, El Fakhri G, Sperling RA, Johnson KA. Association of Pathologic and Volumetric Biomarker Changes With Cognitive Decline in Clinically Normal Adults. Neurology 2023; 101:e2533-e2544. [PMID: 37968130 PMCID: PMC10791053 DOI: 10.1212/wnl.0000000000207962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/03/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Hippocampal volume (HV) atrophy is a well-known biomarker of memory impairment. However, compared with β-amyloid (Aβ) and tau imaging, it is less specific for Alzheimer disease (AD) pathology. This lack of specificity could provide indirect information about potential copathologies that cannot be observed in vivo. In this prospective cohort study, we aimed to assess the associations among Aβ, tau, HV, and cognition, measured over a 10-year follow-up period with a special focus on the contributions of HV atrophy to cognition after adjusting for Aβ and tau. METHODS We enrolled 283 older adults without dementia or overt cognitive impairment in the Harvard Aging Brain Study. In this report, we only analyzed data from individuals with available longitudinal imaging and cognition data. Serial MRI (follow-up duration 1.3-7.0 years), neocortical Aβ imaging on Pittsburgh Compound B PET scans (1.9-8.5 years), entorhinal and inferior temporal tau on flortaucipir PET scans (0.8-6.0 years), and the Preclinical Alzheimer Cognitive Composite (3.0-9.8 years) were prospectively collected. We evaluated the longitudinal associations between Aβ, tau, volume, and cognition data and investigated sequential models to test the contribution of each biomarker to cognitive decline. RESULTS We analyzed data from 128 clinically normal older adults, including 72 (56%) women and 56 (44%) men; median age at inclusion was 73 years (range 63-87). Thirty-four participants (27%) exhibited an initial high-Aβ burden on PET imaging. Faster HV atrophy was correlated with faster cognitive decline (R2 = 0.28, p < 0.0001). When comparing all biomarkers, HV slope was associated with cognitive decline independently of Aβ and tau measures, uniquely accounting for 10% of the variance. Altogether, 45% of the variance in cognitive decline was explained by combining the change measures in the different imaging biomarkers. DISCUSSION In older adults, longitudinal hippocampal atrophy is associated with cognitive decline, independently of Aβ or tau, suggesting that non-AD pathologies (e.g., TDP-43, vascular) may contribute to hippocampal-mediated cognitive decline. Serial HV measures, in addition to AD-specific biomarkers, may help evaluate the contribution of non-AD pathologies that cannot be measured otherwise in vivo.
Collapse
Affiliation(s)
- Bernard J Hanseeuw
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Heidi I L Jacobs
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Aaron P Schultz
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Rachel F Buckley
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Michelle E Farrell
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Nicolas J Guehl
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - John A Becker
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Michael Properzi
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Justin S Sanchez
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Yakeel T Quiroz
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Patrizia Vannini
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jorge Sepulcre
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Hyun-Sik Yang
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jasmeer P Chhatwal
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Jennifer Gatchel
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Gad A Marshall
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Rebecca Amariglio
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Kathryn Papp
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Dorene M Rentz
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Marc Normandin
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Julie C Price
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Brian C Healy
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Georges El Fakhri
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Reisa A Sperling
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Keith A Johnson
- From the Department of Radiology (B.J.H., H.I.L.J., N.J.G., J.A.B., J.S.S., J.S., H.-S.Y., M.N., J.C.P., G.E.F., K.A.J.), Massachusetts General Hospital, the Gordon Center for Medical Imaging, Boston; Department of Neurology (B.J.H.), Cliniques Universitaires Saint-Luc, Brussels, Belgium; Faculty of Health, Medicine and Life Sciences (H.I.L.J.), School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, the Netherlands; Department of Neurology (A.P.S., R.F.B., M.E.F., M.P., Y.T.Q., P.V., J.P.C., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Massachusetts General Hospital; Center for Alzheimer Research and Treatment (R.F.B., P.V., G.A.M., R.A., K.P., D.M.R., R.A.S., K.A.J.), Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston; Melbourne School of Psychological Sciences (R.F.B.), University of Melbourne, Australia; and Department of Psychiatry (J.G.), and Department of Biostatistics (B.C.H.), Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
24
|
Mitsunaga S, Fujito N, Nakaoka H, Imazeki R, Nagata E, Inoue I. Detection of APP gene recombinant in human blood plasma. Sci Rep 2023; 13:21703. [PMID: 38066066 PMCID: PMC10709617 DOI: 10.1038/s41598-023-48993-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
The pathogenesis of Alzheimer's disease (AD) is believed to involve the accumulation of amyloid-β in the brain, which is produced by the sequential cleavage of amyloid precursor protein (APP) by β-secretase and γ-secretase. Recently, analysis of genomic DNA and mRNA from postmortem brain neurons has revealed intra-exonic recombinants of APP (gencDNA), which have been implicated in the accumulation of amyloid-β. In this study, we computationally analyzed publicly available sequence data (SRA) using probe sequences we constructed to screen APP gencDNAs. APP gencDNAs were detected in SRAs constructed from both genomic DNA and RNA obtained from the postmortem brain and in the SRA constructed from plasma cell-free mRNA (cf-mRNA). The SRA constructed from plasma cf-mRNA showed a significant difference in the number of APP gencDNA reads between SAD and NCI: the p-value from the Mann-Whitney U test was 5.14 × 10-6. The transcripts were also found in circulating nucleic acids (CNA) from our plasma samples with NGS analysis. These data indicate that transcripts of APP gencDNA can be detected in blood plasma and suggest the possibility of using them as blood biomarkers for Alzheimer's disease.
Collapse
Affiliation(s)
- Shigeki Mitsunaga
- Laboratory of Human Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan.
| | - Naoko Fujito
- Laboratory of Human Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan
- Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, 411-8540, Japan
| | - Hirofumi Nakaoka
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Ryoko Imazeki
- Department of Neurology, Tokai University School of Medicine, Isehara, Japan
| | - Eiichiro Nagata
- Department of Neurology, Tokai University School of Medicine, Isehara, Japan
| | - Ituro Inoue
- Laboratory of Human Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan.
| |
Collapse
|
25
|
Corriveau-Lecavalier N, Botha H, Graff-Radford J, Switzer AR, Przybelski SA, Wiste HJ, Murray ME, Reichard RR, Dickson DW, Nguyen AT, Ramanan VK, McCarter SJ, Boeve BF, Machulda MM, Fields JA, Stricker NH, Nelson PT, Grothe MJ, Knopman DS, Lowe VJ, Petersen RC, Jack CR, Jones DT. A limbic-predominant amnestic neurodegenerative syndrome associated with TDP-43 pathology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.19.23298314. [PMID: 38045300 PMCID: PMC10690340 DOI: 10.1101/2023.11.19.23298314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Limbic-predominant age-related TDP-43 encephalopathy (LATE) is a neuropathologically-defined disease that affects 40% of persons in advanced age, but its associated neurological syndrome is not defined. LATE neuropathological changes (LATE-NC) are frequently comorbid with Alzheimer's disease neuropathologic changes (ADNC). When seen in isolation, LATE-NC have been associated with a predominantly amnestic profile and slow clinical progression. We propose a set of clinical criteria for a limbic-predominant amnestic neurodegenerative syndrome (LANS) that is highly associated with LATE-NC but also other pathologic entities. The LANS criteria incorporate core, standard and advanced features that are measurable in vivo, including older age at evaluation, mild clinical syndrome, disproportionate hippocampal atrophy, impaired semantic memory, limbic hypometabolism, absence of neocortical degenerative patterns and low likelihood of neocortical tau, with degrees of certainty (highest, high, moderate, low). We operationalized this set of criteria using clinical, imaging and biomarker data to validate its associations with clinical and pathologic outcomes. We screened autopsied patients from Mayo Clinic (n = 922) and ADNI (n = 93) cohorts and applied the LANS criteria to those with an antemortem predominant amnestic syndrome (Mayo, n = 165; ADNI, n = 53). ADNC, ADNC/LATE-NC and LATE-NC accounted for 35%, 37% and 4% of cases in the Mayo cohort, respectively, and 30%, 22%, and 9% of cases in the ADNI cohort, respectively. The LANS criteria effectively categorized these cases, with ADNC having the lowest LANS likelihoods, LATE-NC patients having the highest likelihoods, and ADNC/LATE-NC patients having intermediate likelihoods. A logistic regression model using the LANS features as predictors of LATE-NC achieved a balanced accuracy of 74.6% in the Mayo cohort, and out-of-sample predictions in the ADNI cohort achieved a balanced accuracy of 73.3%. Patients with high LANS likelihoods had a milder and slower clinical course and more severe temporo-limbic degeneration compared to those with low likelihoods. Stratifying ADNC/LATE-NC patients from the Mayo cohort according to their LANS likelihood revealed that those with higher likelihoods had more temporo-limbic degeneration and a slower rate of cognitive decline, and those with lower likelihoods had more lateral temporo-parietal degeneration and a faster rate of cognitive decline. The implementation of LANS criteria has implications to disambiguate the different driving etiologies of progressive amnestic presentations in older age and guide prognosis, treatment, and clinical trials. The development of in vivo biomarkers specific to TDP-43 pathology are needed to refine molecular associations between LANS and LATE-NC and precise antemortem diagnoses of LATE.
