1
|
Garliyev V, Lyssenko CA, Wiener JP, Praticò D, Lyssenko NN. Very low levels of ABCA7 in the cerebrum and Alzheimer's disease onset between the ages of 60 and 80 independently of APOE. J Neuropathol Exp Neurol 2024; 83:808-821. [PMID: 38900184 DOI: 10.1093/jnen/nlae060] [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: 06/21/2024] Open
Abstract
This cross-sectional study addressed the ABCA7-Alzheimer's disease (AD) association. ABCA7 protein levels were quantified in 3 cerebral regions of brain donors with Braak neurofibrillary tangle (NFT) stages 0-V. Ordinal regression models were implemented to estimate the effect of ABCA7 on stopping in an earlier Braak NFT stage versus progressing to the later stages in 2 prespecified age segments. In the final model, high ABCA7 levels in the parietal cortex increased the odds of remaining cognitively healthy (ie, in stages 0/I) versus experiencing AD onset (ie, progressing to stages II-V) in the 61-80 age segment (OR = 2.87, adj 95% CI = 1.41-7.86, adj P = .007, n = 109), after controlling for APOE and other covariates. No ABCA7-AD association was found in the 81-98 age segment (n = 113). Parietal ABCA7 levels in 61-80-year-old with stages II-V were very low, even significantly lower than in 81-98-year-old with stages II-V. ABCA7 levels in the prefrontal cortex and hippocampus predicted AD onset in the 61-80 age segment after adjustment for APOE. ABCA7 levels were also the lowest in 61-80-year-old with frequent neuritic plaques. Thus, very low ABCA7 levels in the cerebrum are associated with AD onset in the 7th-8th decade of life.
Collapse
Affiliation(s)
- Viktor Garliyev
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Catherine A Lyssenko
- Office of Institutional Research & Analysis, University of Pennsylvania, Philadelphia, PA, United States
| | - Joel P Wiener
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Domenico Praticò
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Nicholas N Lyssenko
- Alzheimer's Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
2
|
Katsumata Y, Fardo DW, Shade LMP, Wu X, Karanth SD, Hohman TJ, Schneider JA, Bennett DA, Farfel JM, Gauthreaux K, Mock C, Kukull WA, Abner EL, Nelson PT. Genetic associations with dementia-related proteinopathy: Application of item response theory. Alzheimers Dement 2024; 20:2906-2921. [PMID: 38460116 PMCID: PMC11032554 DOI: 10.1002/alz.13741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 03/11/2024]
Abstract
INTRODUCTION Although dementia-related proteinopathy has a strong negative impact on public health, and is highly heritable, understanding of the related genetic architecture is incomplete. METHODS We applied multidimensional generalized partial credit modeling (GPCM) to test genetic associations with dementia-related proteinopathies. Data were analyzed to identify candidate single nucleotide variants for the following proteinopathies: Aβ, tau, α-synuclein, and TDP-43. RESULTS Final included data comprised 966 participants with neuropathologic and WGS data. Three continuous latent outcomes were constructed, corresponding to TDP-43-, Aβ/Tau-, and α-synuclein-related neuropathology endophenotype scores. This approach helped validate known genotype/phenotype associations: for example, TMEM106B and GRN were risk alleles for TDP-43 pathology; and GBA for α-synuclein/Lewy bodies. Novel suggestive proteinopathy-linked alleles were also discovered, including several (SDHAF1, TMEM68, and ARHGEF28) with colocalization analyses and/or high degrees of biologic credibility. DISCUSSION A novel methodology using GPCM enabled insights into gene candidates for driving misfolded proteinopathies. HIGHLIGHTS Latent factor scores for proteinopathies were estimated using a generalized partial credit model. The three latent continuous scores corresponded well with proteinopathy severity. Novel genes associated with proteinopathies were identified. Several genes had high degrees of biologic credibility for dementia risk factors.
Collapse
Affiliation(s)
- Yuriko Katsumata
- Department of BiostatisticsUniversity of KentuckyLexingtonKentuckyUSA
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - David W. Fardo
- Department of BiostatisticsUniversity of KentuckyLexingtonKentuckyUSA
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | | | - Xian Wu
- Department of BiostatisticsUniversity of KentuckyLexingtonKentuckyUSA
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Shama D. Karanth
- Department of SurgeryCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
- UF Health Cancer CenterUniversity of FloridaGainesvilleFloridaUSA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Julie A. Schneider
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - David A. Bennett
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Jose M. Farfel
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Kathryn Gauthreaux
- National Alzheimer's Coordinating CenterDepartment of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Charles Mock
- National Alzheimer's Coordinating CenterDepartment of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Walter A. Kukull
- National Alzheimer's Coordinating CenterDepartment of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Erin L. Abner
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of Epidemiology and Environmental HealthUniversity of KentuckyLexingtonKentuckyUSA
| | - Peter T. Nelson
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of PathologyDivision of NeuropathologyUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
3
|
Hanyu H, Koyama Y, Umekida K, Momose T, Watanabe S, Sato T. Factors and brain imaging features associated with cognition in oldest-old patients with Alzheimer-type dementia. J Neurol Sci 2024; 458:122929. [PMID: 38377704 DOI: 10.1016/j.jns.2024.122929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/20/2024] [Accepted: 02/09/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND The underlying pathophysiology of cognitive dysfunction in oldest-old patients with Alzheimer-type dementia (AD) has not been clarified to date. We aimed to determine the factors and brain imaging features associated with cognition in oldest-old patients with AD. METHODS We enrolled 456 consecutive outpatients with probable AD (145 men and 311 women, age range: 51-95 years). Demographic factors, such as education level, disease duration at initial visit, body mass index, comorbidities, frailty, and leisure activity, and brain imaging features, including severity of medial temporal lobe (MTL) atrophy, white matter lesions and infarcts, and frequency of posterior cerebral hypoperfusion were compared among pre-old (≤ 74 years), old (75 to 84 years), and oldest-old (≥ 85 years) subgroups. RESULTS The oldest-old subgroup showed significantly longer disease duration, lower education level, more severe frailty, less leisure activity, worse cognitive impairment, a tendency of slower progression of cognitive decline, greater MTL atrophy, more severe white matter hyperintensities and infarcts, and lower frequency of posterior hypoperfusion than the younger age subgroups. Regarding the brain imaging subtypes, there were significantly more patients with the limbic-predominant subtype and fewer patients with the hippocampal-sparing subtype in the oldest-old AD group than the pre-old AD group. CONCLUSIONS Oldest-old patients with AD show different factors and brain imaging features associated with cognition from pre-old and old patients. Our results are expected to provide useful information towards understanding the pathophysiology of oldest-old patients with AD, and for determining their clinical diagnosis and appropriate management methods.
Collapse
Affiliation(s)
- Haruo Hanyu
- Dementia Research Center, Tokyo General Hospital, Tokyo, Japan; Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan.
| | - Yumi Koyama
- Department of Rehabilitation, Tokyo General Hospital, Tokyo, Japan
| | - Kazuki Umekida
- Department of Rehabilitation, Tokyo General Hospital, Tokyo, Japan
| | | | | | - Tomohiko Sato
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
4
|
Kraner SD, Sompol P, Prateeptrang S, Promkan M, Hongthong S, Thongsopha N, Nelson PT, Norris CM. Development of a monoclonal antibody specific for a calpain-generated ∆48 kDa calcineurin fragment, a marker of distressed astrocytes. J Neurosci Methods 2024; 402:110012. [PMID: 37984591 PMCID: PMC10841921 DOI: 10.1016/j.jneumeth.2023.110012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/23/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Calcineurin (CN) is a Ca2+/calmodulin-dependent protein phosphatase. In healthy tissue, CN exists mainly as a full-length (∼60 kDa) highly-regulated protein phosphatase involved in essential cellular functions. However, in diseased or injured tissue, CN is proteolytically converted to a constitutively active fragment that has been causatively-linked to numerous pathophysiologic processes. These calpain-cleaved CN fragments (∆CN) appear at high levels in human brain at early stages of cognitive decline associated with Alzheimer's disease (AD). NEW METHOD We developed a monoclonal antibody to ∆CN, using an immunizing peptide corresponding to the C-terminal end of the ∆CN fragment. RESULTS We obtained a mouse monoclonal antibody, designated 26A6, that selectively detects ∆CN in Western analysis of calpain-cleaved recombinant human CN. Using this antibody, we screened both pathological and normal human brain sections provided by the University of Kentucky's Alzheimer's Disease Research Center. 26A6 showed low reactivity towards normal brain tissue, but detected astrocytes both surrounding AD amyloid plaques and throughout AD brain tissue. In brain tissue with infarcts, there was considerable concentration of 26A6-positive astrocytes within/around infarcts, suggesting a link with anoxic/ischemia pathways. COMPARISON WITH EXISTING METHOD The results obtained with the new monoclonal are similar to those obtained with a polyclonal we had previously developed. However, the monoclonal is an abundant tool available to the dementia research community. CONCLUSIONS The new monoclonal 26A6 antibody is highly selective for the ∆CN proteolytic fragment and labels a subset of astrocytes, and could be a useful tool for marking insidious brain pathology and identifying novel astrocyte phenotypes.
Collapse
Affiliation(s)
| | - Pradoldej Sompol
- Sanders Brown Center on Aging, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Siriyagon Prateeptrang
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Moltira Promkan
- Sanders Brown Center on Aging, USA; Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Suthida Hongthong
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Napasorn Thongsopha
- Sanders Brown Center on Aging, USA; School of Allied Health Science, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Peter T Nelson
- Sanders Brown Center on Aging, USA; Department of Pathology, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher M Norris
- Sanders Brown Center on Aging, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
5
|
Isik AT, Kaya D, Gokden M. Brain Banking in Dementia Studies. Methods Mol Biol 2024; 2785:287-295. [PMID: 38427200 DOI: 10.1007/978-1-0716-3774-6_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
It is now well-established practice in dementia that one clinical entity may be caused by various neurodegenerative disorders, each with different histopathological findings, whereas neuropathologically confirmed patients may have different, unusual, and atypical clinical manifestations.This inconsistency in dementia patients leads to neuropathological examination of cases, and neuropathological examination seems to be an inevitable part of dementia practice, at least until all clinical entities are properly identified for humans.Additionally, the development of disease-modifying therapies and confirmation of the actual accurate diagnosis of the neurodegenerative disease that the drug is thought to modify or act upon are of great importance for neuropathological evaluation in brain banks.Neuropathological processes coexisting among patients diagnosed with established clinical criteria or international guidelines have provided a new perspective in the context of drug development.Here, we review our routinely used methodology in the context of the brain banking process.
Collapse
Affiliation(s)
- Ahmet Turan Isik
- Unit for Aging Brain and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Derya Kaya
- Unit for Aging Brain and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Murat Gokden
- Division of Neuropathology, Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
6
|
Hiya S, Maldonado-Díaz C, Walker JM, Richardson TE. Cognitive symptoms progress with limbic-predominant age-related TDP-43 encephalopathy stage and co-occurrence with Alzheimer disease. J Neuropathol Exp Neurol 2023; 83:2-10. [PMID: 37966908 PMCID: PMC10746699 DOI: 10.1093/jnen/nlad098] [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: 11/17/2023] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is a neuropathologic entity characterized by transactive response DNA-binding protein of 43-kDa (TDP-43)-immunoreactive inclusions that originate in the amygdala and then progress to the hippocampi and middle frontal gyrus. LATE-NC may mimic Alzheimer disease clinically and often co-occurs with Alzheimer disease neuropathologic change (ADNC). This report focuses on the cognitive effects of isolated and concomitant LATE-NC and ADNC. Cognitive/neuropsychological, neuropathologic, genetic, and demographic variables were analyzed in 28 control, 31 isolated LATE-NC, 244 isolated ADNC, and 172 concurrent LATE-NC/ADNC subjects from the National Alzheimer's Coordinating Center. Cases with LATE-NC and ADNC were significantly older than controls; cases with ADNC had a significantly higher proportion of cases with at least one APOE ε4 allele. Both LATE-NC and ADNC exhibited deleterious effects on overall cognition proportional to their neuropathological stages; concurrent LATE-NC/ADNC exhibited the worst overall cognitive effect. Multivariate logistic regression analysis determined an independent risk of cognitive impairment for progressive LATE-NC stages (OR 1.66; p = 0.0256) and ADNC levels (OR 3.41; p < 0.0001). These data add to the existing knowledge on the clinical consequences of LATE-NC pathology and the growing literature on the effects of multiple concurrent neurodegenerative pathologies.
Collapse
Affiliation(s)
- Satomi Hiya
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Carolina Maldonado-Díaz
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jamie M Walker
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Timothy E Richardson
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
7
|
Toniolo S, Di Lorenzo F, Bernardini S, Mercuri NB, Sancesario GM. Blood-Brain Barrier Dysfunction and Aβ42/40 Ratio Dose-Dependent Modulation with the ApoE Genotype within the ATN Framework. Int J Mol Sci 2023; 24:12151. [PMID: 37569528 PMCID: PMC10418506 DOI: 10.3390/ijms241512151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 08/13/2023] Open
Abstract
The definition of Alzheimer's disease (AD) now considers the presence of the markers of amyloid (A), tau deposition (T), and neurodegeneration (N) essential for diagnosis. AD patients have been reported to have increased blood-brain barrier (BBB) dysfunction, but that has not been tested within the ATN framework so far. As the field is moving towards the use of blood-based biomarkers, the relationship between BBB disruption and AD-specific biomarkers requires considerable attention. Moreover, other factors have been previously implicated in modulating BBB permeability, including age, gender, and ApoE status. A total of 172 cognitively impaired individuals underwent cerebrospinal fluid (CSF) analysis for AD biomarkers, and data on BBB dysfunction, demographics, and ApoE status were collected. Our data showed that there was no difference in BBB dysfunction across different ATN subtypes, and that BBB damage was not correlated with cognitive impairment. However, patients with BBB disruption, if measured with a high Qalb, had low Aβ40 levels. ApoE status did not affect BBB function but had a dose-dependent effect on the Aβ42/40 ratio. These results might highlight the importance of understanding dynamic changes across the BBB in future studies in patients with AD.
