1
|
Tyagi SC. Lactobacillus Eats Amyloid Plaque and Post-Biotically Attenuates Senescence Due to Repeat Expansion Disorder and Alzheimer's Disease. Antioxidants (Basel) 2024; 13:1225. [PMID: 39456478 PMCID: PMC11506100 DOI: 10.3390/antiox13101225] [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: 08/22/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Patients with Alzheimer's disease and related dementia (ADRD) are faced with a formidable challenge of focal amyloid deposits and cerebral amyloid angiopathy (CAA). The treatment of amyloid deposits in ADRD by targeting only oxidative stress, inflammation and hyperlipidemia has not yielded significant positive clinical outcomes. The chronic high-fat diet (HFD), or gut dysbiosis, is one of the major contributors of ADRD in part by disrupted transport, epigenetic DNMT1 and the folate 1-carbon metabolism (FOCM) cycle, i.e., rhythmic methylation/de-methylation on DNA, an active part of epigenetic memory during genes turning off and on by the gene writer (DNMT1) and eraser (TET2/FTO) and the transsulfuration pathway by mitochondrial 3-mercaptopyruvate sulfur transferase (3MST)-producing H2S. The repeat CAG expansion and m6A disorder causes senescence and AD. We aim to target the paradigm-shift pathway of the gut-brain microbiome axis that selectively inhibits amyloid deposits and increases mitochondrial transsulfuration and H2S. We have observed an increase in DNMT1 and decreased FTO levels in the cortex of the brain of AD mice. Interestingly, we also observed that probiotic lactobacillus-producing post-biotic folate and lactone/ketone effectively prevented FOCM-associated gut dysbiosis and amyloid deposits. The s-adenosine-methionine (SAM) transporter (SLC25A) was increased by hyperhomocysteinemia (HHcy). Thus, we hypothesize that chronic gut dysbiosis induces SLC25A, the gene writer, and HHcy, and decreases the gene eraser, leading to a decrease in SLC7A and mitochondrial transsulfuration H2S production and bioenergetics. Lactobacillus engulfs lipids/cholesterol and a tri-directional post-biotic, folic acid (an antioxidant and inhibitor of beta amyloid deposits; reduces Hcy levels), and the lactate ketone body (fuel for mitochondria) producer increases SLC7A and H2S (an antioxidant, potent vasodilator and neurotransmitter gas) production and inhibits amyloid deposits. Therefore, it is important to discuss whether lactobacillus downregulates SLC25A and DNMT1 and upregulates TET2/FTO, inhibiting β-amyloid deposits by lowering homocysteine. It is also important to discuss whether lactobacillus upregulates SLC7A and inhibits β-amyloid deposits by increasing the mitochondrial transsulfuration of H2S production.
Collapse
Affiliation(s)
- Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
2
|
Lavrova A, Pham NTT, Vernon CJ, Carlos AF, Petersen RC, Dickson DW, Lowe VJ, Jack CR, Whitwell JL, Josephs KA. A multimodal clinical diagnostic approach using MRI and 18F-FDG-PET for antemortem diagnosis of TDP-43 in cases with low-intermediate Alzheimer's disease neuropathologic changes and primary age-related tauopathy. J Neurol 2024; 271:4105-4118. [PMID: 38578498 PMCID: PMC11233234 DOI: 10.1007/s00415-024-12312-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 04/06/2024]
Abstract
OBJECTIVE To evaluate the utility of clinical assessment scales for MRI and 18F-FDG-PET as potential in vivo predictive diagnostic tools for TAR DNA-binding protein of 43 kDa (TDP-43) proteinopathy in cases with low-intermediate Alzheimer's disease neuropathologic changes (ADNC) and primary age-related tauopathy (PART). METHODS We conducted a cross-sectional analysis on patients with antemortem MRI and 18F-FDG-PET scans and postmortem diagnosis of low-intermediate ADNC or PART (Braak stage ≤ III; Thal β-amyloid phase 0-5). We employed visual imaging scales to grade structural changes on MRI and metabolic changes on 18F-FDG-PET and statistically compared demographic and clinicopathological characteristics between TDP-43 positive and negative cases. Independent regression analyses were performed to assess further influences of pathological characteristics on imaging outcomes. Within-reader repeatability and inter-reader reliability were calculated (CI = 0.95). Additional quantitative region-of-interest analyses of MRI gray matter volumes and PET ligand uptake were performed. RESULTS Of the 64 cases in the study, 20 (31%) were TDP-43 ( +), of which 12 (60%) were female. TDP-43 ( +) cases were more likely to have hippocampal sclerosis (HS) (p = 0.014) and moderate-severe medial temporal lobe atrophy on MRI (p = 0.048). TDP-43( +) cases also showed a trend for less parietal atrophy on MRI (p = 0.086) and more medial temporal lobe hypometabolism on 18F-FDG-PET (p = 0.087) than TDP-43( - ) cases. Regression analysis showed an association between medial temporal hypometabolism and HS (p = 0.0113). ICC values for MRI and PET within one reader were 0.75 and 0.91; across two readers were 0.79 and 0.82. The region-of-interest-based analysis confirmed a significant difference between TDP-43( +) and TDP-43( - ) cases for medial temporal lobe gray matter volume on MRI (p = 0.014) and medial temporal metabolism on PET (p = 0.011). CONCLUSION Visual inspection of the medial temporal lobe on MRI and FDG-PET may help to predict TDP-43 status in the context of low-intermediate ADNC and PART.