Collapse
Affiliation(s)
- Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Heather J. Wiste
- Department of Quantitative Health Sciences, Mayo Clinic Rochester, MN, USA
| | | | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN, USA
| | | | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN, USA
| | | | | | | | - Mary M. Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Julie A. Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Nikki H. Stricker
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Peter T. Nelson
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Michel J. Grothe
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center, Madrid, Spain
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Clifford R. Jack
- Department of Neuroscience, Mayo Clinic Jacksonville, FL, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - David T. Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
26
|
Ortega-Cruz D, Iglesias JE, Rabano A, Strange BA. Hippocampal sclerosis of aging at post-mortem is evident on MRI more than a decade prior. Alzheimers Dement 2023; 19:5307-5315. [PMID: 37366342 PMCID: PMC10751387 DOI: 10.1002/alz.13352] [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: 03/09/2023] [Revised: 05/05/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION Hippocampal sclerosis of aging (HS) is an important component of combined dementia neuropathology. However, the temporal evolution of its histologically-defined features is unknown. We investigated pre-mortem longitudinal hippocampal atrophy associated with HS, as well as with other dementia-associated pathologies. METHODS We analyzed hippocampal volumes from magnetic resonance imaging (MRI) segmentations in 64 dementia patients with longitudinal MRI follow-up and post-mortem neuropathological evaluation, including HS assessment in the hippocampal head and body. RESULTS Significant HS-associated hippocampal volume changes were observed throughout the evaluated timespan, up to 11.75 years before death. These changes were independent of age and Alzheimer's disease (AD) neuropathology and were driven specifically by CA1 and subiculum atrophy. AD pathology, but not HS, was associated significantly with the rate of hippocampal atrophy. DISCUSSION HS-associated volume changes are detectable on MRI earlier than 10 years before death. Based on these findings, volumetric cutoffs could be derived for in vivo differentiation between HS and AD. HIGHLIGHTS Hippocampal atrophy was found in HS+ patients earlier than 10 years before death. These early pre-mortem changes were driven by reduced CA1 and subiculum volumes. Rates of hippocampus and subfield volume decline were independent of HS. In contrast, steeper atrophy rates were associated with AD pathology burden. Differentiation between AD and HS could be facilitated based on these MRI findings.
Collapse
Affiliation(s)
- Diana Ortega-Cruz
- Laboratory for Clinical Neuroscience, Center for Biomedical Technology, Universidad Politécnica de Madrid, IdISSC, 28223, Madrid, Spain
- Alzheimer’s Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, 28031, Madrid, Spain
| | - Juan Eugenio Iglesias
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, 02129, Boston, MA, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, 02139, Boston, MA, USA
- Centre for Medical Image Computing, University College London, WC1V 6LJ, London, UK
| | - Alberto Rabano
- Alzheimer’s Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, 28031, Madrid, Spain
| | - Bryan A. Strange
- Laboratory for Clinical Neuroscience, Center for Biomedical Technology, Universidad Politécnica de Madrid, IdISSC, 28223, Madrid, Spain
- Alzheimer’s Disease Research Unit, CIEN Foundation, Queen Sofia Foundation Alzheimer Center, 28031, Madrid, Spain
| |
Collapse
|
27
|
Boxer AL, Sperling R. Accelerating Alzheimer's therapeutic development: The past and future of clinical trials. Cell 2023; 186:4757-4772. [PMID: 37848035 PMCID: PMC10625460 DOI: 10.1016/j.cell.2023.09.023] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/03/2023] [Accepted: 09/22/2023] [Indexed: 10/19/2023]
Abstract
Alzheimer's disease (AD) research has entered a new era with the recent positive phase 3 clinical trials of the anti-Aβ antibodies lecanemab and donanemab. Why did it take 30 years to achieve these successes? Developing potent therapies for reducing fibrillar amyloid was key, as was selection of patients at relatively early stages of disease. Biomarkers of the target pathologies, including amyloid and tau PET, and insights from past trials were also critical to the recent successes. Moving forward, the challenge will be to develop more efficacious therapies with greater efficiency. Novel trial designs, including combination therapies and umbrella and basket protocols, will accelerate clinical development. Better diversity and inclusivity of trial participants are needed, and blood-based biomarkers may help to improve access for medically underserved groups. Incentivizing innovation in both academia and industry through public-private partnerships, collaborative mechanisms, and the creation of new career paths will be critical to build momentum in these exciting times.
Collapse
Affiliation(s)
- Adam L Boxer
- Memory and Aging Center, Department of Neurology, Weill Institute of Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| | - Reisa Sperling
- Center for Alzheimer Research and Treatment, Department of Neurology, MassGeneral Brigham, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Roelofs A. Cerebral atrophy as a cause of aphasia: From Pick to the modern era. Cortex 2023; 165:101-118. [PMID: 37276800 DOI: 10.1016/j.cortex.2023.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 06/07/2023]
Abstract
In his epoch-making monograph, Wernicke (1874) claimed that atrophy of the brain cannot cause aphasia. Refuting this claim, Pick (1892, 1898, 1901, 1904a) documented in increasing detail several cases of aphasia with circumscribed atrophy of the left temporal lobe, frontal lobe, or both, which persuaded Wernicke (1906). To explain why the atrophy is circumscribed and leads to focal symptoms, Pick (1908a) advanced a functional network account. Behavioral, neuroanatomical, and histopathological studies by Dejerine and Sérieux, Fischer, Alzheimer, Altman, Gans, Onari and Spatz, and Stertz further illuminated the clinical syndromes, the exact spatial distributions of the atrophy, the underlying disease, and its laminar specificity. Unaware of these seminal studies, research from the 1970s until now has independently rediscovered all key findings, and also supports Pick's forgotten functional account of the distribution of atrophy and the focal symptoms. His frontal and temporal forms of aphasia foreshadowed what are now called the nonfluent/agrammatic and semantic variants of primary progressive aphasia. Moreover, aphasic symptoms may occur with frontal degeneration (what used to be called "Pick's disease") that yields personality changes and behavioral disturbances, now called the behavioral variant of frontotemporal dementia.
Collapse
Affiliation(s)
- Ardi Roelofs
- Donders Centre for Cognition, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
29
|
Carlos AF, Tosakulwong N, Weigand SD, Senjem ML, Schwarz CG, Knopman DS, Boeve BF, Petersen RC, Nguyen AT, Reichard RR, Dickson DW, Jack CR, Lowe V, Whitwell JL, Josephs KA. TDP-43 pathology effect on volume and flortaucipir uptake in Alzheimer's disease. Alzheimers Dement 2023; 19:2343-2354. [PMID: 36463537 PMCID: PMC10239529 DOI: 10.1002/alz.12878] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/18/2022] [Accepted: 10/21/2022] [Indexed: 12/07/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) patients ≥70 years show smaller medial temporal volumes despite less 18 F-flortaucipir-positron emission tomography (PET) uptake than younger counterparts. We investigated whether TAR DNA-binding protein 43 (TDP-43) was contributing to this volume-uptake mismatch. METHODS Seventy-seven participants with flortaucipir-PET and volumetric magnetic resonance imaging underwent postmortem AD and TDP-43 pathology assessments. Bivariate-response linear regression estimated the effect of age and TDP-43 pathology on volume and/or flortaucipir standardized uptake volume ratios of the hippocampus, amygdala, entorhinal, inferior temporal, and midfrontal cortices. RESULTS Older participants had lower hippocampal volumes and overall flortaucipir uptake. TDP-43-immunoreactivity correlated with reduced medial temporal volumes but was unrelated to flortaucipir uptake. TDP-43 effect size was consistent across the age spectrum. However, at older ages, the cohort mean volumes moved toward those of TDP-43-positives, reflecting the increasing TDP-43 pathology frequency with age. DISCUSSION TDP-43 pathology is a relevant contributor driving the volume-uptake mismatch in older AD participants. HIGHLIGHTS TDP-43 pathology affects medial temporal volume loss but not tau radiotracer uptake. Greater TDP-43 pathology effect is seen in old age due to its increasing frequency. TDP-43 pathology is a relevant driver of the volume-uptake mismatch in old AD patients.
Collapse
Affiliation(s)
- Arenn F. Carlos
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905 USA
| | - Nirubol Tosakulwong
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905 USA
| | - Stephen D. Weigand
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905 USA
| | - Matthew L. Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905 USA
- Department of Information Technology, Mayo Clinic, Rochester, MN, 55905 USA
| | | | | | | | | | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905 USA
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905 USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224 USA
| | | | - Val Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905 USA
| | | | | |
Collapse
|
30
|
Roelofs A. Accounting for word production, comprehension, and repetition in semantic dementia, Alzheimer's dementia, and mild cognitive impairment. BRAIN AND LANGUAGE 2023; 238:105243. [PMID: 36868157 DOI: 10.1016/j.bandl.2023.105243] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 01/27/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
It has been known since Pick (1892, 1904) that word retrieval is commonly impaired in left temporal lobe degeneration. Individuals with semantic dementia (SD), Alzheimer's dementia (AD), and mild cognitive impairment (MCI) present with word retrieval difficulty, while comprehension is less affected and repetition is preserved. Whereas computational models have elucidated performance in poststroke and progressive aphasias, including SD, simulations are lacking for AD and MCI. Here, the WEAVER++/ARC model, which has provided neurocognitive computational accounts of poststroke and progressive aphasias, is extended to AD and MCI. Assuming a loss of activation capacity in semantic memory in SD, AD, and MCI, the simulations showed that severity variation accounts for 99% of the variance in naming, comprehension, and repetition at the group level and 95% at the individual patient level (N = 49). Other plausible assumptions do less well. This supports a unified account of performance in SD, AD, and MCI.