Collapse
Affiliation(s)
- Sofia Toniolo
- Cognitive Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3AZ, UK
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy (G.M.S.)
| | - Francesco Di Lorenzo
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy (G.M.S.)
- Non-Invasive Brain Simulation Unit, IRCSS Santa Lucia Foundation, 00179 Rome, Italy
| | - Sergio Bernardini
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy (G.M.S.)
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy (G.M.S.)
| | - Giulia Maria Sancesario
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy (G.M.S.)
- Biobank Unit, IRCSS Santa Lucia Foundation, 00179 Rome, Italy
| |
Collapse
|
8
|
Phongpreecha T, Cholerton B, Bhukari S, Chang AL, De Francesco D, Thuraiappah M, Godrich D, Perna A, Becker MG, Ravindra NG, Espinosa C, Kim Y, Berson E, Mataraso S, Sha SJ, Fox EJ, Montine KS, Baker LD, Craft S, White L, Poston KL, Beecham G, Aghaeepour N, Montine TJ. Prediction of neuropathologic lesions from clinical data. Alzheimers Dement 2023; 19:3005-3018. [PMID: 36681388 PMCID: PMC10359434 DOI: 10.1002/alz.12921] [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] [Received: 07/06/2022] [Revised: 11/15/2022] [Accepted: 12/12/2022] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Post-mortem analysis provides definitive diagnoses of neurodegenerative diseases; however, only a few can be diagnosed during life. METHODS This study employed statistical tools and machine learning to predict 17 neuropathologic lesions from a cohort of 6518 individuals using 381 clinical features (Table S1). The multisite data allowed validation of the model's robustness by splitting train/test sets by clinical sites. A similar study was performed for predicting Alzheimer's disease (AD) neuropathologic change without specific comorbidities. RESULTS Prediction results show high performance for certain lesions that match or exceed that of research annotation. Neurodegenerative comorbidities in addition to AD neuropathologic change resulted in compounded, but disproportionate, effects across cognitive domains as the comorbidity number increased. DISCUSSION Certain clinical features could be strongly associated with multiple neurodegenerative diseases, others were lesion-specific, and some were divergent between lesions. Our approach could benefit clinical research, and genetic and biomarker research by enriching cohorts for desired lesions.
Collapse
Affiliation(s)
- Thanaphong Phongpreecha
- Department of Pathology, Stanford University 300 Pasteur Drive Medicine Lane Building L235 Stanford, CA 94305 USA
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University 300 Pasteur Drive, Room H3580 MC 5640 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
| | - Brenna Cholerton
- Department of Pathology, Stanford University 300 Pasteur Drive Medicine Lane Building L235 Stanford, CA 94305 USA
| | - Syed Bhukari
- Department of Pathology, Stanford University 300 Pasteur Drive Medicine Lane Building L235 Stanford, CA 94305 USA
| | - Alan L. Chang
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University 300 Pasteur Drive, Room H3580 MC 5640 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
- Department of Pediatrics, Stanford University 453 Quarry Road MC 5660 Palo Alto, CA 94304 USA
| | - Davide De Francesco
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University 300 Pasteur Drive, Room H3580 MC 5640 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
- Department of Pediatrics, Stanford University 453 Quarry Road MC 5660 Palo Alto, CA 94304 USA
| | - Melan Thuraiappah
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University 300 Pasteur Drive, Room H3580 MC 5640 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
- Department of Pediatrics, Stanford University 453 Quarry Road MC 5660 Palo Alto, CA 94304 USA
| | - Dana Godrich
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami 1501 NW 10 Ave, Miami, Florida 33136 USA
| | - Amalia Perna
- Department of Pathology, Stanford University 300 Pasteur Drive Medicine Lane Building L235 Stanford, CA 94305 USA
| | - Martin G. Becker
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University 300 Pasteur Drive, Room H3580 MC 5640 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
- Department of Pediatrics, Stanford University 453 Quarry Road MC 5660 Palo Alto, CA 94304 USA
| | - Neal G. Ravindra
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University 300 Pasteur Drive, Room H3580 MC 5640 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
- Department of Pediatrics, Stanford University 453 Quarry Road MC 5660 Palo Alto, CA 94304 USA
| | - Camilo Espinosa
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University 300 Pasteur Drive, Room H3580 MC 5640 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
- Department of Pediatrics, Stanford University 453 Quarry Road MC 5660 Palo Alto, CA 94304 USA
| | - Yeasul Kim
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University 300 Pasteur Drive, Room H3580 MC 5640 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
- Department of Pediatrics, Stanford University 453 Quarry Road MC 5660 Palo Alto, CA 94304 USA
| | - Eloise Berson
- Department of Pathology, Stanford University 300 Pasteur Drive Medicine Lane Building L235 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
- Department of Pediatrics, Stanford University 453 Quarry Road MC 5660 Palo Alto, CA 94304 USA
| | - Samson Mataraso
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University 300 Pasteur Drive, Room H3580 MC 5640 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
- Department of Pediatrics, Stanford University 453 Quarry Road MC 5660 Palo Alto, CA 94304 USA
| | - Sharon J. Sha
- Department of Neurology & Neurological Sciences, Stanford University 213 Quarry Road, MC 5979 Palo Alto, CA 94304 USA
| | - Edward J. Fox
- Department of Pathology, Stanford University 300 Pasteur Drive Medicine Lane Building L235 Stanford, CA 94305 USA
| | - Kathleen S. Montine
- Department of Pathology, Stanford University 300 Pasteur Drive Medicine Lane Building L235 Stanford, CA 94305 USA
| | - Laura D. Baker
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine 475 Vine Street, Winston-Salem, NC 27101 USA
| | - Suzanne Craft
- Department of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine 475 Vine Street, Winston-Salem, NC 27101 USA
| | - Lon White
- Pacific Health Research and Education Institute, Hawaii 3375 Koapaka Street, I-540, Honolulu, HI 96819 USA
| | - Kathleen L. Poston
- Department of Neurology & Neurological Sciences, Stanford University 213 Quarry Road, MC 5979 Palo Alto, CA 94304 USA
| | - Gary Beecham
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami 1501 NW 10 Ave, Miami, Florida 33136 USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University 300 Pasteur Drive, Room H3580 MC 5640 Stanford, CA 94305 USA
- Department of Biomedical Data Science, Stanford University 1265 Welch Road MC5464 MSOB West Wing, Third Floor Stanford, CA 94305 USA
- Department of Pediatrics, Stanford University 453 Quarry Road MC 5660 Palo Alto, CA 94304 USA
| | - Thomas J. Montine
- Department of Pathology, Stanford University 300 Pasteur Drive Medicine Lane Building L235 Stanford, CA 94305 USA
| |
Collapse
|
9
|
Maioli H, Mittenzwei R, Shofer JB, Scherpelz KP, Marshall D, Nolan AL, Nelson PT, Keene CD, Latimer CS. Performance of a condensed protocol to assess limbic-predominant age-related TDP-43 encephalopathy neuropathologic change. J Neuropathol Exp Neurol 2023; 82:611-619. [PMID: 37195467 PMCID: PMC10280345 DOI: 10.1093/jnen/nlad035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is a dementia-related proteinopathy common in the elderly population. LATE-NC stages 2 or 3 are consistently associated with cognitive impairment. A condensed protocol (CP) for the assessment of Alzheimer disease neuropathologic change and other disorders associated with cognitive impairment, recommended sampling of small brain portions from specific neuroanatomic regions that were consolidated, resulting in significant cost reduction. Formal evaluation of the CP for LATE-NC staging was not previously performed. Here, we determined the ability of the CP to identify LATE-NC stages 2 or 3. Forty brains donated to the University of Washington BioRepository and Integrated Neuropathology laboratory with known LATE-NC status were resampled. Slides containing brain regions required for LATE-NC staging were immunostained for phospho-TDP-43 and reviewed by 6 neuropathologists blinded to original LATE-NC diagnosis. Overall group performance distinguishing between LATE-NC stages 0-1 and 2-3 was 85% (confidence interval [CI]: 75%-92%). We also used the CP to evaluate LATE-NC in a hospital autopsy cohort, in which LATE-NC was more common in individuals with a history of cognitive impairment, older age, and/or comorbid hippocampal sclerosis. This study shows that the CP can effectively discriminate higher stages of LATE-NC from low or no LATE-NC and that it can be successfully applied in clinical practice using a single tissue block and immunostain.
Collapse
Affiliation(s)
- Heather Maioli
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Rhonda Mittenzwei
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Jane B Shofer
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
- Mental Illness Research, Education, and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, Washington, USA
| | - Kathryn P Scherpelz
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Desiree Marshall
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Amber L Nolan
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Peter T Nelson
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - C Dirk Keene
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Caitlin S Latimer
- Division of Neuropathology, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
10
|
Jackson KL, Luo J, Willroth EC, Ong AD, James BD, Bennett DA, Wilson R, Mroczek DK, Graham EK. Associations Between Loneliness and Cognitive Resilience to Neuropathology in Older Adults. J Gerontol B Psychol Sci Soc Sci 2023; 78:939-947. [PMID: 36789449 PMCID: PMC10214654 DOI: 10.1093/geronb/gbad023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Indexed: 02/16/2023] Open
Abstract
OBJECTIVES Loneliness in the aging population is associated with decreased cognitive function and increased neuropathology; less is understood about the association of loneliness and cognitive resilience (CR), defined as the discordance between a person's actual and expected cognition given their neuropathology. Here we assess the effect of loneliness and change in loneliness on CR at end of life and across older adulthood. METHODS Data were combined from 2 longitudinal studies of older adults. CR proximate to death (CRlast_level) and across time (CRslope) was obtained by independently regressing global cognition and change in cognition onto multiple neuropathology indicators and extracting the resulting residuals. We used a series of simple linear regression models to assess the effect of loneliness level and change on CRlast_level and CRslope. RESULTS Higher baseline loneliness was associated with lower CRlast_level (β = -0.11, 95% confidence interval [95% CI; -0.18, -0.04], p < .01); higher baseline loneliness and increasing loneliness over time was associated with lower CRslope (β = -0.13, 95% CI [-0.22, -0.05], p < .01 and β = -0.12, 95% CI [-0.20, -0.04], p < .01, respectively). Results were robust to covariate inclusion and independent of objective social isolation. DISCUSSION Higher and increasing loneliness was associated with lower CR in the face of neuropathology. These results suggest that some individuals are less resilient to the accumulation of neuropathology than others, and experiencing high/increasing loneliness is a key factor putting some at risk. Interventions aimed at optimizing cognitive function across older adults should include loneliness reduction as a potential area of focus.