Collapse
Affiliation(s)
- Anna Lavrova
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Arenn F Carlos
- Department of Neurology, College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA
| | - Ronald C Petersen
- Department of Neurology, College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA
| | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Keith A Josephs
- Department of Neurology, College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
3
|
Liou JJ, Li J, Berardinelli J, Jin H, Santini T, Noh J, Farhat N, Wu M, Aizenstein H, Mettenburg JM, Yong W, Head E, Ikonomovic M, Ibrahim T, Kofler J. Correlating hippocampal and amygdala volumes with neuropathological burden in neurodegenerative diseases using 7T postmortem MRI. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.15.24307354. [PMID: 38798514 PMCID: PMC11118630 DOI: 10.1101/2024.05.15.24307354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Numerous research groups worldwide have focused on postmortem imaging to bridge the resolution gap between clinical neuroimaging and neuropathology data. We developed a standardized protocol for brain embedding, imaging, and processing, facilitating alignment between antemortem MRI, postmortem MRI, and pathology to observe brain atrophy and structural damage progression over time. Using 7T postmortem ex vivo MRI, we explore the potential correlation of amygdala and hippocampal atrophy with neuropathological burden in both Down syndrome (DS) and Alzheimer's disease (AD) cohorts. Using 7T postmortem ex vivo MRI scans from 66 cases (12 DS and 54 AD) alongside a subset of antemortem scans (n=17), we correlated manually segmented hippocampal and amygdala volumes, adjusted for age, sex, and ApoE4 status, with pathological indicators such as Thal phase, Braak stage, limbic-predominant age-related TDP-43 encephalopathy (LATE) stage, hippocampal sclerosis (HS), and Lewy body (LB) stage. A significant correlation was observed between postmortem and antemortem volumes for the hippocampus, but a similar trend observed for the amygdala did not reach statistical significance. DS individuals exhibited notably smaller hippocampal and amygdala volumes compared to AD subjects. In DS, lower hippocampal and amygdala volumes correlated with more severe Braak stage, without significant associations with Thal phase. LATE and HS pathologies were uncommon in DS cases but trended toward smaller hippocampal volumes. In AD, lower hippocampal volume associated with dementia duration, advanced Thal phase, Braak stage, LATE stage, and HS presence, whereas reduced amygdala volume correlated mainly with severe LATE stage and HS, but not with Thal or Braak stages. No significant LB correlation was detected in either DS or AD cohorts. Hippocampal volume in AD appears influenced by both AD and LATE pathologies, while amygdala volume seems primarily influenced by LATE. In DS, smaller hippocampal volume, relative to AD, appears primarily influenced by tau pathology.