Collapse
Affiliation(s)
- Ardi Roelofs
- Radboud University, Donders Institute for Brain, Cognition and Behaviour, Centre for Cognition, Thomas van Aquinostraat 4, 6525 GD Nijmegen, The Netherlands.
| |
Collapse
|
31
|
Hoenig MC, Drzezga A. Clear-headed into old age: Resilience and resistance against brain aging-A PET imaging perspective. J Neurochem 2023; 164:325-345. [PMID: 35226362 DOI: 10.1111/jnc.15598] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 11/28/2022]
Abstract
With the advances in modern medicine and the adaptation towards healthier lifestyles, the average life expectancy has doubled since the 1930s, with individuals born in the millennium years now carrying an estimated life expectancy of around 100 years. And even though many individuals around the globe manage to age successfully, the prevalence of aging-associated neurodegenerative diseases such as sporadic Alzheimer's disease has never been as high as nowadays. The prevalence of Alzheimer's disease is anticipated to triple by 2050, increasing the societal and economic burden tremendously. Despite all efforts, there is still no available treatment defeating the accelerated aging process as seen in this disease. Yet, given the advances in neuroimaging techniques that are discussed in the current Review article, such as in positron emission tomography (PET) or magnetic resonance imaging (MRI), pivotal insights into the heterogenous effects of aging-associated processes and the contribution of distinct lifestyle and risk factors already have and are still being gathered. In particular, the concepts of resilience (i.e. coping with brain pathology) and resistance (i.e. avoiding brain pathology) have more recently been discussed as they relate to mechanisms that are associated with the prolongation and/or even stop of the progressive brain aging process. Better understanding of the underlying mechanisms of resilience and resistance may one day, hopefully, support the identification of defeating mechanism against accelerating aging.
Collapse
Affiliation(s)
- Merle C Hoenig
- Research Center Juelich, Institute for Neuroscience and Medicine II, Molecular Organization of the Brain, Juelich, Germany.,Department of Nuclear Medicine, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Alexander Drzezga
- Research Center Juelich, Institute for Neuroscience and Medicine II, Molecular Organization of the Brain, Juelich, Germany.,Department of Nuclear Medicine, Faculty of Medicine, University Hospital Cologne, Cologne, Germany.,German Center for Neurodegenerative Diseases, Bonn/Cologne, Germany
| |
Collapse
|
32
|
Du Y, Zhang S, Qiu Q, Zhang J, Fang Y, Zhao L, Wei W, Wang J, Wang J, Li X. The effect of hippocampal radiomic features and functional connectivity on the relationship between hippocampal volume and cognitive function in Alzheimer's disease. J Psychiatr Res 2023; 158:382-391. [PMID: 36646036 DOI: 10.1016/j.jpsychires.2023.01.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Hippocampal volume is associated with cognitive function in Alzheimer's disease (AD). Hippocampal radiomic features and resting-state functional connectivity (rs-FC) are promising biomarkers and correlate with AD pathology. However, few studies have been conducted on how hippocampal biomarkers affect the cognition-structure relationship. Therefore, we aimed to investigate the effects of hippocampal radiomic features and resting-state functional connectivity (rs-FC) on this relationship in AD. We enrolled 70 AD patients and 65 healthy controls (HCs). The FreeSurfer software was used to measure hippocampal volume. We selected hippocampal radiomic features to build a model to distinguish AD patients from HCs and used a seed-based approach to calculate the hippocampal rs-FC. Furthermore, we conducted mediation and moderation analyses to investigate the effect of hippocampal radiomic features and rs-FC on the relationship between hippocampal volume and cognition in AD. The results suggested that hippocampal radiomic features mediated the association between bilateral hippocampal volume and cognition in AD. Additionally, patients with AD showed weaker rs-FC between the bilateral hippocampus and right ventral posterior cingulate cortex and stronger rs-FC between the left hippocampus and left insula than HCs. The rs-FC between the hippocampus and insula moderated the relationship between hippocampal volume and cognition in AD, suggesting that this rs-FC could exacerbate or ameliorate the effects of hippocampal volume on cognition and may be essential in improving cognitive function in AD. Our findings may not only expand existing biological knowledge of the interrelationships among hippocampal biomarkers and cognition but also provide potential targets for treatment strategies for AD.
Collapse
Affiliation(s)
- Yang Du
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Shaowei Zhang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qi Qiu
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jianye Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yuan Fang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Lu Zhao
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Wenjing Wei
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jinghua Wang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jinhong Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Xia Li
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
33
|
Calderón-Garcidueñas L, Torres-Jardón R, Greenough GP, Kulesza R, González-Maciel A, Reynoso-Robles R, García-Alonso G, Chávez-Franco DA, García-Rojas E, Brito-Aguilar R, Silva-Pereyra HG, Ayala A, Stommel EW, Mukherjee PS. Sleep matters: Neurodegeneration spectrum heterogeneity, combustion and friction ultrafine particles, industrial nanoparticle pollution, and sleep disorders-Denial is not an option. Front Neurol 2023; 14:1117695. [PMID: 36923490 PMCID: PMC10010440 DOI: 10.3389/fneur.2023.1117695] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/01/2023] [Indexed: 03/02/2023] Open
Abstract
Sustained exposures to ubiquitous outdoor/indoor fine particulate matter (PM2.5), including combustion and friction ultrafine PM (UFPM) and industrial nanoparticles (NPs) starting in utero, are linked to early pediatric and young adulthood aberrant neural protein accumulation, including hyperphosphorylated tau (p-tau), beta-amyloid (Aβ1 - 42), α-synuclein (α syn) and TAR DNA-binding protein 43 (TDP-43), hallmarks of Alzheimer's (AD), Parkinson's disease (PD), frontotemporal lobar degeneration (FTLD), and amyotrophic lateral sclerosis (ALS). UFPM from anthropogenic and natural sources and NPs enter the brain through the nasal/olfactory pathway, lung, gastrointestinal (GI) tract, skin, and placental barriers. On a global scale, the most important sources of outdoor UFPM are motor traffic emissions. This study focuses on the neuropathology heterogeneity and overlap of AD, PD, FTLD, and ALS in older adults, their similarities with the neuropathology of young, highly exposed urbanites, and their strong link with sleep disorders. Critical information includes how this UFPM and NPs cross all biological barriers, interact with brain soluble proteins and key organelles, and result in the oxidative, endoplasmic reticulum, and mitochondrial stress, neuroinflammation, DNA damage, protein aggregation and misfolding, and faulty complex protein quality control. The brain toxicity of UFPM and NPs makes them powerful candidates for early development and progression of fatal common neurodegenerative diseases, all having sleep disturbances. A detailed residential history, proximity to high-traffic roads, occupational histories, exposures to high-emission sources (i.e., factories, burning pits, forest fires, and airports), indoor PM sources (tobacco, wood burning in winter, cooking fumes, and microplastics in house dust), and consumption of industrial NPs, along with neurocognitive and neuropsychiatric histories, are critical. Environmental pollution is a ubiquitous, early, and cumulative risk factor for neurodegeneration and sleep disorders. Prevention of deadly neurological diseases associated with air pollution should be a public health priority.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- College of Health, The University of Montana, Missoula, MT, United States.,Universidad del Valle de México, Mexico City, Mexico
| | - Ricardo Torres-Jardón
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Glen P Greenough
- Department of Neurology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Randy Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | | | | | | | | | | | | | - Héctor G Silva-Pereyra
- Instituto Potosino de Investigación Científica y Tecnológica A.C., San Luis Potosi, Mexico
| | - Alberto Ayala
- Sacramento Metropolitan Air Quality Management District, Sacramento, CA, United States.,Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV, United States
| | - Elijah W Stommel
- Department of Neurology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Partha S Mukherjee
- Interdisciplinary Statistical Research Unit, Indian Statistical Institute, Kolkata, India
| |
Collapse
|
34
|
Carlos AF, Machulda MM, Rutledge MH, Nguyen AT, Reichard RR, Baker MC, Rademakers R, Dickson DW, Petersen RC, Josephs KA. Comparison of Clinical, Genetic, and Pathologic Features of Limbic and Diffuse Transactive Response DNA-Binding Protein 43 Pathology in Alzheimer's Disease Neuropathologic Spectrum. J Alzheimers Dis 2023; 93:1521-1535. [PMID: 37182869 PMCID: PMC10923399 DOI: 10.3233/jad-221094] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Increasing evidence suggests that TAR DNA-binding protein 43 (TDP-43) pathology in Alzheimer's disease (AD), or AD-TDP, can be diffuse or limbic-predominant. Understanding whether diffuse AD-TDP has genetic, clinical, and pathological features that differ from limbic AD-TDP could have clinical and research implications. OBJECTIVE To better characterize the clinical and pathologic features of diffuse AD-TDP and differentiate it from limbic AD-TDP. METHODS 363 participants from the Mayo Clinic Study of Aging, Alzheimer's Disease Research Center, and Neurodegenerative Research Group with autopsy confirmed AD and TDP-43 pathology were included. All underwent genetic, clinical, neuropsychologic, and neuropathologic evaluations. AD-TDP pathology distribution was assessed using the Josephs 6-stage scale. Stages 1-3 were classified as Limbic, those 4-6 as Diffuse. Multivariable logistic regression was used to identify clinicopathologic features that independently predicted diffuse pathology. RESULTS The cohort was 61% female and old at onset (median: 76 years [IQR:70-82]) and death (median: 88 years [IQR:82-92]). Fifty-four percent were Limbic and 46% Diffuse. Clinically, ∼10-20% increases in odds of being Diffuse associated with 5-year increments in age at onset (p = 0.04), 1-year longer disease duration (p = 0.02), and higher Neuropsychiatric Inventory scores (p = 0.03), while 15-second longer Trailmaking Test-B times (p = 0.02) and higher Block Design Test scores (p = 0.02) independently decreased the odds by ~ 10-15%. There was evidence for association of APOEɛ4 allele with limbic AD-TDP and of TMEM106B rs3173615 C allele with diffuse AD-TDP. Pathologically, widespread amyloid-β plaques (Thal phases: 3-5) decreased the odds of diffuse TDP-43 pathology by 80-90%, while hippocampal sclerosis increased it sixfold (p < 0.001). CONCLUSION Diffuse AD-TDP shows clinicopathologic and genetic features different from limbic AD-TDP.