Collapse
Affiliation(s)
- Kathryn L Jackson
- Department of Medical Social Sciences, Northwestern University, Chicago, Illinois, USA
| | - Jing Luo
- Department of Medical Social Sciences, Northwestern University, Chicago, Illinois, USA
| | - Emily C Willroth
- Department of Psychological and Brain Sciences, Washington University in St. Louis, Missouri, USA
| | - Anthony D Ong
- Department of Psychology, Cornell University, Ithaca, New York, USA
| | - Bryan D James
- Rush Alzheimer’s Disease Center, RUSH University Medical Center, Chicago, Illinois, USA
- Department of Internal Medicine, RUSH Medical Center, Chicago, Illinois, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, RUSH University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, RUSH Medical Center, Chicago, Illinois, USA
| | - Robert Wilson
- Rush Alzheimer’s Disease Center, RUSH University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, RUSH Medical Center, Chicago, Illinois, USA
- Department of Psychiatry and Behavioral Sciences, RUSH Medical Center, Chicago, Illinois, USA
| | - Daniel K Mroczek
- Department of Medical Social Sciences, Northwestern University, Chicago, Illinois, USA
- Department of Psychology, Northwestern University, Chicago, Illinois, USA
| | - Eileen K Graham
- Department of Medical Social Sciences, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
11
|
Wang M, Tang G, Zhou C, Guo H, Hu Z, Hu Q, Li G. Revisiting the intersection of microglial activation and neuroinflammation in Alzheimer's disease from the perspective of ferroptosis. Chem Biol Interact 2023; 375:110387. [PMID: 36758888 DOI: 10.1016/j.cbi.2023.110387] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/12/2023] [Accepted: 02/05/2023] [Indexed: 02/10/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by chronic neuroinflammation with amyloid beta-protein deposition and hyperphosphorylated tau protein. The typical clinical manifestation of AD is progressive memory impairment, and AD is considered a multifactorial disease with various etiologies (genetic factors, aging, lifestyle, etc.) and complicated pathophysiological processes. Previous research identified that neuroinflammation and typical microglial activation are significant mechanisms underlying AD, resulting in dysfunction of the nervous system and progression of the disease. Ferroptosis is a novel modality involved in this process. As an iron-dependent form of cell death, ferroptosis, characterized by iron accumulation, lipid peroxidation, and irreversible plasma membrane disruption, promotes AD by accelerating neuronal dysfunction and abnormal microglial activation. In this case, disturbances in brain iron homeostasis and neuronal ferroptosis aggravate neuroinflammation and lead to the abnormal activation of microglia. Abnormally activated microglia release various pro-inflammatory factors that aggravate the dysregulation of iron homeostasis and neuroinflammation, forming a vicious cycle. In this review, we first introduce ferroptosis, microglia, AD, and their relationship. Second, we discuss the nonnegligible role of ferroptosis in the abnormal microglial activation involved in the chronic neuroinflammation of AD to provide new ideas for the identification of potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Miaomiao Wang
- Queen Mary School, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Gan Tang
- Queen Mary School, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Congfa Zhou
- Department of Anatomy, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Hongmin Guo
- Department of Physiology, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Zihui Hu
- Department of Physiology, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Qixing Hu
- Department of Physiology, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China
| | - Guilin Li
- Department of Physiology, Medical School of Nanchang University, 461 Bayi Road, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
12
|
Iwase T, Yoshida M, Hashizume Y, Inagaki T, Iwasaki Y. Severe cerebrovascular pathology of the first supercentenarian to be autopsied in the world. Neuropathology 2023; 43:181-189. [PMID: 36321363 DOI: 10.1111/neup.12874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 04/04/2023]
Abstract
We report on a 116-year-old Japanese woman who was the first officially documented supercentenarian to be autopsied in the world. She lived a remarkably healthy life until suffering cerebral infarction at 109 years of age. She became Japan's oldest person at 113 years and died in 1995 from colon cancer at 116 years 175 days. Her medical records show the delayed onset of stroke, cancer, dementia, and heart disease and the importance of appropriate medical treatment and intensive dedicated care provided during the last stage of her life. She was the longest-lived person in Japan for 21 years from 1993 until 2014. The neuropathological findings of her autopsied brain were briefly reported in the Japanese literature in 1997. In this study, we reinvestigated her brain and spinal cord in more detail. Severe cerebrovascular lesions and cervical spondylotic myelopathy were found to be the main causes of her disability. Although the density of senile plaques was relatively high, the distribution of neurofibrillary tangles was limited. Ghost tangles and argyrophilic grains were mild. The mildness of tau pathological changes in her neurons, in other words the resistance of neurons to tau pathology, may be a factor responsible for her longevity.
Collapse
Affiliation(s)
- Tamaki Iwase
- Department of Neurology, Nagoya City Koseiin Medical Welfare Center, Aichi, Japan
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Yoshio Hashizume
- Institute for Neuropathology, Fukushimura Hospital, Aichi, Japan
| | - Toshiaki Inagaki
- Department of Rehabilitation, Tosei General Hospital, Aichi, Japan
| | - Yasushi Iwasaki
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| |
Collapse
|
13
|
I F. The unique neuropathological vulnerability of the human brain to aging. Ageing Res Rev 2023; 87:101916. [PMID: 36990284 DOI: 10.1016/j.arr.2023.101916] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Alzheimer's disease (AD)-related neurofibrillary tangles (NFT), argyrophilic grain disease (AGD), aging-related tau astrogliopathy (ARTAG), limbic predominant TDP-43 proteinopathy (LATE), and amygdala-predominant Lewy body disease (LBD) are proteinopathies that, together with hippocampal sclerosis, progressively appear in the elderly affecting from 50% to 99% of individuals aged 80 years, depending on the disease. These disorders usually converge on the same subject and associate with additive cognitive impairment. Abnormal Tau, TDP-43, and α-synuclein pathologies progress following a pattern consistent with an active cell-to-cell transmission and abnormal protein processing in the host cell. However, cell vulnerability and transmission pathways are specific for each disorder, albeit abnormal proteins may co-localize in particular neurons. All these alterations are unique or highly prevalent in humans. They all affect, at first, the archicortex and paleocortex to extend at later stages to the neocortex and other regions of the telencephalon. These observations show that the phylogenetically oldest areas of the human cerebral cortex and amygdala are not designed to cope with the lifespan of actual humans. New strategies aimed at reducing the functional overload of the human telencephalon, including optimization of dream repair mechanisms and implementation of artificial circuit devices to surrogate specific brain functions, appear promising.
Collapse
Affiliation(s)
- Ferrer I
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain; Emeritus Researcher of the Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain; Biomedical Research Network of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain; Institute of Neurosciences, University of Barcelona, Barcelona, Spain; Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
14
|
Donadio V, Sturchio A, Rizzo G, Abu Rumeileh S, Liguori R, Espay AJ. Pathology vs pathogenesis: Rationale and pitfalls in the clinicopathology model of neurodegeneration. HANDBOOK OF CLINICAL NEUROLOGY 2023; 192:35-55. [PMID: 36796947 DOI: 10.1016/b978-0-323-85538-9.00001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
In neurodegenerative disorders, the term pathology is often implicitly referred to as pathogenesis. Pathology has been conceived as a window into the pathogenesis of neurodegenerative disorders. This clinicopathologic framework posits that what can be identified and quantified in postmortem brain tissue can explain both premortem clinical manifestations and the cause of death, a forensic approach to understanding neurodegeneration. As the century-old clinicopathology framework has yielded little correlation between pathology and clinical features or neuronal loss, the relationship between proteins and degeneration is ripe for revisitation. There are indeed two synchronous consequences of protein aggregation in neurodegeneration: the loss of the soluble/normal proteins on one; the accrual of the insoluble/abnormal fraction of these proteins on the other. The omission of the first part in the protein aggregation process is an artifact of the early autopsy studies: soluble, normal proteins have disappeared, with only the remaining insoluble fraction amenable to quantification. We here review the collective evidence from human data suggesting that protein aggregates, known collectively as pathology, are the consequence of many biological, toxic, and infectious exposures, but may not explain alone the cause or pathogenesis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Vincenzo Donadio
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy.
| | - Andrea Sturchio
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, Stockholm, Sweden; James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Giovanni Rizzo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Samir Abu Rumeileh
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
15
|
Abulafia C, Vidal MF, Olivar N, Odzak A, Brusco I, Guinjoan SM, Cardinali DP, Vigo DE. An Exploratory Study of Sleep-Wake Differences of Autonomic Activity in Patients with Mild Cognitive Impairment: The Role of Melatonin as a Modulating Factor. Clin Interv Aging 2023; 18:771-781. [PMID: 37200894 PMCID: PMC10187579 DOI: 10.2147/cia.s394749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 05/08/2023] [Indexed: 05/20/2023] Open
Abstract
Purpose The objective of the present study was to assess sleep-wake differences of autonomic activity in patients with mild cognitive impairment (MCI) compared to control subjects. As a post-hoc objective, we sought to evaluate the mediating effect of melatonin on this association. Patients and Methods A total of 22 MCI patients (13 under melatonin treatment) and 12 control subjects were included in this study. Sleep-wake periods were identified by actigraphy and 24hr-heart rate variability measures were obtained to study sleep-wake autonomic activity. Results MCI patients did not show any significant differences in sleep-wake autonomic activity when compared to control subjects. Post-hoc analyses revealed that MCI patients not taking melatonin displayed lower parasympathetic sleep-wake amplitude than controls not taking melatonin (RMSSD -7 ± 1 vs 4 ± 4, p = 0.004). In addition, we observed that melatonin treatment was associated with greater parasympathetic activity during sleep (VLF 15.5 ± 0.1 vs 15.1 ± 0.1, p = 0.010) and in sleep-wake differences in MCI patients (VLF 0.5 ± 0.1 vs 0.2 ± 0.0, p = 0.004). Conclusion These preliminary findings hint at a possible sleep-related parasympathetic vulnerability in patients at prodromal stages of dementia as well as a potential protective effect of exogenous melatonin in this population.
Collapse
Affiliation(s)
- Carolina Abulafia
- Laboratory of Chronophysiology, Institute for Biomedical Research (BIOMED), Pontifical Catholic University of Argentina (UCA) and CONICET, Buenos Aires, Argentina
- Facultad de Psicología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María F Vidal
- Servicio de Psiquiatría, Departamento de Neurología, Fleni, Buenos Aires, Argentina
| | - Natividad Olivar
- Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Andrea Odzak
- Servicio de Clínica Médica, Hospital Argerich, Buenos Aires, Argentina
| | - Ignacio Brusco
- Hospital de Clínicas “José de San Martín”, Universidad de Buenos Aires, Buenos Aires, Argentina
- Servicio de Clínica Médica, Hospital Argerich, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | | | - Daniel P Cardinali
- Facultad de Ciencias Médicas, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Daniel E Vigo
- Laboratory of Chronophysiology, Institute for Biomedical Research (BIOMED), Pontifical Catholic University of Argentina (UCA) and CONICET, Buenos Aires, Argentina
- Faculty of Psychology and Educational Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
- Correspondence: Daniel E Vigo, Instituto de Investigaciones Biomédicas, Pontificia Universidad Católica Argentina, Alicia Moreau de Justo 1500, 4° piso, Buenos Aires, C1107AAZ, Argentina, Tel +54 0810-2200-822 ext 1152, Email ;
| |
Collapse
|
16
|
Brenowitz WD, Yaffe K. Observational studies in Alzheimer disease: bridging preclinical studies and clinical trials. Nat Rev Neurol 2022; 18:747-757. [PMID: 36316487 PMCID: PMC9894623 DOI: 10.1038/s41582-022-00733-7] [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] [Accepted: 09/28/2022] [Indexed: 11/29/2022]
Abstract
Recent high-profile failures of Alzheimer disease treatments at the clinical trial stage have led to renewed efforts to identify and test novel interventions for Alzheimer disease and related dementias (ADRD). In this Perspective, we highlight the importance of including well-designed observational studies as part of these efforts. Observational research is an important cornerstone for gathering evidence on risk factors and causes of ADRD; this evidence can then be combined with data from preclinical studies and randomized controlled trials to inform the development of effective interventions. Observational study designs can be particularly beneficial for hypothesis generation, posing questions that are unethical or impractical for a trial setting, studying life-course associations, research in populations typically not included in trials, and public health surveillance. Here, we discuss each of these situations in the specific context of ADRD research. We also highlight novel approaches to enhance causal inference and provide a timely discussion on how observational epidemiological studies help provide a bridge between preclinical studies and successful interventions for ADRD.
Collapse
Affiliation(s)
- Willa D Brenowitz
- Departments of Psychiatry and Behavioral Sciences, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Kristine Yaffe
- Departments of Psychiatry and Behavioral Sciences, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
- San Francisco VA Medical Center, San Francisco, CA, USA.
| |
Collapse
|
17
|
Katsumata Y, Shade LM, Hohman TJ, Schneider JA, Bennett DA, Farfel JM, Kukull WA, Fardo DW, Nelson PT. Multiple gene variants linked to Alzheimer's-type clinical dementia via GWAS are also associated with non-Alzheimer's neuropathologic entities. Neurobiol Dis 2022; 174:105880. [PMID: 36191742 PMCID: PMC9641973 DOI: 10.1016/j.nbd.2022.105880] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/26/2022] Open
Abstract
The classic pathologic hallmarks of Alzheimer's disease (AD) are amyloid plaques and neurofibrillary tangles (AD neuropathologic changes, or ADNC). However, brains from individuals clinically diagnosed with "AD-type" (amnestic) dementia usually harbor heterogeneous neuropathologies in addition to, or other than, ADNC. We hypothesized that some AD-type dementia associated genetic single nucleotide variants (SNVs) identified from large genomewide association studies (GWAS) were associated with non-ADNC neuropathologies. To test this hypothesis, we analyzed data from multiple studies with available genotype and neuropathologic phenotype information. Clinical AD/dementia risk alleles of interest were derived from the very large GWAS by Bellenguez et al. (2022) who reported 83 clinical AD/dementia-linked SNVs in addition to the APOE risk alleles. To query the pathologic phenotypes associated with variation of those SNVs, National Alzheimer's disease Coordinating Center (NACC) neuropathologic data were linked to AD Sequencing Project (ADSP) and AD Genomics Consortium (ADGC) data. Separate data were obtained from the harmonized Religious Orders Study and the Rush Memory and Aging Project (ROSMAP). A total of 4811 European participants had at least ADNC neuropathology data and also genotype data available; data were meta-analyzed across cohorts. As expected, a subset of dementia-associated SNVs were associated with ADNC risk in Europeans-e.g., BIN1, PICALM, CR1, MME, and COX7C. Other gene variants linked to (clinical) AD dementia were associated with non-ADNC pathologies. For example, the associations of GRN and TMEM106B SNVs with limbic-predominant age-related TDP-43 neuropathologic changes (LATE-NC) were replicated. In addition, SNVs in TNIP1 and WNT3 previously reported as AD-related were instead associated with hippocampal sclerosis pathology. Some genotype/neuropathology association trends were not statistically significant at P < 0.05 after correcting for multiple testing, but were intriguing. For example, variants in SORL1 and TPCN1 showed trends for association with LATE-NC whereas Lewy body pathology trended toward association with USP6NL and BIN1 gene variants. A smaller cohort of non-European subjects (n = 273, approximately one-half of whom were African-Americans) provided the basis for additional exploratory analyses. Overall, these findings were consistent with the hypothesis that some genetic variants linked to AD dementia risk exert their affect by influencing non-ADNC neuropathologies.