Collapse
|
4
|
Chun MY, Chae W, Seo SW, Jang H, Yun J, Na DL, Kang D, Lee J, Hammers DB, Apostolova LG, Jang SI, Kim HJ. Effects of risk factors on the development and mortality of early- and late-onset dementia: an 11-year longitudinal nationwide population-based cohort study in South Korea. Alzheimers Res Ther 2024; 16:92. [PMID: 38664771 PMCID: PMC11044300 DOI: 10.1186/s13195-024-01436-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Early-onset dementia (EOD, onset age < 65) and late-onset dementia (LOD, onset age ≥ 65) exhibit distinct features. Understanding the risk factors for dementia development and mortality in EOD and LOD respectively is crucial for personalized care. While risk factors are known for LOD development and mortality, their impact on EOD remains unclear. We aimed to investigate how hypertension, diabetes mellitus, hyperlipidemia, atrial fibrillation, and osteoporosis influence the development and mortality of EOD and LOD, respectively. METHODS Using the Korean National Health Insurance Service (NHIS) database, we collected 546,709 dementia-free individuals and followed up for 11 years. In the two study groups, the Younger group (< 65 years old) and the Older group (≥ 65 years old), we applied Cox proportional hazard models to assess risk factors for development of EOD and LOD, respectively. Then, we assessed risk factors for mortality among EOD and LOD. RESULTS Diabetes mellitus and osteoporosis increased the risk of EOD and LOD development. Hypertension increased the risk of EOD, while atrial fibrillation increased the risk of LOD. Conversely, hyperlipidemia exhibited a protective effect against LOD development. Additionally, diabetes mellitus increased mortality in EOD and LOD. Hypertension and atrial fibrillation increased mortality in LOD, while hyperlipidemia decreased mortality in EOD and LOD. CONCLUSIONS Risk factors influencing dementia development and mortality differed in EOD and LOD. Targeted public health interventions addressing age-related risk factors may reduce dementia incidence and mortality.
Collapse
Affiliation(s)
- Min Young Chun
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Department of Neurology, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, South Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, 363 Dongbaekjukjeon-daero, Giheung-gu, , Yongin-si, Gyeonggi-do, 16995, South Korea
| | - Wonjeong Chae
- Office of Strategic Planning, Healthcare Policy and Strategy Task Force, Yonsei University Health System, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Department of Neurology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Jihwan Yun
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Department of Neurology, Soonchunhyang University Bucheon Hospital, 170, Jomaru-ro, Wonmi-Gu, Bucheon-si, Gyeonggi-do, 14574, South Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Dongwoo Kang
- Department of Data Science, Hanmi Pharm. Co., Ltd, 14, Wiryeseong-daero, Songpa-gu, Seoul, South Korea
| | - Jungkuk Lee
- Department of Data Science, Hanmi Pharm. Co., Ltd, 14, Wiryeseong-daero, Songpa-gu, Seoul, South Korea
| | - Dustin B Hammers
- Department of Neurology, Indiana University School of Medicine, 355 W 16th St, Indianapolis, IN, USA
| | - Liana G Apostolova
- Department of Neurology, Indiana University School of Medicine, 355 W 16th St, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine Indianapolis, 355W 16th St, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 355W 16th St, Indianapolis, IN, USA
| | - Sung-In Jang
- Department of Preventive Medicine, College of Medicine, Yonsei University, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, South Korea.
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
- Department of Digital Health, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
- Department of Neurology, Indiana University School of Medicine, 355 W 16th St, Indianapolis, IN, USA.
| |
Collapse
|
5
|
Chung M, Carter EK, Veire AM, Dammer EB, Chang J, Duong DM, Raj N, Bassell GJ, Glass JD, Gendron TF, Nelson PT, Levey AI, Seyfried NT, McEachin ZT. Cryptic exon inclusion is a molecular signature of LATE-NC in aging brains. Acta Neuropathol 2024; 147:29. [PMID: 38308693 PMCID: PMC10838224 DOI: 10.1007/s00401-023-02671-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/30/2023] [Accepted: 12/17/2023] [Indexed: 02/05/2024]
Abstract
The aggregation, mislocalization, and phosphorylation of TDP-43 are pathologic hallmarks of several neurodegenerative diseases and provide a defining criterion for the neuropathologic diagnosis of Limbic-predominant Age-related TDP-43 Encephalopathy (LATE). LATE neuropathologic changes (LATE-NC) are often comorbid with other neurodegenerative pathologies including Alzheimer's disease neuropathologic changes (ADNC). We examined whether TDP-43 regulated cryptic exons accumulate in the hippocampus of neuropathologically confirmed LATE-NC cases. We found that several cryptic RNAs are robustly expressed in LATE-NC cases with or without comorbid ADNC and correlate with pTDP-43 abundance; however, the accumulation of cryptic RNAs is more robust in LATE-NC with comorbid ADNC. Additionally, cryptic RNAs can robustly distinguish LATE-NC from healthy controls and AD cases. These findings expand our current understanding and provide novel potential biomarkers for LATE pathogenesis.