Collapse
Affiliation(s)
- Arenn F. Carlos
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mary M. Machulda
- Department of Psychology and Psychiatry, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew C. Baker
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rosa Rademakers
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL 32224, USA
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Flanders 2000, Belgium
| | - Dennis W. Dickson
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | |
Collapse
|
35
|
Mitroshina EV, Pakhomov AM, Krivonosov MI, Yarkov RS, Gavrish MS, Shkirin AV, Ivanchenko MV, Vedunova MV. Novel Algorithm of Network Calcium Dynamics Analysis for Studying the Role of Astrocytes in Neuronal Activity in Alzheimer's Disease Models. Int J Mol Sci 2022; 23:ijms232415928. [PMID: 36555569 PMCID: PMC9781291 DOI: 10.3390/ijms232415928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
Accumulated experimental data strongly suggest that astrocytes play an important role in the pathogenesis of neurodegeneration, including Alzheimer's disease (AD). The effect of astrocytes on the calcium activity of neuron-astroglia networks in AD modelling was the object of the present study. We have expanded and improved our approach's capabilities to analyze calcium activity. We have developed a novel algorithm to construct dynamic directed graphs of both astrocytic and neuronal networks. The proposed algorithm allows us not only to identify functional relationships between cells and determine the presence of network activity, but also to characterize the spread of the calcium signal from cell to cell. Our study showed that Alzheimer's astrocytes can change the functional pattern of the calcium activity of healthy nerve cells. When healthy nerve cells were cocultivated with astrocytes treated with Aβ42, activation of calcium signaling was found. When healthy nerve cells were cocultivated with 5xFAD astrocytes, inhibition of calcium signaling was observed. In this regard, it seems relevant to further study astrocytic-neuronal interactions as an important factor in the regulation of the functional activity of brain cells during neurodegenerative processes. The approach to the analysis of streaming imaging data developed by the authors is a promising tool for studying the collective calcium dynamics of nerve cells.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
- Correspondence: ; Tel.: +7-950-604-5137
| | - Alexander M. Pakhomov
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
- Institute of Applied Physics RAS, 46 Ulyanov Street, Nizhny Novgorod 603950, Russia
| | - Mikhail I. Krivonosov
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
- Department of Applied Mathematics, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Roman S. Yarkov
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Maria S. Gavrish
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Alexey V. Shkirin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Vavilova St. 38, Moscow 119991, Russia
- Laser Physics Department, National Research Nuclear University MEPhI, Kashirskoe Sh. 31, Moscow 115409, Russia
| | - Mikhail V. Ivanchenko
- Department of Applied Mathematics, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Maria V. Vedunova
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| |
Collapse
|
36
|
Riku Y, Yoshida M, Iwasaki Y, Sobue G, Katsuno M, Ishigaki S. TDP-43 Proteinopathy and Tauopathy: Do They Have Pathomechanistic Links? Int J Mol Sci 2022; 23:ijms232415755. [PMID: 36555399 PMCID: PMC9779029 DOI: 10.3390/ijms232415755] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Transactivation response DNA binding protein 43 kDa (TDP-43) and tau are major pathological proteins of neurodegenerative disorders, of which neuronal and glial aggregates are pathological hallmarks. Interestingly, accumulating evidence from neuropathological studies has shown that comorbid TDP-43 pathology is observed in a subset of patients with tauopathies, and vice versa. The concomitant pathology often spreads in a disease-specific manner and has morphological characteristics in each primary disorder. The findings from translational studies have suggested that comorbid TDP-43 or tau pathology has clinical impacts and that the comorbid pathology is not a bystander, but a part of the disease process. Shared genetic risk factors or molecular abnormalities between TDP-43 proteinopathies and tauopathies, and direct interactions between TDP-43 and tau aggregates, have been reported. Further investigations to clarify the pathogenetic factors that are shared by a broad spectrum of neurodegenerative disorders will establish key therapeutic targets.
Collapse
Affiliation(s)
- Yuichi Riku
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute 480-1195, Japan
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 744-8550, Japan
- Correspondence: or
| | - Mari Yoshida
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute 480-1195, Japan
| | - Yasushi Iwasaki
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute 480-1195, Japan
| | - Gen Sobue
- Graduate School of Medicine, Aichi Medical University, Nagakute 480-1195, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 744-8550, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya 744-8550, Japan
| | - Shinsuke Ishigaki
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan
| |
Collapse
|
37
|
Gauthreaux K, Mock C, Teylan MA, Culhane JE, Chen YC, Chan KCG, Katsumata Y, Nelson PT, Kukull WA. Symptomatic Profile and Cognitive Performance in Autopsy-Confirmed Limbic-Predominant Age-Related TDP-43 Encephalopathy With Comorbid Alzheimer Disease. J Neuropathol Exp Neurol 2022; 81:975-987. [PMID: 36264254 PMCID: PMC9677237 DOI: 10.1093/jnen/nlac093] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Transactive response DNA-binding protein 43 kDa (TDP-43) proteinopathy is the hallmark of limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC). LATE-NC is a common copathology with Alzheimer disease neuropathologic change (ADNC). Data from the National Alzheimer's Coordinating Center were analyzed to compare clinical features and copathologies of autopsy-confirmed ADNC with versus without comorbid LATE-NC. A total of 735 participants with ADNC alone and 365 with ADNC with LATE-NC were included. Consistent with prior work, brains with LATE-NC had more severe ADNC, more hippocampal sclerosis, and more brain arteriolosclerosis copathologies. Behavioral symptoms and cognitive performance on neuropsychological tests were compared, stratified by ADNC severity (low/intermediate vs high). Participants with ADNC and LATE-NC were older, had higher ADNC burden, and had worse cognitive performance than participants with ADNC alone. In the low/intermediate ADNC strata, participants with comorbid LATE-NC had higher prevalence of behavioral symptoms (apathy, disinhibition, agitation, personality change). They also had worsened performance in episodic memory and language/semantic memory. Differences narrowed in the high ADNC strata, with worsened performance in only episodic memory in the comorbid LATE-NC group. The co-occurrence of LATE-NC with ADNC is associated with a different pattern of behavioral and cognitive performance than ADNC alone, particularly in people with low/intermediate ADNC burden.
Collapse
Affiliation(s)
- Kathryn Gauthreaux
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
| | - Charles Mock
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
| | - Merilee A Teylan
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
| | - Jessica E Culhane
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
| | - Yen-Chi Chen
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Kwun C G Chan
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Division of Neuropathology, Department of Pathology, University of Kentucky, Lexington, Kentucky, USA
| | - Walter A Kukull
- From the Department of Epidemiology, National Alzheimer’s Coordinating Center, University of Washington, Seattle, Washington, USA
| |
Collapse
|
38
|
Blasiak J, Sobczuk P, Pawlowska E, Kaarniranta K. Interplay between aging and other factors of the pathogenesis of age-related macular degeneration. Ageing Res Rev 2022; 81:101735. [PMID: 36113764 DOI: 10.1016/j.arr.2022.101735] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/03/2022] [Accepted: 09/12/2022] [Indexed: 01/31/2023]
Abstract
Age-related macular degeneration (AMD) is a complex eye disease with the retina as the target tissue and aging as per definition the most serious risk factor. However, the retina contains over 60 kinds of cells that form different structures, including the neuroretina and retinal pigment epithelium (RPE) which can age at different rates. Other established or putative AMD risk factors can differentially affect the neuroretina and RPE and can differently interplay with aging of these structures. The occurrence of β-amyloid plaques and increased levels of cholesterol in AMD retinas suggest that AMD may be a syndrome of accelerated brain aging. Therefore, the question about the real meaning of age in AMD is justified. In this review we present and update information on how aging may interplay with some aspects of AMD pathogenesis, such as oxidative stress, amyloid beta formation, circadian rhythm, metabolic aging and cellular senescence. Also, we show how this interplay can be specific for photoreceptors, microglia cells and RPE cells as well as in Bruch's membrane and the choroid. Therefore, the process of aging may differentially affect different retinal structures. As an accurate quantification of biological aging is important for risk stratification and early intervention for age-related diseases, the determination how photoreceptors, microglial and RPE cells age in AMD may be helpful for a precise diagnosis and treatment of this largely untreatable disease.