Collapse
Affiliation(s)
- Yuriko Katsumata
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Lincoln M Shade
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julie A Schneider
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Jose M Farfel
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Walter A Kukull
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA; Department of Pathology, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
18
|
Cerebrospinal Fluid Biomarker Profile in TDP-43-Related Genetic Frontotemporal Dementia. J Pers Med 2022; 12:jpm12101747. [PMID: 36294886 PMCID: PMC9605286 DOI: 10.3390/jpm12101747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/06/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Cerebrospinal fluid (CSF) biomarkers, namely total tau, phospho-tau and amyloid beta peptides, have received much attention specifically regarding Alzheimer’s disease (AD), since they can detect the biochemical fingerprint of AD and serve as a diagnostic tool for accurate and early diagnosis during life. In the same way, biomarkers for other neurodegenerative disease pathologies are also needed. We present a case series of six patients with genetic frontotemporal dementia (FTD), with TDP-43 underlying proteinopathy, in an attempt to assess TDP-43 as a novel biomarker alone and in combination with established AD biomarkers for this specific patient group, based on the principles of personalized and precision medicine. Our results indicate that genetic TDP-43-FTD is characterized by increased CSF TPD-43 and increased TDP-43 × τΤ/τP-181 combination. Hence, TDP-43 combined with tau proteins could be a useful tool for the diagnosis of genetic FTD with TDP-43 underling histopathology, supplementing clinical, neuropsychological and imaging data.
Collapse
|
19
|
Hromadkova L, Siddiqi MK, Liu H, Safar JG. Populations of Tau Conformers Drive Prion-like Strain Effects in Alzheimer's Disease and Related Dementias. Cells 2022; 11:2997. [PMID: 36230957 PMCID: PMC9562632 DOI: 10.3390/cells11192997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Recent findings of diverse populations of prion-like conformers of misfolded tau protein expand the prion concept to Alzheimer's disease (AD) and monogenic frontotemporal lobar degeneration (FTLD)-MAPT P301L, and suggest that distinct strains of misfolded proteins drive the phenotypes and progression rates in many neurodegenerative diseases. Notable progress in the previous decades has generated many lines of proof arguing that yeast, fungal, and mammalian prions determine heritable as well as infectious traits. The extraordinary phenotypic diversity of human prion diseases arises from structurally distinct prion strains that target, at different progression speeds, variable brain structures and cells. Although human prion research presents beneficial lessons and methods to study the mechanism of strain diversity of protein-only pathogens, the fundamental molecular mechanism by which tau conformers are formed and replicate in diverse tauopathies is still poorly understood. In this review, we summarize up to date advances in identification of diverse tau conformers through biophysical and cellular experimental paradigms, and the impact of heterogeneity of pathological tau strains on personalized structure- and strain-specific therapeutic approaches in major tauopathies.
Collapse
Affiliation(s)
- Lenka Hromadkova
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | - He Liu
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jiri G. Safar
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
20
|
Zhao Z, Zhang L, Luo W, Cao Z, Zhu Q, Kong X, Zhu K, Zhang J, Wu D. Layer-specific microstructural patterns of anterior hippocampus in Alzheimer's disease with ex vivo diffusion MRI at 14.1 T. Hum Brain Mapp 2022; 44:458-471. [PMID: 36053237 PMCID: PMC9842914 DOI: 10.1002/hbm.26062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/20/2022] [Accepted: 08/16/2022] [Indexed: 01/25/2023] Open
Abstract
High-resolution ex vivo diffusion MRI (dMRI) can provide exquisite mesoscopic details and microstructural information of the human brain. Microstructural pattern of the anterior part of human hippocampus, however, has not been well elucidated with ex vivo dMRI, either in normal or disease conditions. The present study collected high-resolution (0.1 mm isotropic) dMRI of post-mortem anterior hippocampal tissues from four Alzheimer's diseases (AD), three primary age-related tauopathy (PART), and three healthy control (HC) brains on a 14.1 T spectrometer. We evaluated how AD affected dMRI-based microstructural features in different layers and subfields of anterior hippocampus. In the HC samples, we found higher anisotropy, lower diffusivity, and more streamlines in the layers within cornu ammonis (CA) than those within dentate gyrus (DG). Comparisons between disease groups showed that (1) anisotropy measurements in the CA layers of AD, especially stratum lacunosum (SL) and stratum radiatum (SR), had higher regional variability than the other two groups; (2) streamline density in the DG layers showed a gradually increased variance from HC to PART to AD; (3) AD also showed the higher variability in terms of inter-layer connectivity than HC or PART. Moreover, voxelwise correlation analysis between the coregistered dMRI and histopathology images revealed significant correlations between dMRI measurements and the contents of amyloid beta (Aβ)/tau protein in specific layers of AD samples. These findings may reflect layer-specific microstructural characteristics in different hippocampal subfields at the mesoscopic resolution, which were associated with protein deposition in the anterior hippocampus of AD patients.
Collapse
Affiliation(s)
- Zhiyong Zhao
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| | - Lei Zhang
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of NeurobiologyZhejiang University School of MedicineHangzhouChina
| | - Wanrong Luo
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| | - Zuozhen Cao
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| | - Qinfeng Zhu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| | - Xueqian Kong
- Department of ChemistryZhejiang UniversityHangzhouChina
| | - Keqing Zhu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of NeurobiologyZhejiang University School of MedicineHangzhouChina
| | - Jing Zhang
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of NeurobiologyZhejiang University School of MedicineHangzhouChina,Department of Pathology, The First Affiliated Hospital and School of MedicineZhejiang UniversityHangzhouChina
| | - Dan Wu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| |
Collapse
|
21
|
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: 10] [Impact Index Per Article: 3.3] [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
|
22
|
Wang SHJ, Guo Y, Ervin JF, Lusk JB, Luo S. Neuropathological associations of limbic-predominant age-related TDP-43 encephalopathy neuropathological change (LATE-NC) differ between the oldest-old and younger-old. Acta Neuropathol 2022; 144:45-57. [PMID: 35551470 PMCID: PMC9997084 DOI: 10.1007/s00401-022-02432-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/08/2023]
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathological change (LATE-NC) is most often seen in the oldest-old (≥ 90 years of age) but can also be present in the younger-old (< 90 years of age). In this study, we compared the neuropathological associations of LATE-NC and contribution of LATE-NC to cognitive impairment between the oldest-old and younger-old. We observed significant differences in the prevalence of LATE-NC and its association with other co-pathologies in these two age groups. LATE-NC was present in 30.9% (34/110) of the oldest-old but only 9.4% (19/203) of the younger-old. Participants of the oldest-old with LATE-NC were more likely to have hippocampal sclerosis (HS) (55.9% vs. 10.5%, p < 0.001) and moderate to severe arteriolosclerosis (82.4% vs. 50%, p = 0.007), but not intermediate to high Alzheimer's disease neuropathologic change (ADNC) (70.6% vs. 59.2%, p = 0.486) or Lewy body disease (LBD) (20.6% vs. 26.3%, p = 0.793). Participants of the younger-old with LATE-NC were more likely to have intermediate to high ADNC (94.7% vs. 55.4%, p < 0.001) and LBD (63.2% vs. 28.8%, p = 0.013) in addition to hippocampal sclerosis (42.1% vs. 6.5%, p < 0.001), and moderate to severe arteriolosclerosis (42.1% vs. 15.2%, p = 0.020). Of note, participants with LATE-NC and no to low ADNC were very rare in the younger-old (< 1%) but relatively common in the oldest-old (9.1%). Logistic regression modeling showed that in the oldest-old, both intermediate to high ADNC and LATE-NC were independently associated with higher odds of having dementia (OR: 5.09, 95% CI [1.99, 13.06], p < 0.001 for ADNC; OR: 3.28, 95% CI [1.25, 8.57], p = 0.015 for LATE-NC). In the younger-old, by contrast, intermediate to high ADNC and LBD were independently associated with higher odds of having dementia (OR: 4.43, 95% CI [2.27, 8.63], p < 0.001 for ADNC; OR: 2.55, 95% CI [1.21, 5.35], p < 0.014 for LBD), whereas LATE-NC did not show an independent association with dementia. Overall, LATE-NC is strongly associated with arteriolosclerosis and HS in both groups; however, in the younger-old, LATE-NC is associated with other neurodegenerative pathologies, such as ADNC and LBD; whereas in the oldest-old, LATE-NC can exist independent of significant ADNC.
Collapse
Affiliation(s)
- Shih-Hsiu J Wang
- Department of Pathology, Duke University Medical Center, 214MA Davison Bldg., 40 Duke Medicine Circle, Durham, NC, 27710, USA.
- Department of Neurology, Duke University Medical Center, Durham, USA.
| | - Yuanyuan Guo
- Department of Biostatics and Bioinformatics, Duke University Medical Center, Durham, USA
| | - John F Ervin
- Department of Neurology, Duke University Medical Center, Durham, USA
| | - Jay B Lusk
- Department of Pathology, Duke University Medical Center, 214MA Davison Bldg., 40 Duke Medicine Circle, Durham, NC, 27710, USA
- Department of Neurology, Duke University Medical Center, Durham, USA
| | - Sheng Luo
- Department of Biostatics and Bioinformatics, Duke University Medical Center, Durham, USA
| |
Collapse
|
23
|
Salinas J, Beiser AS, Samra JK, O'Donnell A, DeCarli CS, Gonzales MM, Aparicio HJ, Seshadri S. Association of Loneliness With 10-Year Dementia Risk and Early Markers of Vulnerability for Neurocognitive Decline. Neurology 2022; 98:e1337-e1348. [PMID: 35131906 PMCID: PMC8967424 DOI: 10.1212/wnl.0000000000200039] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 12/27/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Loneliness is common, and its prevalence is rising. The relationship of loneliness with subsequent dementia and the early preclinical course of Alzheimer disease and related dementia (ADRD) remains unclear. Thus, the primary objective of this study was to determine the association of loneliness with 10-year all-cause dementia risk and early cognitive and neuroanatomic imaging markers of ADRD vulnerability. METHODS This was a retrospective analysis of prospectively collected data from the population-based Framingham Study cohorts (September 9, 1948-December 31, 2018). Eligible participants had loneliness assessed and were dementia-free at baseline. Loneliness was recorded with the Center for Epidemiologic Studies Depression Scale, defined conservatively as feeling lonely ≥3 days in the past week. The main outcomes were incident dementia over a 10-year period, cognition, and MRI brain volumes and white matter injury. RESULTS Of 2,308 participants (mean age 73 [SD 9] years, 56% women) who met eligibility in the dementia sample, 14% (329 of 2,308) developed dementia and 6% (144 of 2,308) were lonely. Lonely (versus not lonely) adults had higher 10-year dementia risk (age-, sex-, and education-adjusted hazard ratio 1.54, 95% CI 1.06-2.24). Lonely participants <80 years of age without APOE ε4 alleles had a 3-fold greater risk (adjusted hazard ratio 3.03, 95% CI, 1.63-5.62). Among 1,875 persons without dementia who met eligibility in the cognition sample (mean age 62 [SD 9] years, 54% women), loneliness associated with poorer executive function, lower total cerebral volume, and greater white matter injury. DISCUSSION Over 10 years of close clinical dementia surveillance in this cohort study, loneliness was associated with increased dementia risk; this tripled in adults whose baseline risk would otherwise be relatively low on the basis of age and genetic risk, representing a majority of the US population. Loneliness was also associated with worse neurocognitive markers of ADRD vulnerability, suggesting an early pathogenic role. These findings may have important clinical and public health implications given observed loneliness trends. CLASSIFICATION OF EVIDENCE This study provides Class I evidence that loneliness increases the 10-year risk of developing dementia.