Collapse
Affiliation(s)
- Mingee Chung
- Department of Cell Biology, Emory University, Atlanta, GA, 30322, USA
- Laboratory for Translational Cell Biology, Emory University, Atlanta, GA, 30322, USA
| | - E Kathleen Carter
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Austin M Veire
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Jianjun Chang
- Department of Cell Biology, Emory University, Atlanta, GA, 30322, USA
| | - Duc M Duong
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Nisha Raj
- Department of Cell Biology, Emory University, Atlanta, GA, 30322, USA
- Laboratory for Translational Cell Biology, Emory University, Atlanta, GA, 30322, USA
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University, Atlanta, GA, 30322, USA
- Laboratory for Translational Cell Biology, Emory University, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA, 30322, USA
| | - Jonathan D Glass
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA, 30322, USA
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Peter T Nelson
- Department of Pathology and Sanders-Brown Center On Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Allan I Levey
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA, 30322, USA.
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA.
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA.
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA, 30322, USA.
| | - Zachary T McEachin
- Department of Cell Biology, Emory University, Atlanta, GA, 30322, USA.
- Laboratory for Translational Cell Biology, Emory University, Atlanta, GA, 30322, USA.
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA.
- Center for Neurodegenerative Diseases, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
6
|
Agra Almeida Quadros AR, Li Z, Wang X, Ndayambaje IS, Aryal S, Ramesh N, Nolan M, Jayakumar R, Han Y, Stillman H, Aguilar C, Wheeler HJ, Connors T, Lopez-Erauskin J, Baughn MW, Melamed Z, Beccari MS, Olmedo Martínez L, Canori M, Lee CZ, Moran L, Draper I, Kopin AS, Oakley DH, Dickson DW, Cleveland DW, Hyman BT, Das S, Ertekin-Taner N, Lagier-Tourenne C. Cryptic splicing of stathmin-2 and UNC13A mRNAs is a pathological hallmark of TDP-43-associated Alzheimer's disease. Acta Neuropathol 2024; 147:9. [PMID: 38175301 PMCID: PMC10766724 DOI: 10.1007/s00401-023-02655-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024]
Abstract
Nuclear clearance and cytoplasmic accumulations of the RNA-binding protein TDP-43 are pathological hallmarks in almost all patients with amyotrophic lateral sclerosis (ALS) and up to 50% of patients with frontotemporal dementia (FTD) and Alzheimer's disease. In Alzheimer's disease, TDP-43 pathology is predominantly observed in the limbic system and correlates with cognitive decline and reduced hippocampal volume. Disruption of nuclear TDP-43 function leads to abnormal RNA splicing and incorporation of erroneous cryptic exons in numerous transcripts including Stathmin-2 (STMN2, also known as SCG10) and UNC13A, recently reported in tissues from patients with ALS and FTD. Here, we identify both STMN2 and UNC13A cryptic exons in Alzheimer's disease patients, that correlate with TDP-43 pathology burden, but not with amyloid-β or tau deposits. We also demonstrate that processing of the STMN2 pre-mRNA is more sensitive to TDP-43 loss of function than UNC13A. In addition, full-length RNAs encoding STMN2 and UNC13A are suppressed in large RNA-seq datasets generated from Alzheimer's disease post-mortem brain tissue. Collectively, these results open exciting new avenues to use STMN2 and UNC13A as potential therapeutic targets in a broad range of neurodegenerative conditions with TDP-43 proteinopathy including Alzheimer's disease.
Collapse
Affiliation(s)
- Ana Rita Agra Almeida Quadros
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Zhaozhi Li
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xue Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - I Sandra Ndayambaje
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sandeep Aryal
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Nandini Ramesh
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Matthew Nolan
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Rojashree Jayakumar
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yi Han
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hannah Stillman
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Corey Aguilar
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hayden J Wheeler
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Theresa Connors
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jone Lopez-Erauskin
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Michael W Baughn
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Ze'ev Melamed
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Melinda S Beccari
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Laura Olmedo Martínez
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Canori
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Chao-Zong Lee
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura Moran
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Derek H Oakley
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Don W Cleveland
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Bradley T Hyman
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sudeshna Das
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA.
| | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Neurology, MassGeneral Institute for Neurodegenerative Diseases (MIND), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA.