Collapse
Affiliation(s)
- Janusz Blasiak
- Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland.
| | - Piotr Sobczuk
- Emergency Medicine and Disaster Medicine Department, Medical University of Lodz, Pomorska 251, 92-209 Lodz, Poland; Department of Orthopaedics and Traumatology, Polish Mothers' Memorial Hospital - Research Institute, Rzgowska 281, 93-338 Lodz, Poland
| | - Elzbieta Pawlowska
- Department of Pediatric Dentistry, Medical University of Lodz, Pomorska 251, 92-216 Lodz, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, KYS, P.O. Box 100, FI-70029 Finland
| |
Collapse
|
39
|
Quintas-Neves M, Teylan MA, Morais-Ribeiro R, Almeida F, Mock CN, Kukull WA, Crary JF, Oliveira TG. Divergent magnetic resonance imaging atrophy patterns in Alzheimer's disease and primary age-related tauopathy. Neurobiol Aging 2022; 117:1-11. [PMID: 35640459 DOI: 10.1016/j.neurobiolaging.2022.04.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 10/18/2022]
Abstract
Our study compared brain MRI with neuropathological findings in patients with primary age-related tauopathy (PART) and Alzheimer's disease (AD), while assessing the relationship between brain atrophy and clinical impairment. We analyzed 233 participants: 32 with no plaques ("definite" PART-BRAAK stage higher than 0 and CERAD 0), and 201 cases within the AD spectrum, with 25 with sparse (CERAD 1), 76 with moderate (CERAD 2), and 100 with severe (CERAD 3) degrees of neuritic plaques. Upon correcting for age, sex, and age difference at MRI and death, there were significantly higher levels of atrophy in CERAD 3 compared to CERAD 1-2 and a trend compared to PART (p = 0.06). In the anterior temporal region, there was a trend for higher levels of atrophy in PART compared to Alzheimer's disease spectrum cases with CERAD 1 (p = 0.08). We then assessed the correlation between regional brain atrophy and CDR sum of boxes score for PART and AD, and found that overall cognition deficits are directly correlated with regional atrophy in the AD continuum, but not in definite PART. We further observed correlations between regional brain atrophy with multiple neuropsychological metrics in AD, with PART showing specific correlations between language deficits and anterior temporal atrophy. Overall, these findings support PART as an independent pathologic process from AD.
Collapse
Affiliation(s)
- Miguel Quintas-Neves
- Department of Neuroradiology, Hospital de Braga, Braga, Portugal; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Merilee A Teylan
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA, USA
| | - Rafaela Morais-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Francisco Almeida
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Charles N Mock
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA, USA
| | - Walter A Kukull
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA, USA
| | - John F Crary
- Neuropathology Brain Bank & Research Core, Department of Pathology, Nash Family Department of Neuroscience, Department of Artificial Intelligence & Human Health, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tiago Gil Oliveira
- Department of Neuroradiology, Hospital de Braga, Braga, Portugal; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
40
|
Smirnov DS, Salmon DP, Galasko D, Edland SD, Pizzo DP, Goodwill V, Hiniker A. TDP-43 Pathology Exacerbates Cognitive Decline in Primary Age-Related Tauopathy. Ann Neurol 2022; 92:425-438. [PMID: 35696592 PMCID: PMC9391297 DOI: 10.1002/ana.26438] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Primary age-related tauopathy (PART) refers to tau neurofibrillary tangles restricted largely to the medial temporal lobe in the absence of significant beta-amyloid plaques. PART has been associated with cognitive impairment, but contributions from concomitant limbic age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) are underappreciated. METHODS We compare prevalence of LATE-NC and vascular copathologies in age- and Braak-matched patients with PART (n = 45, Braak stage I-IV, Thal phase 0-2) or early stage Alzheimer disease neuropathologic change (ADNC; n = 51, Braak I-IV, Thal 3-5), and examine their influence on clinical and cognitive decline. RESULTS Concomitant LATE-NC and vascular pathology were equally common, and cognition was equally impaired, in PART (Mini-Mental State Examination [MMSE] = 24.8 ± 6.9) and ADNC (MMSE = 24.2 ± 6.0). Patients with LATE-NC were more impaired than those without LATE-NC on the MMSE (by 5.8 points, 95% confidence interval [CI] = 3.0-8.6), Mattis Dementia Rating Scale (DRS; 17.5 points, 95% CI = 7.1-27.9), Clinical Dementia Rating, sum of boxes scale (CDR-sob; 5.2 points, 95% CI = 2.1-8.2), memory composite (0.8 standard deviations [SD], 95% CI = 0.1-1.6), and language composite (1.1 SD, 95% CI = 0.2-2.0), and more likely to receive a dementia diagnosis (odds ratio = 4.8, 95% CI = 1.5-18.0). Those with vascular pathology performed worse than those without on the DRS (by 10.2 points, 95% CI = 0.1-20.3) and executive composite (1.3 SD, 95% CI = 0.3-2.3). Cognition declined similarly in PART and ADNC over the 5 years preceding death; however, LATE-NC was associated with more rapid decline on the MMSE (β = 1.9, 95% CI = 0.9-3.0), DRS (β = 7.8, 95% CI = 3.4-12.7), CDR-sob (β = 1.9, 95% CI = 0.4-3.7), language composite (β = 0.5 SD, 95% CI = 0.1-0.8), and vascular pathology with more rapid decline on the DRS (β = 5.2, 95% CI = 0.6-10.2). INTERPRETATION LATE-NC, and to a lesser extent vascular copathology, exacerbate cognitive impairment and decline in PART and early stage ADNC. ANN NEUROL 2022;92:425-438.
Collapse
Affiliation(s)
- Denis S. Smirnov
- Department of Neurosciences, University of California, San Diego
| | - David P. Salmon
- Department of Neurosciences, University of California, San Diego
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego
- Veterans Affairs San Diego Healthcare System
| | - Steven D. Edland
- Department of Neurosciences, University of California, San Diego
- School of Public Health, University of California, San Diego
| | - Donald P. Pizzo
- Department of Pathology, University of California, San Diego
| | | | - Annie Hiniker
- Department of Pathology, University of California, San Diego
| |
Collapse
|
41
|
Josephs KA, Weigand SD, Whitwell JL. Characterizing Amyloid-Positive Individuals With Normal Tau PET Levels After 5 Years: An ADNI Study. Neurology 2022; 98:e2282-e2292. [PMID: 35314506 PMCID: PMC9162162 DOI: 10.1212/wnl.0000000000200287] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 02/10/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Individuals with biomarker evidence of β-amyloid (Aβ) deposition are increasingly being enrolled in clinical treatment trials but there is a need to identify markers to predict which of these individuals will also develop tau deposition. We aimed to determine whether Aβ-positive individuals can remain tau-negative for at least 5 years and identify characteristics that could distinguish between these individuals and those who develop high tau within this period. METHODS Tau PET positivity was defined using a Gaussian mixture model with log-transformed standard uptake value ratio values from 7 temporal and medial parietal regions using all participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI) with flortaucipir PET. Tau PET scans were classified as normal if the posterior probability of elevated tau was less than 1%. Aβ PET positivity was defined based on ADNI cutpoints. We identified all Aβ-positive individuals from ADNI who had normal tau PET more than 5 years after their first abnormal Aβ PET (amyloid with low tau [ALT] group) and all Aβ-positive individuals with abnormal tau PET within 5 years (biomarker AD). In a case-control design, logistic regression was used to model the odds of biomarker AD vs ALT accounting for sex, age, APOE ε4 carriership, Aβ Centiloid, and hippocampal volume. RESULTS We identified 45 individuals meeting criteria for ALT and 157 meeting criteria for biomarker AD. The ALT group had a lower proportion of APOE ε4 carriers, lower Aβ Centiloid, larger hippocampal volumes, and more preserved cognition, and were less likely to develop dementia, than the biomarker AD group. APOE ε4, higher Aβ Centiloid, and hippocampal atrophy were independently associated with increased odds of abnormal tau within 5 years. A Centiloid value of 50 effectively discriminated biomarker AD and ALT with 80% sensitivity and specificity. The majority of the ALT participants did not develop dementia throughout the 5-year interval. DISCUSSION Aβ-positive individuals can remain tau-negative for at least 5 years. Baseline characteristics can help identify these ALT individuals who are less likely to develop dementia. Conservative Aβ cutpoints should be utilized for clinical trials to better capture individuals with high risk of developing biomarker AD.