Collapse
Affiliation(s)
- Joel Salinas
- From the Department of Neurology (J.S.), Center for Cognitive Neurology, New York University Grossman School of Medicine, New York; The Framingham Study (J.S., A.S.B., J.K.S., A.O., H.J.A., S.S.); Department of Biostatistics (A.S.B., J.K.S., A.O.), Boston University School of Public Health; Department of Neurology (A.S.B., H.J.A., S.S.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), University of California Davis; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.M.G., S.S.), University of Texas Health Sciences Center, San Antonio.
| | - Alexa S Beiser
- From the Department of Neurology (J.S.), Center for Cognitive Neurology, New York University Grossman School of Medicine, New York; The Framingham Study (J.S., A.S.B., J.K.S., A.O., H.J.A., S.S.); Department of Biostatistics (A.S.B., J.K.S., A.O.), Boston University School of Public Health; Department of Neurology (A.S.B., H.J.A., S.S.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), University of California Davis; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.M.G., S.S.), University of Texas Health Sciences Center, San Antonio
| | - Jasmeet K Samra
- From the Department of Neurology (J.S.), Center for Cognitive Neurology, New York University Grossman School of Medicine, New York; The Framingham Study (J.S., A.S.B., J.K.S., A.O., H.J.A., S.S.); Department of Biostatistics (A.S.B., J.K.S., A.O.), Boston University School of Public Health; Department of Neurology (A.S.B., H.J.A., S.S.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), University of California Davis; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.M.G., S.S.), University of Texas Health Sciences Center, San Antonio
| | - Adrienne O'Donnell
- From the Department of Neurology (J.S.), Center for Cognitive Neurology, New York University Grossman School of Medicine, New York; The Framingham Study (J.S., A.S.B., J.K.S., A.O., H.J.A., S.S.); Department of Biostatistics (A.S.B., J.K.S., A.O.), Boston University School of Public Health; Department of Neurology (A.S.B., H.J.A., S.S.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), University of California Davis; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.M.G., S.S.), University of Texas Health Sciences Center, San Antonio
| | - Charles S DeCarli
- From the Department of Neurology (J.S.), Center for Cognitive Neurology, New York University Grossman School of Medicine, New York; The Framingham Study (J.S., A.S.B., J.K.S., A.O., H.J.A., S.S.); Department of Biostatistics (A.S.B., J.K.S., A.O.), Boston University School of Public Health; Department of Neurology (A.S.B., H.J.A., S.S.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), University of California Davis; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.M.G., S.S.), University of Texas Health Sciences Center, San Antonio
| | - Mitzi M Gonzales
- From the Department of Neurology (J.S.), Center for Cognitive Neurology, New York University Grossman School of Medicine, New York; The Framingham Study (J.S., A.S.B., J.K.S., A.O., H.J.A., S.S.); Department of Biostatistics (A.S.B., J.K.S., A.O.), Boston University School of Public Health; Department of Neurology (A.S.B., H.J.A., S.S.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), University of California Davis; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.M.G., S.S.), University of Texas Health Sciences Center, San Antonio
| | - Hugo J Aparicio
- From the Department of Neurology (J.S.), Center for Cognitive Neurology, New York University Grossman School of Medicine, New York; The Framingham Study (J.S., A.S.B., J.K.S., A.O., H.J.A., S.S.); Department of Biostatistics (A.S.B., J.K.S., A.O.), Boston University School of Public Health; Department of Neurology (A.S.B., H.J.A., S.S.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), University of California Davis; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.M.G., S.S.), University of Texas Health Sciences Center, San Antonio
| | - Sudha Seshadri
- From the Department of Neurology (J.S.), Center for Cognitive Neurology, New York University Grossman School of Medicine, New York; The Framingham Study (J.S., A.S.B., J.K.S., A.O., H.J.A., S.S.); Department of Biostatistics (A.S.B., J.K.S., A.O.), Boston University School of Public Health; Department of Neurology (A.S.B., H.J.A., S.S.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), University of California Davis; and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (M.M.G., S.S.), University of Texas Health Sciences Center, San Antonio
| |
Collapse
|
24
|
Gal J, Katsumata Y, Zhu H, Srinivasan S, Chen J, Johnson LA, Wang WX, Golden LR, Wilcock DM, Jicha GA, Cykowski MD, Nelson PT. Apolipoprotein E Proteinopathy Is a Major Dementia-Associated Pathologic Biomarker in Individuals with or without the APOE Epsilon 4 Allele. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:564-578. [PMID: 34954207 PMCID: PMC8895423 DOI: 10.1016/j.ajpath.2021.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/27/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022]
Abstract
The amygdala is vulnerable to multiple or "mixed" mis-aggregated proteins associated with neurodegenerative conditions that can manifest clinically with amnestic dementia; the amygdala region is often affected even at earliest disease stages. With the original intent of identifying novel dementia-associated proteins, the detergent-insoluble proteome was characterized from the amygdalae of 40 participants from the University of Kentucky Alzheimer's Disease Center autopsy cohort. These individuals encompassed a spectrum of clinical conditions (cognitively normal to severe amnestic dementia). Polypeptides from the detergent-insoluble fraction were interrogated using liquid chromatography-electrospray ionization-tandem mass spectrometry. As anticipated, portions of peptides previously associated with neurologic diseases were enriched from subjects with dementia. Among all detected peptides, Apolipoprotein E (ApoE) stood out: even more than the expected Tau, APP/Aβ, and α-Synuclein peptides, ApoE peptides were strongly enriched in dementia cases, including from individuals lacking the APOE ε4 genotype. The amount of ApoE protein detected in detergent-insoluble fractions was robustly associated with levels of complement proteins C3 and C4. Immunohistochemical staining of APOE ε3/ε3 subjects' amygdalae confirmed ApoE co-localization with C4 in amyloid plaques. Thus, analyses of human amygdala proteomics indicate that rather than being only an "upstream" genetic risk factor, ApoE is an aberrantly aggregated protein in its own right, and show that the ApoE protein may play active disease-driving mechanistic roles in persons lacking the APOE ε4 allele.
Collapse
Affiliation(s)
- Jozsef Gal
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, Kentucky,Department of Neuroscience, University of Kentucky, Lexington, Kentucky
| | - Yuriko Katsumata
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky,Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky,Research & Development, Lexington VA Medical Center, Lexington, Kentucky
| | - Sukanya Srinivasan
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky
| | - Lance Allen Johnson
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky,Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| | - Wang-Xia Wang
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky,Department of Pathology, University of Kentucky, Lexington, Kentucky
| | | | - Donna M. Wilcock
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky,Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky,Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Gregory A. Jicha
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky,Department of Neurology, University of Kentucky, Lexington, Kentucky
| | | | - Peter Tobias Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky; Department of Pathology, University of Kentucky, Lexington, Kentucky.
| |
Collapse
|
25
|
Emotion Recognition Deficits in the Differential Diagnosis of Amnestic Mild Cognitive Impairment: A Cognitive Marker for the Limbic-Predominant Phenotype. J Int Neuropsychol Soc 2022; 28:203-209. [PMID: 33745493 DOI: 10.1017/s1355617721000254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Late-onset amnestic mild cognitive impairment (aMCI) with long disease course and slow progression has been recently recognized as a possible phenotypical expression of a limbic-predominant neurodegenerative disorder. Basic emotion recognition ability crucially depending on temporo-limbic integrity is supposed to be impaired in this group of MCI subjects presenting a selective vulnerability of medio-temporal and limbic regions. However, no study specifically investigated this issue. METHODS Hereby, we enrolled 30 aMCI with a biomarker-based diagnosis of Alzheimer's disease (i.e., aMCI-AD, n = 16) or a biomarker evidence of selective medio-temporal and limbic degeneration (aMCI-mTLD, n = 14). Ekman-60 Faces Test (Ek-60F) was administered to each subject, comparing the performance with that of 20 healthy controls (HCs). RESULTS aMCI-mTLD subjects showed significantly lower Ek-60F global scores compared to HC (p = 0.001), whose performance was comparable to aMCI-AD. Fear (p = 0.02), surprise (p = 0.005), and anger (p = 0.01) recognition deficits characterized the aMCI-mTLD performance. Fear recognition scores were significantly lower in aMCI-mTLD compared to aMCI-AD (p = 0.04), while no differences were found in other emotions. CONCLUSIONS Impaired social cognition, suggested by defective performance in emotion recognition tasks, may be a useful cognitive marker to detect limbic-predominant aMCI subjects among the heterogeneous aMCI population.
Collapse
|
26
|
Abstract
The key pathological hallmarks-extracellular plaques and intracellular neurofibrillary tangles (NFT)-described by Alois Alzheimer in his seminal 1907 article are still central to the postmortem diagnosis of Alzheimer's disease (AD), but major advances in our understanding of the underlying pathophysiology as well as significant progress in clinical diagnosis and therapy have changed the perspective and importance of neuropathologic evaluation of the brain. The notion that the pathological processes underlying AD already start decades before symptoms are apparent in patients has brought a major change reflected in the current neuropathological classification of AD neuropathological changes (ADNC). The predictable progression of beta-amyloid (Aβ) plaque pathology from neocortex, over limbic structures, diencephalon, and basal ganglia, to brainstem and cerebellum is captured in phases described by Thal and colleagues. The progression of NFT pathology from the transentorhinal region to the limbic system and ultimately the neocortex is described in stages proposed by Braak and colleagues. The density of neuritic plaque pathology is determined by criteria defined by the Consortium to establish a registry for Alzheimer's diseases (CERAD). While these changes neuropathologically define AD, it becomes more and more apparent that the majority of patients present with a multitude of additional pathological changes which are possible contributing factors to the clinical presentation and disease progression. The impact of co-existing Lewy body pathology has been well studied, but the importance of more recently described pathologies including limbic-predominant age-related TDP-43 encephalopathy (LATE), chronic traumatic encephalopathy (CTE), and aging-related tau astrogliopathy (ARTAG) still needs to be evaluated in large cohort studies. In addition, it is apparent that vascular pathology plays an important role in the AD patient population, but a lack of standardized reporting criteria has hampered progress in elucidating the importance of these changes for clinical presentation and disease progression. More recently a key role was ascribed to the immune response to pathological protein aggregates, and it will be important to analyze these changes systematically to better understand the temporal and spatial distribution of the immune response in AD and elucidate their importance for the disease process. Advances in digital pathology and technologies such as single cell sequencing and digital spatial profiling have opened novel avenues for improvement of neuropathological diagnosis and advancing our understanding of underlying molecular processes. Finally, major strides in biomarker-based diagnosis of AD and recent advances in targeted therapeutic approaches may have shifted the perspective but also highlight the continuous importance of postmortem analysis of the brain in neurodegenerative diseases.
Collapse
Affiliation(s)
- Jorge A Trejo-Lopez
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Anthony T Yachnis
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Stefan Prokop
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
27
|
Libard S, Giedraitis V, Kilander L, Ingelsson M, Alafuzoff I. Mixed Pathologies in a Subject with a Novel PSEN1 G206R Mutation. J Alzheimers Dis 2022; 90:1601-1614. [PMID: 36314207 PMCID: PMC9789486 DOI: 10.3233/jad-220655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND There are more than 300 presenilin-1 (PSEN1) mutations identified but a thorough postmortem neuropathological assessment of the mutation carriers is seldom performed. OBJECTIVE To assess neuropathological changes (NC) in a 73-year-old subject with the novel PSEN1 G206R mutation suffering from cognitive decline in over 20 years. To compare these findings with an age- and gender-matched subject with sporadic Alzheimer's disease (sAD). METHODS The brains were assessed macro- and microscopically and the proteinopathies were staged according to current recommendations. RESULTS The AD neuropathological change (ADNC) was more extensive in the mutation carrier, although both individuals reached a high level of ADNC. The transactive DNA binding protein 43 pathology was at the end-stage in the index subject, a finding not previously described in familial AD. This pathology was moderate in the sAD subject. The PSEN1 G206R subject displayed full-blown alpha-synuclein pathology, while this proteinopathy was absent in the sAD case. Additionally, the mutation carrier displayed pronounced neuroinflammation, not previously described in association with PSEN1 mutations. CONCLUSION Our findings are exceptional, as the PSEN1 G206R subject displayed an end-stage pathology of every common proteinopathy. It is unclear whether the observed alterations are caused by the mutation or are related to a cross-seeding mechanisms. The pronounced neuroinflammation in the index patient can be reactive to the extensive NC or a contributing factor to the proteinopathies. Thorough postmortem neuropathological and genetic assessment of subjects with familial AD is warranted, for further understanding of a dementing illness.
Collapse
Affiliation(s)
- Sylwia Libard
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Surgical Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Lena Kilander
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Department of Medicine and Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Irina Alafuzoff
- Department of Surgical Pathology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
28
|
Savola S, Kaivola K, Raunio A, Kero M, Mäkelä M, Pärn K, Palta P, Tanskanen M, Tuimala J, Polvikoski T, Tienari PJ, Paetau A, Myllykangas L. Primary Age‐Related Tauopathy (PART) in a Finnish Population‐Based Study of the Oldest Old (Vantaa 85+). Neuropathol Appl Neurobiol 2021; 48:e12788. [PMID: 34927275 PMCID: PMC9305229 DOI: 10.1111/nan.12788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 11/16/2021] [Accepted: 12/12/2021] [Indexed: 11/26/2022]
Abstract
Aims Few studies have investigated primary age‐related tauopathy (PART) in a population‐based setting. Here, we assessed its prevalence, genetic background, comorbidities and features of cognitive decline in an unselected elderly population. Methods The population‐based Vantaa 85+ study includes all 601 inhabitants of Vantaa aged ≥ 85 years in 1991. Neuropathological assessment was possible in 301. Dementia (DSM IIIR criteria) and Mini‐Mental State Examination (MMSE) scores were assessed at the baseline of the study and follow‐ups. PART subjects were identified according to the criteria by Crary et al and were compared with subjects with mild and severe Alzheimer's disease (AD) neuropathological changes. The effects of other neuropathologies were taken into account using multivariate and sensitivity assays. Genetic analyses included APOE genotypes and 29 polymorphisms of the MAPT 3′ untranslated region (3′UTR region). Results The frequency of PART was 20% (n = 61/301, definite PART 5%). When PART subjects were compared with those with severe AD pathology, dementia was less common, its age at onset was higher and duration shorter. No such differences were seen when compared with those with milder AD pathology. However, both AD groups showed a steeper decline in MMSE scores in follow‐ups compared with PART. APOE ε4 frequency was lower, and APOE ε2 frequency higher in the PART group compared with each AD group. The detected nominally significant associations between PART and two MAPT 3′UTR polymorphisms and haplotypes did not survive Bonferroni correction. Conclusions PART is common among very elderly. PART subjects differ from individuals with AD‐type changes in the pattern of cognitive decline, associated genetic and neuropathological features.