| |
Collapse
|
7
|
Chang K, Ling JP, Redding-Ochoa J, An Y, Li L, Dean SA, Blanchard TG, Pylyukh T, Barrett A, Irwin KE, Moghekar A, Resnick SM, Wong PC, Troncoso JC. Loss of TDP-43 splicing repression occurs early in the aging population and is associated with Alzheimer's disease neuropathologic changes and cognitive decline. Acta Neuropathol 2023; 147:4. [PMID: 38133681 DOI: 10.1007/s00401-023-02653-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 12/23/2023]
Abstract
LATE-NC, the neuropathologic changes of limbic-predominant age-related TAR DNA-binding protein 43 kDa (TDP-43) encephalopathy are frequently associated with Alzheimer's disease (AD) and cognitive impairment in older adults. The association of TDP-43 proteinopathy with AD neuropathologic changes (ADNC) and its impact on specific cognitive domains are not fully understood and whether loss of TDP-43 function occurs early in the aging brain remains unknown. Here, using a large set of autopsies from the Baltimore Longitudinal Study of Aging (BLSA) and another younger cohort, we were able to study brains from subjects 21-109 years of age. Examination of these brains show that loss of TDP-43 splicing repression, as judged by TDP-43 nuclear clearance and expression of a cryptic exon in HDGFL2, first occurs during the 6th decade, preceding by a decade the appearance of TDP-43+ neuronal cytoplasmic inclusions (NCIs). We corroborated this observation using a monoclonal antibody to demonstrate a cryptic exon-encoded neoepitope within HDGFL2 in neurons exhibiting nuclear clearance of TDP-43. TDP-43 nuclear clearance is associated with increased burden of tau pathology. Age at death, female sex, high CERAD neuritic plaque score, and high Braak neurofibrillary stage significantly increase the odds of LATE-NC. Faster rates of cognitive decline on verbal memory (California Verbal Learning Test immediate recall), visuospatial ability (Card Rotations Test), mental status (MMSE) and semantic fluency (Category Fluency Test) were associated with LATE-NC. Notably, the effects of LATE-NC on verbal memory and visuospatial ability are independent of ADNC. However, the effects of TDP-43 nuclear clearance in absence of NCI on the longitudinal trajectories and levels of cognitive measures are not significant. These results establish that loss of TDP-43 splicing repression is an early event occurring in the aging population during the development of TDP-43 proteinopathy and is associated with increased tau pathology. Furthermore, LATE-NC correlates with high levels of ADNC but also has an impact on specific memory and visuospatial functions in aging that is independent of AD.
Collapse
Affiliation(s)
- Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department and Graduate Institute of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, 100225, Taiwan
| | - Jonathan P Ling
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Javier Redding-Ochoa
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Ling Li
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Office of the Chief Medical Examiner, State of Maryland, Baltimore, MD, 21223, USA
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Stephanie A Dean
- Office of the Chief Medical Examiner, State of Maryland, Baltimore, MD, 21223, USA
| | - Thomas G Blanchard
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Tatiana Pylyukh
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Alexander Barrett
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Katherine E Irwin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Philip C Wong
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Juan C Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
8
|
Barnett D, Bohmbach K, Grelot V, Charlet A, Dallérac G, Ju YH, Nagai J, Orr AG. Astrocytes as Drivers and Disruptors of Behavior: New Advances in Basic Mechanisms and Therapeutic Targeting. J Neurosci 2023; 43:7463-7471. [PMID: 37940585 PMCID: PMC10634555 DOI: 10.1523/jneurosci.1376-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/13/2023] [Accepted: 08/22/2023] [Indexed: 11/10/2023] Open
Abstract
Astrocytes are emerging as key regulators of cognitive function and behavior. This review highlights some of the latest advances in the understanding of astrocyte roles in different behavioral domains across lifespan and in disease. We address specific molecular and circuit mechanisms by which astrocytes modulate behavior, discuss their functional diversity and versatility, and highlight emerging astrocyte-targeted treatment strategies that might alleviate behavioral and cognitive dysfunction in pathologic conditions. Converging evidence across different model systems and manipulations is revealing that astrocytes regulate behavioral processes in a precise and context-dependent manner. Improved understanding of these astrocytic functions may generate new therapeutic strategies for various conditions with cognitive and behavioral impairments.