Collapse
Affiliation(s)
- Keith A Josephs
- From the Departments of Neurology (K.A.J.), Health Sciences Research (Division of Biomedical Informatics and Statistics) (S.D.W.), and Radiology (J.W.), Mayo Clinic, Rochester, MN
| | - Stephen D Weigand
- From the Departments of Neurology (K.A.J.), Health Sciences Research (Division of Biomedical Informatics and Statistics) (S.D.W.), and Radiology (J.W.), Mayo Clinic, Rochester, MN
| | - Jennifer L Whitwell
- From the Departments of Neurology (K.A.J.), Health Sciences Research (Division of Biomedical Informatics and Statistics) (S.D.W.), and Radiology (J.W.), Mayo Clinic, Rochester, MN
| |
Collapse
|
42
|
Planche V, Manjon JV, Mansencal B, Lanuza E, Tourdias T, Catheline G, Coupé P. Structural progression of Alzheimer’s disease over decades: the MRI staging scheme. Brain Commun 2022; 4:fcac109. [PMID: 35592489 PMCID: PMC9113086 DOI: 10.1093/braincomms/fcac109] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/10/2022] [Accepted: 04/26/2022] [Indexed: 11/25/2022] Open
Abstract
The chronological progression of brain atrophy over decades, from pre-symptomatic to dementia stages, has never been formally depicted in Alzheimer’s disease. This is mainly due to the lack of cohorts with long enough MRI follow-ups in cognitively unimpaired young participants at baseline. To describe a spatiotemporal atrophy staging of Alzheimer’s disease at the whole-brain level, we built extrapolated lifetime volumetric models of healthy and Alzheimer’s disease brain structures by combining multiple large-scale databases (n = 3512 quality controlled MRI from 9 cohorts of subjects covering the entire lifespan, including 415 MRI from ADNI1, ADNI2 and AIBL for Alzheimer’s disease patients). Then, we validated dynamic models based on cross-sectional data using external longitudinal data. Finally, we assessed the sequential divergence between normal aging and Alzheimer’s disease volumetric trajectories and described the following staging of brain atrophy progression in Alzheimer’s disease: (i) hippocampus and amygdala; (ii) middle temporal gyrus; (iii) entorhinal cortex, parahippocampal cortex and other temporal areas; (iv) striatum and thalamus and (v) middle frontal, cingular, parietal, insular cortices and pallidum. We concluded that this MRI scheme of atrophy progression in Alzheimer’s disease was close but did not entirely overlap with Braak staging of tauopathy, with a ‘reverse chronology’ between limbic and entorhinal stages. Alzheimer’s disease structural progression may be associated with local tau accumulation but may also be related to axonal degeneration in remote sites and other limbic-predominant associated proteinopathies.
Collapse
Affiliation(s)
- Vincent Planche
- Univ. Bordeaux, CNRS, UMR 5293, Institut des Maladies Neurodégénératives, F-33000 Bordeaux, France
- Centre Mémoire Ressources Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, F-33000 Bordeaux, France
| | - José V. Manjon
- Instituto de Aplicaciones de las Tecnologías de la Información y de las Comunicaciones Avanzadas (ITACA), Universitat Politècnica de València, Camino de Vera s/n, 46022, Valencia, Spain
| | - Boris Mansencal
- CNRS, Univ. Bordeaux, Bordeaux INP, LABRI, UMR5800, F-33400 Talence, France
| | - Enrique Lanuza
- Univ. Valencia, Dept. of Cell Biology, Burjassot 46100, Valencia, Spain
| | - Thomas Tourdias
- Inserm U1215 - Neurocentre Magendie, Bordeaux F-33000, France
- Service de Neuroimagerie diagnostique et thérapeutique, CHU de Bordeaux, F-33000 Bordeaux, France
| | - Gwenaëlle Catheline
- Univ. Bordeaux, CNRS, UMR 5287, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, F-33000 Bordeaux, France
| | - Pierrick Coupé
- CNRS, Univ. Bordeaux, Bordeaux INP, LABRI, UMR5800, F-33400 Talence, France
| |
Collapse
|
43
|
Latimer CS, Stair JG, Hincks JC, Currey HN, Bird TD, Keene CD, Kraemer BC, Liachko NF. TDP-43 promotes tau accumulation and selective neurotoxicity in bigenic Caenorhabditis elegans. Dis Model Mech 2022; 15:275149. [PMID: 35178571 PMCID: PMC9066518 DOI: 10.1242/dmm.049323] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/11/2022] [Indexed: 11/20/2022] Open
Abstract
Although amyloid β (Aβ) and tau aggregates define the neuropathology of Alzheimer's disease (AD), TDP-43 has recently emerged as a co-morbid pathology in more than half of patients with AD. Individuals with concomitant Aβ, tau and TDP-43 pathology experience accelerated cognitive decline and worsened brain atrophy, but the molecular mechanisms of TDP-43 neurotoxicity in AD are unknown. Synergistic interactions among Aβ, tau and TDP-43 may be responsible for worsened disease outcomes. To study the biology underlying this process, we have developed new models of protein co-morbidity using the simple animal Caenorhabditis elegans. We demonstrate that TDP-43 specifically enhances tau but not Aβ neurotoxicity, resulting in neuronal dysfunction, pathological tau accumulation and selective neurodegeneration. Furthermore, we find that synergism between tau and TDP-43 is rescued by loss-of-function of the robust tau modifier sut-2. Our results implicate enhanced tau neurotoxicity as the primary driver underlying worsened clinical and neuropathological phenotypes in AD with TDP-43 pathology, and identify cell-type specific sensitivities to co-morbid tau and TDP-43. Determining the relationship between co-morbid TDP-43 and tau is crucial to understand, and ultimately treat, mixed pathology AD.
Collapse
Affiliation(s)
- Caitlin S. Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Jade G. Stair
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Joshua C. Hincks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Heather N. Currey
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Thomas D. Bird
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Department of Neurology, University of Washington, Seattle, WA 98104, USA,Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Brian C. Kraemer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA,Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - Nicole F. Liachko
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA,Author for correspondence ()
| |
Collapse
|
44
|
Krishnadas N, Doré V, Groot C, Lamb F, Bourgeat P, Burnham SC, Huang K, Goh AMY, Masters CL, Villemagne VL, Rowe CC. Mesial temporal tau in amyloid-β-negative cognitively normal older persons. Alzheimers Res Ther 2022; 14:51. [PMID: 35395950 PMCID: PMC8991917 DOI: 10.1186/s13195-022-00993-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/23/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Tau deposition in the mesial temporal lobe (MTL) in the absence of amyloid-β (Aβ-) occurs with aging. The tau PET tracer 18F-MK6240 has low non-specific background binding so is well suited to exploration of early-stage tau deposition. The aim of this study was to investigate the associations between MTL tau, age, hippocampal volume (HV), cognition, and neocortical tau in Aβ- cognitively unimpaired (CU) individuals. METHODS One hundred and ninety-nine Aβ- participants (Centiloid < 25) who were CU underwent 18F-MK6240 PET at age 75 ± 5.2 years. Tau standardized uptake value ratio (SUVR) was estimated in mesial temporal (Me), temporoparietal (Te), and rest of the neocortex (R) regions and four Me sub-regions. Tau SUVR were analyzed as continuous variables and compared between high and low MTL SUVR groups. RESULTS In this cohort with a stable clinical classification of CU for a mean of 5.3 years prior to and at the time of tau PET, MTL tau was visually observed in 9% of the participants and was limited to Braak stages I-II. MTL tau was correlated with age (r = 0.24, p < 0.001). Age contributed to the variance in cognitive scores but MTL tau did not. MTL tau was not greater with subjective memory complaint, nor was there a correlation between MTL tau and Aβ Centiloid value, but high tau was associated with smaller HV. Participants with MTL tau had higher tau SUVR in the neocortex but this was driven by the cerebellar reference region and was not present when using white matter normalization. CONCLUSIONS In an Aβ- CU cohort, tau tracer binding in the mesial temporal lobe was age-related and associated with smaller hippocampi, but not with subjective or objective cognitive impairment.
Collapse
Affiliation(s)
- Natasha Krishnadas
- Florey Department of Neurosciences & Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Molecular Imaging & Therapy, Austin Health, 145 Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, 145 Studley Rd, Heidelberg, VIC, 3084, Australia
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Melbourne, Victoria, Australia
| | - Colin Groot
- Department of Molecular Imaging & Therapy, Austin Health, 145 Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Fiona Lamb
- Department of Molecular Imaging & Therapy, Austin Health, 145 Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Pierrick Bourgeat
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Brisbane, QLD, Australia
| | - Samantha C Burnham
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Melbourne, Victoria, Australia
| | - Kun Huang
- Department of Molecular Imaging & Therapy, Austin Health, 145 Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Anita M Y Goh
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, 3010, Australia
- National Ageing Research Institute, Parkville, VIC, 3052, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience & Mental Health, Parkville, VIC, 3052, Australia
| | | | - Christopher C Rowe
- Florey Department of Neurosciences & Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia.
- Department of Molecular Imaging & Therapy, Austin Health, 145 Studley Rd, Heidelberg, VIC, 3084, Australia.