Collapse
Affiliation(s)
- Sara Savola
- Department of Pathology University of Helsinki Helsinki Finland
- Department of Pathology, HUS Diagnostic Center Helsinki University Hospital Helsinki Finland
| | - Karri Kaivola
- Translational Immunology, Research Programs Unit University of Helsinki Helsinki Finland
- Department of Neurology University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Anna Raunio
- Department of Pathology University of Helsinki Helsinki Finland
- Department of Pathology, HUS Diagnostic Center Helsinki University Hospital Helsinki Finland
| | - Mia Kero
- Department of Pathology University of Helsinki Helsinki Finland
- Department of Pathology, HUS Diagnostic Center Helsinki University Hospital Helsinki Finland
| | - Mira Mäkelä
- Department of Pathology University of Helsinki Helsinki Finland
- Department of Pathology, HUS Diagnostic Center Helsinki University Hospital Helsinki Finland
| | - Kalle Pärn
- Institute for Molecular Medicine Finland (FIMM), HiLIFE University of Helsinki Helsinki Finland
| | - Priit Palta
- Institute for Molecular Medicine Finland (FIMM), HiLIFE University of Helsinki Helsinki Finland
| | - Maarit Tanskanen
- Department of Pathology University of Helsinki Helsinki Finland
- Department of Pathology, HUS Diagnostic Center Helsinki University Hospital Helsinki Finland
| | - Jarno Tuimala
- Department of Pathology University of Helsinki Helsinki Finland
| | - Tuomo Polvikoski
- Translational and Clinical Research Institute Newcastle University Newcastle upon Tyne United Kingdom
| | - Pentti J. Tienari
- Translational Immunology, Research Programs Unit University of Helsinki Helsinki Finland
- Department of Neurology University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Anders Paetau
- Department of Pathology University of Helsinki Helsinki Finland
- Department of Pathology, HUS Diagnostic Center Helsinki University Hospital Helsinki Finland
| | - Liisa Myllykangas
- Department of Pathology University of Helsinki Helsinki Finland
- Department of Pathology, HUS Diagnostic Center Helsinki University Hospital Helsinki Finland
| |
Collapse
|
29
|
Jellinger KA. Recent update on the heterogeneity of the Alzheimer’s disease spectrum. J Neural Transm (Vienna) 2021; 129:1-24. [DOI: 10.1007/s00702-021-02449-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/25/2021] [Indexed: 02/03/2023]
|
30
|
Symons GF, Clough M, Fielding J, O'Brien WT, Shepherd CE, Wright DK, Shultz SR. The Neurological Consequences of Engaging in Australian Collision Sports. J Neurotrauma 2021; 37:792-809. [PMID: 32056505 DOI: 10.1089/neu.2019.6884] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Collision sports are an integral part of Australian culture. The most common collision sports in Australia are Australian rules football, rugby union, and rugby league. Each of these sports often results in participants sustaining mild brain traumas, such as concussive and subconcussive injuries. However, the majority of previous studies and reviews pertaining to the neurological implications of sustaining mild brain traumas, while engaging in collision sports, have focused on those popular in North America and Europe. As part of this 2020 International Neurotrauma Symposium special issue, which highlights Australian neurotrauma research, this article will therefore review the burden of mild brain traumas in Australian collision sports athletes. Specifically, this review will first provide an overview of the consequences of mild brain trauma in Australian collision sports, followed by a summary of the previous studies that have investigated neurocognition, ocular motor function, neuroimaging, and fluid biomarkers, as well as neuropathological outcomes in Australian collision sports athletes. A review of the literature indicates that although Australians have contributed to the field, several knowledge gaps and limitations currently exist. These include important questions related to sex differences, the identification and implementation of blood and imaging biomarkers, the need for consistent study designs and common data elements, as well as more multi-modal studies. We conclude that although Australia has had an active history of investigating the neurological impact of collision sports participation, further research is clearly needed to better understand these consequences in Australian athletes and how they can be mitigated.
Collapse
Affiliation(s)
- Georgia F Symons
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Meaghan Clough
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Joanne Fielding
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - William T O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Claire E Shepherd
- Neuroscience Research Australia, The University of New South Wales, Sydney, New South Wales, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
31
|
Phenotypic diversity in ALS and the role of poly-conformational protein misfolding. Acta Neuropathol 2021; 142:41-55. [PMID: 32930869 DOI: 10.1007/s00401-020-02222-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/30/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022]
Abstract
In many types of familial amyotrophic lateral sclerosis (fALS), mutations cause proteins to gain toxic properties that mediate neurodegenerative processes. It is becoming increasingly clear that the proteins involved in ALS, and those responsible for a host of other neurodegenerative diseases, share many characteristics with a growing number of prion diseases. ALS is a heterogenous disease in which the majority of cases are sporadic in their etiology. Studies investigating the inherited forms of the disease are now beginning to provide evidence that some of this heterogeneity may be due to the existence of distinct conformations that ALS-linked proteins can adopt to produce the equivalent of prion strains. In this review, we discuss the in vitro and in vivo evidence that has been generated to better understand the characteristics of these proteins and how their tertiary structure may impact the disease phenotype.
Collapse
|
32
|
Tau and TDP-43 synergy: a novel therapeutic target for sporadic late-onset Alzheimer's disease. GeroScience 2021; 43:1627-1634. [PMID: 34185246 PMCID: PMC8492812 DOI: 10.1007/s11357-021-00407-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/16/2021] [Indexed: 11/10/2022] Open
Abstract
Alzheimer’s disease (AD) is traditionally defined by the presence of two types of protein aggregates in the brain: amyloid plaques comprised of the protein amyloid-β (Aβ) and neurofibrillary tangles containing the protein tau. However, a large proportion (up to 57%) of AD patients also have TDP-43 aggregates present as an additional comorbid pathology. The presence of TDP-43 aggregates in AD correlates with hippocampal sclerosis, worse brain atrophy, more severe cognitive impairment, and more rapid cognitive decline. In patients with mixed Aβ, tau, and TDP-43 pathology, TDP-43 may interact with neurodegenerative processes in AD, worsening outcomes. While considerable progress has been made to characterize TDP-43 pathology in AD and late-onset dementia, there remains a critical need for mechanistic studies to understand underlying disease biology and develop therapeutic interventions. This perspectives article reviews the current understanding of these processes from autopsy cohort studies and model organism-based research, and proposes targeting neurotoxic synergies between tau and TDP-43 as a new therapeutic strategy for AD with comorbid TDP-43 pathology.
Collapse
|
33
|
Jellinger KA. Pathobiological Subtypes of Alzheimer Disease. Dement Geriatr Cogn Disord 2021; 49:321-333. [PMID: 33429401 DOI: 10.1159/000508625] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/11/2020] [Indexed: 11/19/2022] Open
Abstract
Alzheimer disease (AD), the most common form of dementia, is a heterogenous disorder with various pathobiological subtypes. In addition to the 4 major subtypes based on the distribution of tau pathology and brain atrophy (typical, limbic predominant, hippocampal sparing, and minimal atrophy [MA]), several other clinical variants showing distinct regional patterns of tau burden have been identified: nonamnestic, corticobasal syndromal, primary progressive aphasia, posterior cortical atrophy, behavioral/dysexecutive, and mild dementia variants. Among the subtypes, differences were found in age at onset, sex distribution, cognitive status, disease duration, APOE genotype, and biomarker levels. The patterns of key network destructions parallel the tau and atrophy patterns of the AD subgroups essentially. Interruption of key networks, in particular the default-mode network that is responsible for cognitive decline, is consistent in hetero-genous AD groups. AD pathology is often associated with co-pathologies: cerebrovascular lesions, Lewy pathology, and TDP-43 proteinopathies. These mixed pathologies essentially influence the clinical picture of AD and may accel-erate disease progression. Unraveling the heterogeneity among the AD spectrum entities is important for opening a window to pathogenic mechanisms affecting the brain and enabling precision medicine approaches as a basis for developing preventive and ultimately successful disease-modifying therapies for AD.
Collapse
|
34
|
Rodrigo-Herrero S, Luque-Tirado A, Méndez-Barrio C, García-Solís D, Bernal Sánchez-Arjona M, Oropesa-Ruiz JM, Maillet D, Franco-Macías E. TMA-93 Validation by Alzheimer's Disease Biomarkers: A Comparison with the Free and Cued Selective Reminding Test on a Biobank Sample. J Alzheimers Dis 2021; 82:401-410. [PMID: 34024831 DOI: 10.3233/jad-210115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The Memory Associative Test TMA-93 examines visual relational binding, characteristically affected in early-AD stages. OBJECTIVE We aim to validate the TMA-93 by biomarkers determination and compare its diagnostic characteristics with the Free and Cued Selective Reminding Test (FCSRT). METHODS Retrospective analysis of a Biobank database. Patients' records initially consulted for memory complaints, scored MMSE≥22, had TMA-93 and FCSRT tested, and AD biomarker determination (Amyloid-PET or CSF), either positive or negative, were selected. As cutoffs, we considered the 10-percentile for TMA-93 (P10/TMA-93), and "total free recall" (TFR) 21/22, total recall (TR) 43/44, and Cued Index < 0.77 for FCSRT from previous Spanish validation and normative studies. Diagnostic utilities were calculated using ROC curves and compared by the DeLong method. We studied if one test improved the other test's prediction, following a forward stepwise logistic regression model. RESULTS We selected 105 records: 64 "positive" and 41 "negative" biomarkers. TMA-93 total score diagnostic utility (AUC = 0.72; 95%CI:0.62-0.82) was higher than those of the FCSRT: TFR (AUC = 0.70; 95%CI: 0.60-0.80), TR (AUC = 0.63; 95%CI:0.53-0.74), and Cued Index (AUC = 0.62; 95%CI:0.52-0.73). The P10/TMA-93 cutoff showed 86%sensitivity, similar to that of the most sensitive FCSRT cutoff (TFR21/22, 89%) and 29%specificity, lower than that of the most specific FCSRT cutoff (Cued Index < 0.77, 57%). 32.8%of the positive-biomarker group scored above CI/0.77 but below p10TMA-93. The addition of TMA-93 total score to FCSRT variables improved significantly the biomarkers results' prediction. CONCLUSION TMA-93 demonstrated "reasonable" diagnostic utility, similar to FCSRT, for discriminating AD biomarker groups. TMA-93 total score improved the AD biomarker result prediction when added to FCSRT variables.
Collapse
Affiliation(s)
| | | | | | - David García-Solís
- Nuclear Medicine Unit, Virgen del Rocio University Hospital, Seville, Spain
| | | | | | - Didier Maillet
- Neurology Service, Saint-Louis Hospital (AP-HP), Paris, France
| | | |
Collapse
|
35
|
Positive Association Between Serum Insulin-Like Growth Factor-1 and Cognition in Patients with Cerebral Small Vessel Disease. J Stroke Cerebrovasc Dis 2021; 30:105790. [PMID: 33878547 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 11/20/2022] Open
Abstract
Cognitive impairment is one of the main complications of cerebral small vessel disease (CSVD). Serum insulin-like growth factor-1 (IGF-1) might serve as a marker for the risk of cognitive decline in patients with CSVD. We investigated the association of IGF-1 with the development of cognitive impairment in patients with CSVD. We included 216 patients with CVSD (mean age, 67.57 ± 8.53 years; 31.9% female). We compared 117 (54.2%) patients who developed cognitive impairment with 99 (45.8%) patients without cognitive impairment. Patients who developed cognitive impairment had significantly lower levels of IGF-I (p < 0 .001), suggesting that altered IGF-1 signaling may be a risk factor for cognitive decline in patients with CSVD.
Collapse
|
36
|
Dannenhoffer-Lafage T, Best RB. A Data-Driven Hydrophobicity Scale for Predicting Liquid-Liquid Phase Separation of Proteins. J Phys Chem B 2021; 125:4046-4056. [PMID: 33876938 DOI: 10.1021/acs.jpcb.0c11479] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
An accurate model for macroscale disordered assemblies of biological macromolecules such as those formed in so-called membraneless organelles would greatly assist in studying their structure, function, and dynamics. Recent evidence has suggested that liquid-liquid phase separation (LLPS) underlies the formation of membraneless organelles. While the general mechanism of exchange of macromolecule/water for macromolecule/macromolecule interactions is known to be the driving force for LLPS, the specific interactions involved are not well understood. One way that protein-water and protein-protein interactions have been understood historically is via hydrophobicity scales. However, these scales are typically optimized for describing these relative interactions in certain cases, such as protein folding or insertion of proteins into membranes. To better describe the relative interactions of proteins that undergo LLPS, we have developed a new, data-driven hydrophobicity scale. To determine the new scale, we used coarse-grained molecular dynamics simulations using the hydrophobicity scale coarse-grained model, which relates the interactions between amino acids to their hydrophobicity. Hydrophobicity values were determined via the force-balance method on a library of proteins that includes unfolded, intrinsically disordered, and phase-separating proteins (PSP). The resulting hydrophobicity scale can better predict whether a given protein will undergo LLPS at physiological conditions by using coarse-grained molecular dynamics simulations than existing hydrophobicity scales. This new scale confirms the importance of π-π interactions between amino acids as important drivers of LLPS. This new hydrophobicity scale provides a convenient and compact description of protein-protein interactions for proteins that undergo LLPS and could be used to develop new models to describe interactions between PSP and other components, such as nucleic acids.