Collapse
Affiliation(s)
- Daniel Barnett
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, New York 10021
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10021
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, New York 10021
| | - Kirsten Bohmbach
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Valentin Grelot
- Institute of Cellular and Integrative Neuroscience, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, 67000, France
| | - Alexandre Charlet
- Institute of Cellular and Integrative Neuroscience, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, 67000, France
| | - Glenn Dallérac
- Centre National de la Recherche Scientifique and Paris-Saclay University, Paris-Saclay Institute for Neurosciences, Paris, 91400, France
| | - Yeon Ha Ju
- Department of Psychiatry and Neuroscience, University of Texas-Austin Dell Medical School, Austin, Texas 78712
| | - Jun Nagai
- RIKEN Center for Brain Science, Laboratory for Glia-Neuron Circuit Dynamics, Saitama, 351-0198, Japan
| | - Anna G Orr
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, New York 10021
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10021
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, New York 10021
| |
Collapse
|
9
|
Nag S, Schneider JA. Limbic-predominant age-related TDP43 encephalopathy (LATE) neuropathological change in neurodegenerative diseases. Nat Rev Neurol 2023; 19:525-541. [PMID: 37563264 PMCID: PMC10964248 DOI: 10.1038/s41582-023-00846-7] [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: 06/26/2023] [Indexed: 08/12/2023]
Abstract
TAR DNA-binding protein 43 (TDP43) is a focus of research in late-onset dementias. TDP43 pathology in the brain was initially identified in amyotrophic lateral sclerosis and frontotemporal lobar degeneration, and later in Alzheimer disease (AD), other neurodegenerative diseases and ageing. Limbic-predominant age-related TDP43 encephalopathy (LATE), recognized as a clinical entity in 2019, is characterized by amnestic dementia resembling AD dementia and occurring most commonly in adults over 80 years of age. Neuropathological findings in LATE, referred to as LATE neuropathological change (LATE-NC), consist of neuronal and glial cytoplasmic TDP43 localized predominantly in limbic areas with or without coexisting hippocampal sclerosis and/or AD neuropathological change and without frontotemporal lobar degeneration or amyotrophic lateral sclerosis pathology. LATE-NC is frequently associated with one or more coexisting pathologies, mainly AD neuropathological change. The focus of this Review is the pathology, genetic risk factors and nature of the cognitive impairments and dementia in pure LATE-NC and in LATE-NC associated with coexisting pathologies. As the clinical and cognitive profile of LATE is currently not easily distinguishable from AD dementia, it is important to develop biomarkers to aid in the diagnosis of this condition in the clinic. The pathogenesis of LATE-NC should be a focus of future research to form the basis for the development of preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Sukriti Nag
- Rush Alzheimer's Disease Center, Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA.
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
10
|
Fang Y, Si X, Wang J, Wang Z, Chen Y, Liu Y, Yan Y, Tian J, Zhang B, Pu J. Alzheimer Disease and Epilepsy: A Mendelian Randomization Study. Neurology 2023; 101:e399-e409. [PMID: 37225432 PMCID: PMC10435057 DOI: 10.1212/wnl.0000000000207423] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/03/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Observational studies suggested a bidirectional relationship between Alzheimer disease (AD) and epilepsies. However, it remains debated whether and in which direction a causal association exists. This study aims to explore the relationship between genetic predisposition to AD, CSF biomarkers of AD (β-amyloid [Aβ] 42 and phosphorylated tau [pTau]), and epilepsies with 2-sample, bidirectional Mendelian randomization (MR) method. METHODS Genetic instruments were obtained from large-scale genome-wide meta-analysis of AD (Ncase/proxy = 111,326, Ncontrol = 677,663), CSF biomarkers of AD (Aβ42 and pTau, N = 13,116), and epilepsy (Ncase = 15,212, Ncontrol = 29,677) of European ancestry. Epilepsy phenotypes included all epilepsy, generalized epilepsy, focal epilepsy, childhood absence epilepsy, juvenile absence epilepsy, juvenile myoclonic epilepsy, generalized epilepsy with tonic-clonic seizures, focal epilepsy with hippocampal sclerosis (focal HS), and lesion-negative focal epilepsy. Main analyses were performed using generalized summary data-based MR. Sensitivity analyses included inverse variance weighted, MR pleiotropy residual sum and outlier, MR-Egger, weighted mode, and weighted median. RESULTS For forward analysis, genetic predisposition to AD was associated with an increased risk of generalized epilepsy (odds ratio [OR] 1.053, 95% CI 1.002-1.105, p = 0.038) and focal HS (OR 1.013, 95% CI 1.004-1.022, p = 0.004). These associations were consistent across sensitivity analyses and replicated using a separate set of genetic instruments from another AD genome-wide association study. For reverse analysis, there was a suggestive effect of focal HS on AD (OR 3.994, 95% CI 1.172-13.613, p = 0.027). In addition, genetically predicted lower CSF Aβ42 was associated with an increased risk of generalized epilepsy (β = 0.090, 95% CI 0.022-0.158, p = 0.010). DISCUSSION This MR study supports a causal link between AD, amyloid pathology, and generalized epilepsy. This study also indicates a close association between AD and focal HS. More effort should be made to screen seizure in AD, unravel its clinical implications, and explore its role as a putative modifiable risk factor.