- Florey Institute of Neuroscience & Mental Health, Parkville, VIC, 3052, Australia.
| |
Collapse
|
45
|
Riku Y, Iwasaki Y, Ishigaki S, Akagi A, Hasegawa M, Nishioka K, Li Y, Riku M, Ikeuchi T, Fujioka Y, Miyahara H, Sone J, Hattori N, Yoshida M, Katsuno M, Sobue G. Motor neuron TDP-43 proteinopathy in progressive supranuclear palsy and corticobasal degeneration. Brain 2022; 145:2769-2784. [PMID: 35274674 DOI: 10.1093/brain/awac091] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/29/2022] [Accepted: 02/15/2022] [Indexed: 11/12/2022] Open
Abstract
Transactive response DNA-binding protein 43 kDa (TDP-43) is mislocalized from the nucleus and aggregates within the cytoplasm of affected neurons in amyotrophic lateral sclerosis (ALS) cases. TDP-43 pathology has also been found in brain tissues under non-ALS conditions, suggesting mechanistic links between TDP-43-related ALS (ALS-TDP) and various neurological disorders. This study aimed to assess TDP-43 pathology in the spinal cord motor neurons of tauopathies. We examined 106 spinal cords from consecutively autopsied cases with progressive supranuclear palsy (PSP, n = 26), corticobasal degeneration (CBD, n = 12), globular glial tauopathy (GGT, n = 5), Alzheimer's disease (AD, n = 21), or Pick disease (PiD, n = 6) and neurologically healthy controls (n = 36). Ten of the PSP cases (38%) and seven of the CBD cases (58%) showed mislocalization and cytoplasmic aggregation of TDP-43 in spinal cord motor neurons, which was prominent in the cervical cord. TDP-43-aggregates were found to be skein-like, round-shaped, granular, or dot-like and contained insoluble C-terminal fragments showing blotting pattern of ALS or frontotemporal lobar degeneration (FTLD). The lower motor neurons also showed cystatin-C aggregates, although Bunina bodies were absent in hematoxylin-eosin staining. The spinal cord TDP-43 pathology was often associated with TDP-43 pathology of the primary motor cortex. Positive correlations were shown between the severities of TDP-43 and 4-repeat (4R)-tau aggregates in the cervical cord. TDP-43 and 4R-tau aggregates burdens positively correlated with microglial burden in anterior horn. TDP-43 pathology of spinal cord motor neuron did not develop in an age-dependent manner and was not found in the AD, PiD, GGT, and control groups. Next, we assessed splicing factor proline/glutamine rich (SFPQ) expression in spinal cord motor neurons; SFPQ is a recently-identified regulator of ALS/FTLD pathogenesis, and it is also reported that interaction between SFPQ and fused-in-sarcoma (FUS) regulates splicing of microtubule-associated protein tau exon 10. Immunofluorescent and proximity-ligation assays revealed altered SFPQ/FUS-interactions in the neuronal nuclei of PSP, CBD, and ALS-TDP cases but not in AD, PiD, and GGT cases. Moreover, SFPQ expression was depleted in neurons containing TDP-43 or 4R-tau aggregates of PSP and CBD cases. Our results indicate that PSP and CBD may have properties of systematic motor neuron TDP-43 proteinopathy, suggesting mechanistic links with ALS-TDP. SFPQ dysfunction, arising from altered interaction with FUS, may be a candidate of the common pathway.
Collapse
Affiliation(s)
- Yuichi Riku
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan.,Department of Neurology, Graduate School of Nagoya University, Aichi, Japan
| | - Yasushi Iwasaki
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Shinsuke Ishigaki
- Department of Neurology, Graduate School of Nagoya University, Aichi, Japan
| | - Akio Akagi
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Masato Hasegawa
- Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kenya Nishioka
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yuanzhe Li
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Miho Riku
- Department of Pathology, Aichi Medical University, Aichi, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yusuke Fujioka
- Department of Neurology, Graduate School of Nagoya University, Aichi, Japan
| | - Hiroaki Miyahara
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Jun Sone
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Mari Yoshida
- Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Masahisa Katsuno
- Department of Neurology, Graduate School of Nagoya University, Aichi, Japan
| | - Gen Sobue
- Department of Neurology, Graduate School of Nagoya University, Aichi, Japan.,Aichi Medical University, Aichi, Japan
| |
Collapse
|
46
|
Picca A, Guerra F, Calvani R, Coelho-Junior HJ, Bucci C, Marzetti E. Circulating extracellular vesicles: friends and foes in neurodegeneration. Neural Regen Res 2022; 17:534-542. [PMID: 34380883 PMCID: PMC8504375 DOI: 10.4103/1673-5374.320972] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/19/2021] [Accepted: 04/21/2021] [Indexed: 12/01/2022] Open
Abstract
Extracellular vesicles have been identified as pivotal mediators of intercellular communication with critical roles in physiological and pathological conditions. Via this route, several molecules (e.g., nucleic acids, proteins, metabolites) can be transferred to proximal and distant targets to convey specific information. Extracellular vesicle-associated cargo molecules have been proposed as markers of several disease conditions for their potential of tracking down the generating cell. Indeed, circulating extracellular vesicles may represent biomarkers of dysfunctional cellular quality control systems especially in conditions characterized by the accrual of intracellular misfolded proteins. Furthermore, the identification of extracellular vesicles as tools for the delivery of nucleic acids or other cargo molecules to diseased tissues makes these circulating shuttles possible targets for therapeutic development. The increasing interest in the study of extracellular vesicles as biomarkers resides mainly in the fact that the identification of peripheral levels of extracellular vesicle-associated proteins might reflect molecular events occurring in hardly accessible tissues, such as the brain, thereby serving as a "brain liquid biopsy". The exploitation of extracellular vesicles for diagnostic and therapeutic purposed might offer unprecedented opportunities to develop personalized approaches. Here, we discuss the bright and dark sides of extracellular vesicles in the setting of two main neurodegenerative diseases (i.e., Parkinson's and Alzheimer's diseases). A special focus will be placed on the possibility of using extracellular vesicles as biomarkers for the two conditions to enable disease tracking and treatment monitoring.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Hélio José Coelho-Junior
- Università Cattolica del Sacro Cuore, Institute of Internal Medicine and Geriatrics, Rome, Italy
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Institute of Internal Medicine and Geriatrics, Rome, Italy
| |
Collapse
|
47
|
Raghavan S, Przybelski SA, Reid RI, Lesnick TG, Ramanan VK, Botha H, Matchett BJ, Murray ME, Reichard RR, Knopman DS, Graff-Radford J, Jones DT, Lowe VJ, Mielke MM, Machulda MM, Petersen RC, Kantarci K, Whitwell JL, Josephs KA, Jack CR, Vemuri P. White matter damage due to vascular, tau, and TDP-43 pathologies and its relevance to cognition. Acta Neuropathol Commun 2022; 10:16. [PMID: 35123591 PMCID: PMC8817561 DOI: 10.1186/s40478-022-01319-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/27/2022] Open
Abstract
Multi-compartment modelling of white matter microstructure using Neurite Orientation Dispersion and Density Imaging (NODDI) can provide information on white matter health through neurite density index and free water measures. We hypothesized that cerebrovascular disease, Alzheimer's disease, and TDP-43 proteinopathy would be associated with distinct NODDI readouts of white matter damage which would be informative for identifying the substrate for cognitive impairment. We identified two independent cohorts with multi-shell diffusion MRI, amyloid and tau PET, and cognitive assessments: specifically, a population-based cohort of 347 elderly randomly sampled from the Olmsted county, Minnesota, population and a clinical research-based cohort of 61 amyloid positive Alzheimer's dementia participants. We observed an increase in free water and decrease in neurite density using NODDI measures in the genu of the corpus callosum associated with vascular risk factors, which we refer to as the vascular white matter component. Tau PET signal reflective of 3R/4R tau deposition was associated with worsening neurite density index in the temporal white matter where we measured parahippocampal cingulum and inferior temporal white matter bundles. Worsening temporal white matter neurite density was associated with (antemortem confirmed) FDG TDP-43 signature. Post-mortem neuropathologic data on a small subset of this sample lend support to our findings. In the community-dwelling cohort where vascular disease was more prevalent, the NODDI vascular white matter component explained variability in global cognition (partial R2 of free water and neurite density = 8.3%) and MMSE performance (8.2%) which was comparable to amyloid PET (7.4% for global cognition and 6.6% for memory). In the AD dementia cohort, tau deposition was the greatest contributor to cognitive performance (9.6%), but there was also a non-trivial contribution of the temporal white matter component (8.5%) to cognitive performance. The differences observed between the two cohorts were reflective of their distinct clinical composition. White matter microstructural damage assessed using advanced diffusion models may add significant value for distinguishing the underlying substrate (whether cerebrovascular disease versus neurodegenerative disease caused by tau deposition or TDP-43 pathology) for cognitive impairment in older adults.