Collapse
Affiliation(s)
- Thomas Dannenhoffer-Lafage
- Laboratory of Chemical Physics, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0520, United States
| | - Robert B Best
- Laboratory of Chemical Physics, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0520, United States
| |
Collapse
|
37
|
Agrawal S, Yu L, Kapasi A, James BD, Arfanakis K, Barnes LL, Bennett DA, Nag S, Schneider JA. Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change and microvascular pathologies in community-dwelling older persons. Brain Pathol 2021; 31:e12939. [PMID: 33624322 PMCID: PMC8363209 DOI: 10.1111/bpa.12939] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Limbic-predominant age-related transactive response DNA-binding protein 43 (TDP-43) encephalopathy neuropathologic change (LATE-NC) and microvascular pathologies, including microinfarcts, cerebral amyloid angiopathy (CAA), and arteriolosclerosis are common in old age. A relationship between LATE-NC and arteriolosclerosis has been reported in some but not all studies. The objectives of this study were to investigate the frequency of co-occurring LATE-NC and microvascular pathologies and test the hypothesis that arteriolosclerosis, specifically, is related to LATE-NC in brains from community-dwelling older persons. Analyses included 749 deceased participants with completed data on LATE-NC and microvascular pathology from 3 longitudinal clinical pathologic studies of aging. Given the specific interest in arteriolosclerosis, we expanded the examination of arteriolosclerosis to include not only the basal ganglia but also two additional white matter regions from anterior and posterior watershed territories. Ordinal logistic regression models examined the association of microvascular pathology with LATE-NC. LATE-NC was present in 409 (54.6%) decedents, of which 354 (86.5%) had one or multiple microvascular pathologies including 132 (32.3%) with moderate-severe arteriolosclerosis in basal ganglia, 195 (47.6%) in anterior watershed, and 144 (35.2%) in posterior watershed; 170 (41.5%) with moderate-severe CAA, and 150 (36.6%) with microinfarcts. In logistic regression models, only posterior watershed arteriolosclerosis, but not other regions of arteriolosclerosis was associated with a higher odds of more advanced LATE-NC stages (Odds Ratio = 1.12; 95% Confidence Interval = 1.01-1.25) after controlling for demographics, AD, and other age-related pathologies. Capillary CAA, but not the severity of CAA was associated with an increased odds of LATE-NC burden (Odds Ratio = 1.71; 95% Confidence Interval = 1.13-2.58). Findings were unchanged in analyses controlling for APOE ε4, vascular risk factors, or vascular diseases. These findings suggest that LATE-NC with microvascular pathology is a very common mixed pathology and small vessel disease pathology may contribute to LATE-NC in the aging brain.
Collapse
Affiliation(s)
- Sonal Agrawal
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.,Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.,Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Alifiya Kapasi
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.,Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Bryan D James
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.,Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.,Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.,Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.,Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.,Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Sukriti Nag
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.,Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.,Department of Pathology, Rush University Medical Center, Chicago, IL, USA.,Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
38
|
Robinson JL, Porta S, Garrett FG, Zhang P, Xie SX, Suh E, Van Deerlin VM, Abner EL, Jicha GA, Barber JM, Lee VMY, Lee EB, Trojanowski JQ, Nelson PT. Limbic-predominant age-related TDP-43 encephalopathy differs from frontotemporal lobar degeneration. Brain 2021; 143:2844-2857. [PMID: 32830216 DOI: 10.1093/brain/awaa219] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/01/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
TAR-DNA binding protein-43 (TDP-43) proteinopathy is seen in multiple brain diseases. A standardized terminology was recommended recently for common age-related TDP-43 proteinopathy: limbic-predominant, age-related TDP-43 encephalopathy (LATE) and the underlying neuropathological changes, LATE-NC. LATE-NC may be co-morbid with Alzheimer's disease neuropathological changes (ADNC). However, there currently are ill-defined diagnostic classification issues among LATE-NC, ADNC, and frontotemporal lobar degeneration with TDP-43 (FTLD-TDP). A practical challenge is that different autopsy cohorts are composed of disparate groups of research volunteers: hospital- and clinic-based cohorts are enriched for FTLD-TDP cases, whereas community-based cohorts have more LATE-NC cases. Neuropathological methods also differ across laboratories. Here, we combined both cases and neuropathologists' diagnoses from two research centres-University of Pennsylvania and University of Kentucky. The study was designed to compare neuropathological findings between FTLD-TDP and pathologically severe LATE-NC. First, cases were selected from the University of Pennsylvania with pathological diagnoses of either FTLD-TDP (n = 33) or severe LATE-NC (mostly stage 3) with co-morbid ADNC (n = 30). Sections from these University of Pennsylvania cases were cut from amygdala, anterior cingulate, superior/mid-temporal, and middle frontal gyrus. These sections were stained for phospho-TDP-43 immunohistochemically and evaluated independently by two University of Kentucky neuropathologists blinded to case data. A simple set of criteria hypothesized to differentiate FTLD-TDP from LATE-NC was generated based on density of TDP-43 immunoreactive neuronal cytoplasmic inclusions in the neocortical regions. Criteria-based sensitivity and specificity of differentiating severe LATE-NC from FTLD-TDP cases with blind evaluation was ∼90%. Another proposed neuropathological feature related to TDP-43 proteinopathy in aged individuals is 'Alpha' versus 'Beta' in amygdala. Alpha and Beta status was diagnosed by neuropathologists from both universities (n = 5 raters). There was poor inter-rater reliability of Alpha/Beta classification (mean κ = 0.31). We next tested a separate cohort of cases from University of Kentucky with either FTLD-TDP (n = 8) or with relatively 'pure' severe LATE-NC (lacking intermediate or severe ADNC; n = 14). The simple criteria were applied by neuropathologists blinded to the prior diagnoses at University of Pennsylvania. Again, the criteria for differentiating LATE-NC from FTLD-TDP was effective, with sensitivity and specificity ∼90%. If more representative cases from each cohort (including less severe TDP-43 proteinopathy) had been included, the overall accuracy for identifying LATE-NC was estimated at >98% for both cohorts. Also across both cohorts, cases with FTLD-TDP died younger than those with LATE-NC (P < 0.0001). We conclude that in most cases, severe LATE-NC and FTLD-TDP can be differentiated by applying simple neuropathological criteria.
Collapse
Affiliation(s)
- John L Robinson
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Sílvia Porta
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Filip G Garrett
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Panpan Zhang
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Department of Biostatistics, Epidemiology and Informatics, University of Pennsyvania, Philadelphia, PA, USA
| | - Sharon X Xie
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Department of Biostatistics, Epidemiology and Informatics, University of Pennsyvania, Philadelphia, PA, USA
| | - EunRan Suh
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Vivianna M Van Deerlin
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Erin L Abner
- Department of Epidemiology, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Gregory A Jicha
- Department of Neurology, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Justin M Barber
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Virginia M-Y Lee
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Edward B Lee
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Alzheimer's Disease Core Center, University of Pennsyvania, Philadelphia, PA, USA.,Center for Neurodegenerative Disease Research, University of Pennsyvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, University of Pennsyvania, Philadelphia, PA, USA
| | - Peter T Nelson
- Department of Pathology, University of Kentucky, Lexington, KY, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
39
|
Wang X, Zhang L, Lu H, Wu JL, Liang HZ, Liu C, Tao QQ, Wu ZY, Zhu KQ. Primary age-related tauopathy in a Chinese cohort. J Zhejiang Univ Sci B 2021; 21:256-262. [PMID: 32133802 DOI: 10.1631/jzus.b1900262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Primary age-related tauopathy (PART) is characterized by the presence of tau neurofibrillary tangles (NFTs) which are typically observed in Alzheimer's disease (AD) brains, with few or without β-amyloid (Aβ) plaques. The diagnosis of PART can be categorized into "definite" or "possible" depending on the amount of Aβ plaques. Definite PART is diagnosed when NFTs are observed and the Braak stage is ≤IV, with Thal Aβ Phase 0 (Crary et al., 2014). According to the neuropathological diagnostic criteria, we reported that PART was frequently observed in the Chinese population according to our findings from specimens in our brain bank, with 47% of brain bank subjects meeting the criteria for PART. There is no consensus on the nature of PART. It remains to be elucidated whether PART is an early form of AD or a novel tauopathy (Duyckaerts et al., 2015; Jellinger et al., 2015).
Collapse
Affiliation(s)
- Xin Wang
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China.,Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lei Zhang
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China.,Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hui Lu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Juan-Li Wu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Hua-Zheng Liang
- Department of Neurology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200081, China.,Department of Neurology, Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai 200081, China.,Brain Structure and Function Group, Neuroscience Research Australia, Randwick 2031, Australia
| | - Chong Liu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China.,Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qing-Qing Tao
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhi-Ying Wu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ke-Qing Zhu
- China Brain Bank and Department of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, and Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, China.,Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
40
|
Humphrey WO, Martindale R, Pendlebury WW, DeWitt JC. Primary age-related tauopathy (PART) in the general autopsy setting: Not just a disease of the elderly. Brain Pathol 2021; 31:381-384. [PMID: 33147361 PMCID: PMC8018030 DOI: 10.1111/bpa.12919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/20/2020] [Accepted: 10/29/2020] [Indexed: 11/29/2022] Open
Affiliation(s)
- William O Humphrey
- Department of Pathology and Laboratory Medicine, The University of Vermont Medical Center, Burlington, VT, USA
| | - Rachel Martindale
- Department of Pathology and Laboratory Medicine, The University of Vermont Medical Center, Burlington, VT, USA
| | - William W Pendlebury
- Department of Pathology and Laboratory Medicine, The University of Vermont Medical Center, Burlington, VT, USA
| | - John C DeWitt
- Department of Pathology and Laboratory Medicine, The University of Vermont Medical Center, Burlington, VT, USA
| |
Collapse
|
41
|
Beach TG, Malek-Ahmadi M. Alzheimer's Disease Neuropathological Comorbidities are Common in the Younger-Old. J Alzheimers Dis 2021; 79:389-400. [PMID: 33285640 PMCID: PMC8034496 DOI: 10.3233/jad-201213] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Clinicopathological studies have demonstrated that Alzheimer's disease dementia (ADD) is often accompanied by clinically undetectable comorbid neurodegenerative and cerebrovascular disease that alter the rate of cognitive decline. Aside from causing increased variability in clinical response, it is possible that the major ADD comorbidities may not respond to ADD-specific molecular therapeutics. OBJECTIVE As most reports have focused on comorbidity in the oldest-old, its extent in younger age groups that are more likely to be involved in clinical trials is largely unknown; our objective is to provide this information. METHODS We conducted a survey of neuropathological comorbidities in sporadic ADD using data from the US National Alzheimer's Coordinating Center. Subject data was restricted to those with dementia and meeting National Institute on Aging-Alzheimer's Association intermediate or high AD Neuropathological Change levels, excluding those with known autosomal dominant AD-related mutations. RESULTS Highly prevalent ADD comorbidities are not restricted to the oldest-old but are common even in early-onset ADD. The percentage of cases with ADD as the sole major neuropathological diagnosis is highest in the under-60 group, where "pure" ADD cases are still in the minority at 44%. After this AD as a sole major pathology in ADD declines to roughly 20%in the 70s and beyond. Lewy body disease is the most common comorbidity at younger ages but actually is less common at later ages, while for most others, their prevalence increases with age. CONCLUSION Alzheimer's disease neuropathological comorbidities are highly prevalent even in the younger-old.
Collapse
|
42
|
de Boer EMJ, Orie VK, Williams T, Baker MR, De Oliveira HM, Polvikoski T, Silsby M, Menon P, van den Bos M, Halliday GM, van den Berg LH, Van Den Bosch L, van Damme P, Kiernan MC, van Es MA, Vucic S. TDP-43 proteinopathies: a new wave of neurodegenerative diseases. J Neurol Neurosurg Psychiatry 2020; 92:jnnp-2020-322983. [PMID: 33177049 PMCID: PMC7803890 DOI: 10.1136/jnnp-2020-322983] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/28/2020] [Accepted: 09/13/2020] [Indexed: 12/31/2022]
Abstract
Inclusions of pathogenic deposits containing TAR DNA-binding protein 43 (TDP-43) are evident in the brain and spinal cord of patients that present across a spectrum of neurodegenerative diseases. For instance, the majority of patients with sporadic amyotrophic lateral sclerosis (up to 97%) and a substantial proportion of patients with frontotemporal lobar degeneration (~45%) exhibit TDP-43 positive neuronal inclusions, suggesting a role for this protein in disease pathogenesis. In addition, TDP-43 inclusions are evident in familial ALS phenotypes linked to multiple gene mutations including the TDP-43 gene coding (TARDBP) and unrelated genes (eg, C9orf72). While TDP-43 is an essential RNA/DNA binding protein critical for RNA-related metabolism, determining the pathophysiological mechanisms through which TDP-43 mediates neurodegeneration appears complex, and unravelling these molecular processes seems critical for the development of effective therapies. This review highlights the key physiological functions of the TDP-43 protein, while considering an expanding spectrum of neurodegenerative diseases associated with pathogenic TDP-43 deposition, and dissecting key molecular pathways through which TDP-43 may mediate neurodegeneration.