Collapse
Affiliation(s)
- Yi Fang
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoli Si
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiali Wang
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyun Wang
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Chen
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Liu
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaping Yan
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Tian
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baorong Zhang
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiali Pu
- From the Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
11
|
Vasileva L, Gaynanova G, Valeeva F, Belyaev G, Zueva I, Bushmeleva K, Sibgatullina G, Samigullin D, Vyshtakalyuk A, Petrov K, Zakharova L, Sinyashin O. Mitochondria-Targeted Delivery Strategy of Dual-Loaded Liposomes for Alzheimer's Disease Therapy. Int J Mol Sci 2023; 24:10494. [PMID: 37445673 DOI: 10.3390/ijms241310494] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Liposomes modified with tetradecyltriphenylphosphonium bromide with dual loading of α-tocopherol and donepezil hydrochloride were successfully designed for intranasal administration. Physicochemical characteristics of cationic liposomes such as the hydrodynamic diameter, zeta potential, and polydispersity index were within the range from 105 to 115 nm, from +10 to +23 mV, and from 0.1 to 0.2, respectively. In vitro release curves of donepezil hydrochloride were analyzed using the Korsmeyer-Peppas, Higuchi, First-Order, and Zero-Order kinetic models. Nanocontainers modified with cationic surfactant statistically better penetrate into the mitochondria of rat motoneurons. Imaging of rat brain slices revealed the penetration of nanocarriers into the brain. Experiments on transgenic mice with an Alzheimer's disease model (APP/PS1) demonstrated that the intranasal administration of liposomes within 21 days resulted in enhanced learning abilities and a reduction in the formation rate of Aβ plaques in the entorhinal cortex and hippocampus of the brain.
Collapse
Affiliation(s)
- Leysan Vasileva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Str., 420088 Kazan, Russia
| | - Gulnara Gaynanova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Str., 420088 Kazan, Russia
| | - Farida Valeeva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Str., 420088 Kazan, Russia
| | - Grigory Belyaev
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Str., 420088 Kazan, Russia
| | - Irina Zueva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Str., 420088 Kazan, Russia
| | - Kseniya Bushmeleva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Str., 420088 Kazan, Russia
| | - Guzel Sibgatullina
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, 2/31 Lobachevsky Str., 420111 Kazan, Russia
| | - Dmitry Samigullin
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, 2/31 Lobachevsky Str., 420111 Kazan, Russia
- Institute for Radio-Electronics and Telecommunications, Kazan National Research Technical University Named after A.N. Tupolev-KAI, 10 K. Marx St., 420111 Kazan, Russia
| | - Alexandra Vyshtakalyuk
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Str., 420088 Kazan, Russia
| | - Konstantin Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Str., 420088 Kazan, Russia
| | - Lucia Zakharova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Str., 420088 Kazan, Russia
| | - Oleg Sinyashin
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Str., 420088 Kazan, Russia
| |
Collapse
|
12
|
Licht-Murava A, Meadows SM, Palaguachi F, Song SC, Jackvony S, Bram Y, Zhou C, Schwartz RE, Froemke RC, Orr AL, Orr AG. Astrocytic TDP-43 dysregulation impairs memory by modulating antiviral pathways and interferon-inducible chemokines. SCIENCE ADVANCES 2023; 9:eade1282. [PMID: 37075107 PMCID: PMC10115456 DOI: 10.1126/sciadv.ade1282] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/20/2023] [Indexed: 05/03/2023]
Abstract
Transactivating response region DNA binding protein 43 (TDP-43) pathology is prevalent in dementia, but the cell type-specific effects of TDP-43 pathology are not clear, and therapeutic strategies to alleviate TDP-43-linked cognitive decline are lacking. We found that patients with Alzheimer's disease or frontotemporal dementia have aberrant TDP-43 accumulation in hippocampal astrocytes. In mouse models, induction of widespread or hippocampus-targeted accumulation in astrocytic TDP-43 caused progressive memory loss and localized changes in antiviral gene expression. These changes were cell-autonomous and correlated with impaired astrocytic defense against infectious viruses. Among the changes, astrocytes had elevated levels of interferon-inducible chemokines, and neurons had elevated levels of the corresponding chemokine receptor CXCR3 in presynaptic terminals. CXCR3 stimulation altered presynaptic function and promoted neuronal hyperexcitability, akin to the effects of astrocytic TDP-43 dysregulation, and blockade of CXCR3 reduced this activity. Ablation of CXCR3 also prevented TDP-43-linked memory loss. Thus, astrocytic TDP-43 dysfunction contributes to cognitive impairment through aberrant chemokine-mediated astrocytic-neuronal interactions.