Collapse
Affiliation(s)
| | - Scott A. Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905 USA
| | - Robert I. Reid
- Department of Information Technology, Mayo Clinic, Rochester, MN 55905 USA
| | - Timothy G. Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905 USA
| | | | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN 55905 USA
| | | | | | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905 USA
| | | | | | - David T. Jones
- Department of Neurology, Mayo Clinic, Rochester, MN 55905 USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Michelle M. Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905 USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905 USA
| | - Mary M. Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905 USA
| | | | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Jennifer L. Whitwell
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | | | - Clifford R. Jack
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| |
Collapse
|
48
|
Buciuc M, Martin PR, Tosakulwong N, Murray ME, Petrucelli L, Senjem ML, Spychalla AJ, Knopman DS, Boeve BF, Petersen RC, Parisi JE, Reichard RR, Dickson DW, Jack CR, Whitwell JL, Josephs KA. TDP-43-associated atrophy in brains with and without frontotemporal lobar degeneration. Neuroimage Clin 2022; 34:102954. [PMID: 35168140 PMCID: PMC8850800 DOI: 10.1016/j.nicl.2022.102954] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 11/29/2022]
Abstract
Transactive response DNA-binding protein of ∼43 kDa (TDP-43), a primary pathologic substrate in tau-negative frontotemporal lobar degeneration (FTLD), is also often found in the brains of elderly individuals without FTLD and is a key player in the process of neurodegeneration in brains with and without FTLD. It is unknown how rates and trajectories of TDP-43-associated brain atrophy compare between these two groups. Additionally, non-FTLD TDP-43 inclusions are not homogeneous and can be divided into two morphologic types: type-α and neurofibrillary tangle-associated type-β. Therefore, we explored whether neurodegeneration also varies due to the morphologic type. In this longitudinal retrospective study of 293 patients with 843 MRI scans spanning over ∼10 years, we used a Bayesian hierarchical linear model to quantify similarities and differences between the non-FTLD TDP-43 (type-α/type-β) and FTLD-TDP (n = 68) in both regional volume at various timepoints before death and annualized rate of atrophy. Since Alzheimer's disease (AD) is a frequent co-pathology in non-FTLD TDP-43, we further divided types α/β based on presence/absence of intermediate-high likelihood AD: AD-TDP type-β (n = 90), AD-TDP type-α (n = 104), and Pure-TDP (n = 31, all type-α). FTLD-TDP was associated with faster atrophy rates in the inferior temporal lobe and temporal pole compared to all non-FTLD TDP-43 groups. The atrophy rate in the frontal lobe was modulated by age with younger FTLD-TDP having the fastest rates. Older FTLD-TDP showed a limbic predominant pattern of neurodegeneration. AD-TDP type-α showed faster rates of hippocampal atrophy and smaller volumes of amygdala, temporal pole, and inferior temporal lobe compared to AD-TDP type-β. Pure-TDP was associated with slowest rates and less atrophy in all brain regions. The results suggest that there are differences and similarities in longitudinal brain volume loss between FTLD-TDP and non-FTLD TDP-43. Within FTLD-TDP age plays a role in which brain regions are the most affected. Additionally, brain atrophy regional rates also vary by non-FTLD TDP-43 type.
Collapse
Affiliation(s)
- Marina Buciuc
- Department of Neurology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Peter R Martin
- Department of Quantitative Health Sciences, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Nirubol Tosakulwong
- Department of Quantitative Health Sciences, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA.
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA.
| | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Anthony J Spychalla
- Department of Radiology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - David S Knopman
- Department of Neurology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Ronald C Petersen
- Department of Neurology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Joseph E Parisi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - R Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA.
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Jennifer L Whitwell
- Department of Radiology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Keith A Josephs
- Department of Neurology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| |
Collapse
|
49
|
Chen Y, Hu D, Zhao L, Tang W, Li B. Unraveling metabolic alterations in transgenic mouse model of Alzheimer's disease using MALDI MS imaging with 4-aminocinnoline-3-carboxamide matrix. Anal Chim Acta 2022; 1192:339337. [PMID: 35057932 DOI: 10.1016/j.aca.2021.339337] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 11/01/2022]
Abstract
Revealing the metabolic abnormalities of central and peripheral systems in Alzheimer's disease (AD) mouse model is of paramount importance for understanding AD disease. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) is a powerful label-free technique that has been extensively utilized for the interrogation of spatial changes of various metabolites in neurodegenerative disease. However, technical limitations still exist in MALDI MS, and there is a need to improve the performance of traditional MALDI for a deeper investigation of metabolic alterations in the AD mouse model. In this work, 4-aminocinnoline-3-carboxamide (4-AC) was developed into a novel dual-polarity MALDI matrix. Compared with traditionally used MALDI matrices such as 2,5-dihydroxybenzoic acid (DHB) and 9-aminoacridine (9-AA), 4-AC exhibited superior performance in UV absorption at 355 nm, ion yields, background interference, and vacuum stability, making it an ideal MALDI matrix for comprehensive evaluation of metabolic alteration in the brain and serum of APP/PS1 transgenic mouse model of AD. In total, 93 metabolites exhibited different levels of regional changes in the brain of AD mice as compared to the age-matched controls. Moreover, in the serum of AD mice, 81 altered metabolites distinguishing the AD group from the control were observed by using multivariate statistical analysis. It is expected that the application of the MALDI MSI method developed in this work to visualize the spatio-chemical change of various metabolites may improve our understanding of the etiopathogenesis of AD.
Collapse
Affiliation(s)
- Yanwen Chen
- State Key Laboratory of Natural Medicines and School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Dejun Hu
- State Key Laboratory of Natural Medicines and School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lisha Zhao
- State Key Laboratory of Natural Medicines and School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Weiwei Tang
- State Key Laboratory of Natural Medicines and School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Bin Li
- State Key Laboratory of Natural Medicines and School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
50
|
Watanabe H, Hikida S, Ikeda M, Mori E. OUP accepted manuscript. Brain Commun 2022; 4:fcac015. [PMID: 35686225 PMCID: PMC9171501 DOI: 10.1093/braincomms/fcac015] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/07/2021] [Accepted: 01/31/2022] [Indexed: 11/26/2022] Open
Abstract
Primary progressive aphasia, a neurodegenerative syndrome, presents mainly with language impairment. Both semantic and logopenic variants are fluent variants of primary progressive aphasia. Before the research criteria of primary progressive aphasia were proposed, progressive fluent aphasias, such as progressive anomic aphasia, transcortical sensory aphasia and Wernicke’s aphasia, were reported as classical progressive fluent aphasias seen in Alzheimer’s disease. However, since the research criteria of primary progressive aphasia were established, classical fluent variants (other than semantic and logopenic variants) have been neglected and have not been included in the current classification of primary progressive aphasia. This study aimed to determine whether unclassified fluent variants (other than semantic and logopenic variants) can be manifestations of primary progressive aphasia. This study also reconfirmed the characteristics of classical progressive fluent aphasia, such as progressive anomic aphasia, progressive transcortical sensory aphasia and progressive Wernicke’s aphasia as unclassified fluent variants of primary progressive aphasia, using comparison with the current model of primary progressive aphasia. Twelve consecutive patients with an unclassified fluent variant other than semantic or logopenic variant underwent language, neurological, neuropsychological and neuroimaging (MRI and single-photon emission computed tomography) testing. Based on comprehensive language tests, we redefined the diagnoses as primary progressive anomic aphasia (n = 8), primary progressive transcortical sensory aphasia (n = 3) and primary progressive Wernicke’s aphasia (n = 1). Anomic aphasia was characterized by anomia but preserved repetition and comprehension; transcortical sensory aphasia by relatively preserved repetition but poor word comprehension; and Wernicke’s aphasia by poor repetition and word comprehension. In patients with anomic aphasia, voxel-based morphometry of MRI data revealed cortical atrophy, which was most prominent in the temporoparietal lobes, with no obvious lateralization; in two-thirds of patients with transcortical sensory aphasia and in one patient with Wernicke’s aphasia, it revealed atrophy, predominantly in the left temporoparietal lobe. Statistical analysis of single-photon emission computed tomography using three-dimensional stereotactic surface projections revealed patterns of left-sided hypoperfusion in the majority of patients. The temporal and parietal lobes were involved in all cases; the degree of hypoperfusion was higher in patients with transcortical sensory aphasia or Wernicke’s aphasia than in patients with anomic aphasia. The present study demonstrated the clinical and imaging features of 12 patients with an unclassified fluent variant of primary progressive aphasia, which we redefined as primary progressive anomic aphasia, primary progressive transcortical sensory aphasia and primary progressive Wernicke’s aphasia. Classical fluent variants other than semantic and logopenic variants can be found in primary progressive aphasia.
Collapse
Affiliation(s)
- Hiroyuki Watanabe
- Correspondence to: Hiroyuki Watanabe Department of Behavioral Neurology and Neuropsychiatry Osaka University United Graduate School of Child Development 2-2, Yamadaoka, Suita Osaka 565-0871, Japan E-mail:
| | - Sakura Hikida
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
- Brain Function Center, Nippon Life Hospital, Osaka, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
- Brain Function Center, Nippon Life Hospital, Osaka, Japan
| | - Etsuro Mori
- Department of Behavioral Neurology and Neuropsychiatry, Osaka University United Graduate School of Child Development, Suita, Japan
- Brain Function Center, Nippon Life Hospital, Osaka, Japan
| |
Collapse
|