Collapse
Affiliation(s)
- Eva Maria Johanna de Boer
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Viyanti K Orie
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Timothy Williams
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Mark R Baker
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Clinical Neurophysiology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Hugo M De Oliveira
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Tuomo Polvikoski
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Neuropathology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Matthew Silsby
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Parvathi Menon
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Mehdi van den Bos
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Glenda M Halliday
- Brain and Mind Center, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Leonard H van den Berg
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Philip van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
- Department of Neurology, University Hospital Leuven, Leuven, Belgium
| | - Matthew C Kiernan
- Brain and Mind Center, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Michael A van Es
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Steve Vucic
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
43
|
Rubinski A, Franzmeier N, Neitzel J, Ewers M. FDG-PET hypermetabolism is associated with higher tau-PET in mild cognitive impairment at low amyloid-PET levels. ALZHEIMERS RESEARCH & THERAPY 2020; 12:133. [PMID: 33076977 PMCID: PMC7574434 DOI: 10.1186/s13195-020-00702-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/05/2020] [Indexed: 12/04/2022]
Abstract
Background FDG-PET hypermetabolism can be observed in mild cognitive impairment (MCI), but the link to primary pathologies of Alzheimer’s diseases (AD) including amyloid and tau is unclear. Methods Using voxel-based regression, we assessed local interactions between amyloid- and tau-PET on spatially matched FDG-PET in 72 MCI patients. Control groups included cerebrospinal fluid biomarker characterized cognitively normal (CN, n = 70) and AD dementia subjects (n = 95). Results In MCI, significant amyloid-PET by tau-PET interactions were found in frontal, lateral temporal, and posterior parietal regions, where higher local tau-PET was associated with higher spatially corresponding FDG-PET at low levels of local amyloid-PET. FDG-PET in brain regions with a significant local amyloid- by tau-PET interaction was higher compared to that in CN and AD dementia and associated with lower episodic memory. Conclusion Higher tau-PET in the presence of low amyloid-PET is associated with abnormally increased glucose metabolism that is accompanied by episodic memory impairment.
Collapse
Affiliation(s)
- Anna Rubinski
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Julia Neitzel
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Feodor-Lynen-Straße 17, 81377, Munich, Germany. .,German Center for Neurodegenerative Diseases, Munich, Germany.
| | | |
Collapse
|
44
|
Trejo-Lopez JA, Sorrentino ZA, Riffe CJ, Lloyd GM, Labuzan SA, Dickson DW, Yachnis AT, Prokop S, Giasson BI. Novel monoclonal antibodies targeting the RRM2 domain of human TDP-43 protein. Neurosci Lett 2020; 738:135353. [PMID: 32905837 DOI: 10.1016/j.neulet.2020.135353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 10/23/2022]
Abstract
Transactive response DNA-binding protein of 43 kilodaltons (TDP-43) is a 414 amino acid protein that under physiologic conditions localizes to the nucleus and participates in the regulation of RNA metabolism through two RNA recognition motifs (RRM1 and RRM2). In neurodegenerative diseases, TDP-43 may become hyperphosphorylated, ubiquitinated, and aggregate into cytoplasmic inclusions. TDP-43 is now well-characterized as a pathologic protein of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with TDP-43 proteinopathy (FTLD-TDP). Additionally, a common TDP-43 proteinopathy arising outside of the context of ALS and FTLD-TDP has been recently described, termed "limbic predominant age-related TDP-43 encephalopathy (LATE)." In the current study, two novel mouse-derived monoclonal antibodies, 2G11 and 2H1, raised against an epitope within the RRM2 domain of TDP-43 (residues 198-216), were characterized for specificity and immunohistochemical application in human brain from cases of Alzheimer's disease (AD), Lewy Body Disease (LBD), amyotrophic lateral sclerosis (ALS), and frontotemporal lobe degeneration with TDP-43 inclusions (FTLD-TDP). Immunoblot analysis of these antibodies in HEK293T cells revealed efficient detection of intact human TDP-43 protein, and in N2A cells showed no reactivity for mouse TDP-43. Immunohistochemically applied to formalin-fixed paraffin-embedded tissues, 2G11 and 2H1 robustly identified the classic inclusions of ALS and FTLD-TDP, and efficaciously provided a diagnosis of LATE in cases of AD and LBD. These novel antibodies label aberrant intracytoplasmic protein inclusions without relying on hyperphosphorylated epitopes, and provide elegant discrimination between TDP-43 and tau neurofibrillary tangles within neurodegenerative comorbidity.
Collapse
Affiliation(s)
- Jorge A Trejo-Lopez
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA; Department of Department of Laboratory Medicine and Anatomic Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Zachary A Sorrentino
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| | - Cara J Riffe
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| | - Grace M Lloyd
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| | - Sydney A Labuzan
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Anthony T Yachnis
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Stefan Prokop
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA; Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Benoit I Giasson
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
45
|
Jellinger KA. Neuropathological assessment of the Alzheimer spectrum. J Neural Transm (Vienna) 2020; 127:1229-1256. [PMID: 32740684 DOI: 10.1007/s00702-020-02232-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer disease (AD), the most common form of dementia globally, classically defined a clinicopathological entity, is a heterogenous disorder with various pathobiological subtypes, currently referred to as Alzheimer continuum. Its morphological hallmarks are extracellular parenchymal β-amyloid (amyloid plaques) and intraneuronal (tau aggregates forming neurofibrillary tangles) lesions accompanied by synaptic loss and vascular amyloid deposits, that are essential for the pathological diagnosis of AD. In addition to "classical" AD, several subtypes with characteristic regional patterns of tau pathology have been described that show distinct clinical features, differences in age, sex distribution, biomarker levels, and patterns of key network destructions responsible for cognitive decline. AD is a mixed proteinopathy (amyloid and tau), frequently associated with other age-related co-pathologies, such as cerebrovascular lesions, Lewy and TDP-43 pathologies, hippocampal sclerosis, or argyrophilic grain disease. These and other co-pathologies essentially influence the clinical picture of AD and may accelerate disease progression. The purpose of this review is to provide a critical overview of AD pathology, its defining pathological substrates, and the heterogeneity among the Alzheimer spectrum entities that may provide a broader diagnostic coverage of this devastating disorder as a basis for implementing precision medicine approaches and for ultimate development of successful disease-modifying drugs for AD.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
46
|
Hickman RA, Flowers XE, Wisniewski T. Primary Age-Related Tauopathy (PART): Addressing the Spectrum of Neuronal Tauopathic Changes in the Aging Brain. Curr Neurol Neurosci Rep 2020; 20:39. [PMID: 32666342 DOI: 10.1007/s11910-020-01063-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Primary age-related tauopathy (PART) was recently proposed as a pathologic diagnosis for brains that harbor neurofibrillary tangles (Braak stage ≤ 4) with little, if any, amyloid burden. We sought to review the clinicopathologic findings related to PART. RECENT FINDINGS Most adult human brains show at least focal tauopathic changes, and the majority of individuals with PART do not progress to dementia. Older age and cognitive impairment correlate with increased Braak stage, and multivariate analyses suggest that the rate of cognitive decline is less than matched patients with Alzheimer disease (AD). It remains unclear whether PART is a distinct tauopathic entity separate from AD or rather represents an earlier histologic stage of AD. Cognitive decline in PART is usually milder than AD and correlates with tauopathic burden. Biomarker and ligand-based radiologic studies will be important to define PART antemortem and prospectively follow its natural history.
Collapse
Affiliation(s)
- Richard A Hickman
- Department of Pathology and Cell Biology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 630 West 168th Street, PH 15-124, New York, NY, 10032, USA.
| | - Xena E Flowers
- Department of Pathology and Cell Biology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 630 West 168th Street, PH 15-124, New York, NY, 10032, USA
| | - Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry, Center for Cognitive Neurology, NYU School of Medicine, Science Building, Rm 1017, 435 East 30th Street, New York, NY, 10016, USA
| |
Collapse
|
47
|
Rosa G, Giannotti C, Martella L, Massa F, Serafini G, Pardini M, Nobili FM, Monacelli F. Brain Aging, Cardiovascular Diseases, Mixed Dementia, and Frailty in the Oldest Old: From Brain Phenotype to Clinical Expression. J Alzheimers Dis 2020; 75:1083-1103. [DOI: 10.3233/jad-191075] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Gianmarco Rosa
- Department of Internal Medicine and Medical Specialties, DIMI, Section of Cardiovascular Diseases, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Giannotti
- Department of Internal Medicine and Medical Specialties, DIMI, Section of Geriatrics, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lucia Martella
- Department of Internal Medicine and Medical Specialties, DIMI, Section of Geriatrics, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Flavio Mariano Nobili
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health DINOGMI, Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties, DIMI, Section of Geriatrics, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | |
Collapse
|
48
|
Josephs KA, Mackenzie I, Frosch MP, Bigio EH, Neumann M, Arai T, Dugger BN, Ghetti B, Grossman M, Hasegawa M, Herrup K, Holton J, Jellinger K, Lashley T, McAleese KE, Parisi JE, Revesz T, Saito Y, Vonsattel JP, Whitwell JL, Wisniewski T, Hu W. LATE to the PART-y. Brain 2020; 142:e47. [PMID: 31359030 PMCID: PMC6736234 DOI: 10.1093/brain/awz224] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
| | - Ian Mackenzie
- Department of Pathology and Laboratory Medicine, University of British Columbia and Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Matthew P Frosch
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eileen H Bigio
- Feinberg School of Medicine, Northwesterm University, Chicago, IL, USA
| | - Manuela Neumann
- Department of Neuropathology, University of Tübingen and German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Tetsuaki Arai
- Department of Psychiatry, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, UC Davis, Sacramento, CA, USA
| | - Bernardino Ghetti
- Pathology and Laboratory Medicine, Indiana University, Indiana, IL, USA
| | - Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Masato Hasegawa
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Karl Herrup
- Department of Neurology, Alzheimer's Disease Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Janice Holton
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Kurt Jellinger
- Institute of Clinical Neurobiology, Medical University of Vienna, Vienna, Austria
| | - Tammaryn Lashley
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Joseph E Parisi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Tamas Revesz
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Yuko Saito
- National Center of Neurology and Pathology Brain Bank, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | | | | | | | - William Hu
- Department of Neurology and Center for Neurodegenerative Diseases Research, Emory University, Atlanta, GA, USA
| |
Collapse
|
49
|
Ooi L, Dottori M, Cook AL, Engel M, Gautam V, Grubman A, Hernández D, King AE, Maksour S, Targa Dias Anastacio H, Balez R, Pébay A, Pouton C, Valenzuela M, White A, Williamson R. If Human Brain Organoids Are the Answer to Understanding Dementia, What Are the Questions? Neuroscientist 2020; 26:438-454. [PMID: 32281909 PMCID: PMC7539594 DOI: 10.1177/1073858420912404] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Because our beliefs regarding our individuality, autonomy, and personhood are intimately bound up with our brains, there is a public fascination with cerebral organoids, the "mini-brain," the "brain in a dish". At the same time, the ethical issues around organoids are only now being explored. What are the prospects of using human cerebral organoids to better understand, treat, or prevent dementia? Will human organoids represent an improvement on the current, less-than-satisfactory, animal models? When considering these questions, two major issues arise. One is the general challenge associated with using any stem cell-generated preparation for in vitro modelling (challenges amplified when using organoids compared with simpler cell culture systems). The other relates to complexities associated with defining and understanding what we mean by the term "dementia." We discuss 10 puzzles, issues, and stumbling blocks to watch for in the quest to model "dementia in a dish."
Collapse
Affiliation(s)
- Lezanne Ooi
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia.,School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | - Mirella Dottori
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia.,School of Medicine, University of Wollongong, Wollongong, New South Wales, Australia.,Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, Australia.,Department of Biomedical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony L Cook
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Martin Engel
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia.,School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | - Vini Gautam
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Alexandra Grubman
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Damián Hernández
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, Australia.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, East Melbourne, Victoria, Australia.,Department of Surgery, University of Melbourne, Parkville, Victoria, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Simon Maksour
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia.,School of Medicine, University of Wollongong, Wollongong, New South Wales, Australia
| | - Helena Targa Dias Anastacio
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia.,School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | - Rachelle Balez
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia.,School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | - Alice Pébay
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, Australia.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, East Melbourne, Victoria, Australia.,Department of Surgery, University of Melbourne, Parkville, Victoria, Australia
| | - Colin Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Michael Valenzuela
- Regenerative Neuroscience Group, Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Anthony White
- Queensland Institute of Medical Research Berghofer, Brisbane, Queensland, Australia
| | - Robert Williamson
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
50
|
Babinchak WM, Surewicz WK. Liquid-Liquid Phase Separation and Its Mechanistic Role in Pathological Protein Aggregation. J Mol Biol 2020; 432:1910-1925. [PMID: 32169484 DOI: 10.1016/j.jmb.2020.03.004] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/14/2022]
Abstract
Liquid-liquid phase separation (LLPS) of proteins underlies the formation of membrane-less organelles. While it has been recognized for some time that these organelles are of key importance for normal cellular functions, a growing number of recent observations indicate that LLPS may also play a role in disease. In particular, numerous proteins that form toxic aggregates in neurodegenerative diseases, such as amyotrophic lateral sclerosis, frontotemporal lobar degeneration, and Alzheimer's disease, were found to be highly prone to phase separation, suggesting that there might be a strong link between LLPS and the pathogenic process in these disorders. This review aims to assess the molecular basis of this link through exploration of the intermolecular interactions that underlie LLPS and aggregation and the underlying mechanisms facilitating maturation of liquid droplets into more stable assemblies, including so-called labile fibrils, hydrogels, and pathological amyloids. Recent insights into the structural basis of labile fibrils and potential mechanisms by which these relatively unstable structures could transition into more stable pathogenic amyloids are also discussed. Finally, this review explores how the environment of liquid droplets could modulate protein aggregation by altering kinetics of protein self-association, affecting folding of protein monomers, or changing aggregation pathways.
Collapse
Affiliation(s)
- W Michael Babinchak
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Witold K Surewicz
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|