Collapse
Affiliation(s)
- Avital Licht-Murava
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Samantha M. Meadows
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Fernando Palaguachi
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Soomin C. Song
- Skirball Institute, Neuroscience Institute, Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA
| | - Stephanie Jackvony
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Yaron Bram
- Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Constance Zhou
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Medicine–Rockefeller–Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY USA
| | - Robert E. Schwartz
- Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Robert C. Froemke
- Skirball Institute, Neuroscience Institute, Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA
| | - Adam L. Orr
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Anna G. Orr
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Medicine–Rockefeller–Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY USA
| |
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: 5] [Impact Index Per Article: 5.0] [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
|
Nelson PT, Lee EB, Cykowski MD, Alafuzoff I, Arfanakis K, Attems J, Brayne C, Corrada MM, Dugger BN, Flanagan ME, Ghetti B, Grinberg LT, Grossman M, Grothe MJ, Halliday GM, Hasegawa M, Hokkanen SRK, Hunter S, Jellinger K, Kawas CH, Keene CD, Kouri N, Kovacs GG, Leverenz JB, Latimer CS, Mackenzie IR, Mao Q, McAleese KE, Merrick R, Montine TJ, Murray ME, Myllykangas L, Nag S, Neltner JH, Newell KL, Rissman RA, Saito Y, Sajjadi SA, Schwetye KE, Teich AF, Thal DR, Tomé SO, Troncoso JC, Wang SHJ, White CL, Wisniewski T, Yang HS, Schneider JA, Dickson DW, Neumann M. LATE-NC staging in routine neuropathologic diagnosis: an update. Acta Neuropathol 2023; 145:159-173. [PMID: 36512061 PMCID: PMC9849315 DOI: 10.1007/s00401-022-02524-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022]
Abstract
An international consensus report in 2019 recommended a classification system for limbic-predominant age-related TDP-43 encephalopathy neuropathologic changes (LATE-NC). The suggested neuropathologic staging system and nomenclature have proven useful for autopsy practice and dementia research. However, some issues remain unresolved, such as cases with unusual features that do not fit with current diagnostic categories. The goal of this report is to update the neuropathologic criteria for the diagnosis and staging of LATE-NC, based primarily on published data. We provide practical suggestions about how to integrate available genetic information and comorbid pathologies [e.g., Alzheimer's disease neuropathologic changes (ADNC) and Lewy body disease]. We also describe recent research findings that have enabled more precise guidance on how to differentiate LATE-NC from other subtypes of TDP-43 pathology [e.g., frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS)], and how to render diagnoses in unusual situations in which TDP-43 pathology does not follow the staging scheme proposed in 2019. Specific recommendations are also made on when not to apply this diagnostic term based on current knowledge. Neuroanatomical regions of interest in LATE-NC are described in detail and the implications for TDP-43 immunohistochemical results are specified more precisely. We also highlight questions that remain unresolved and areas needing additional study. In summary, the current work lays out a number of recommendations to improve the precision of LATE-NC staging based on published reports and diagnostic experience.
Collapse
Affiliation(s)
- Peter T Nelson
- University of Kentucky, Rm 575 Todd Building, Lexington, KY, USA.
| | - Edward B Lee
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Konstantinos Arfanakis
- Rush University Medical Center, Chicago, IL, USA
- Illinois Institute of Technology, Chicago, IL, USA
| | | | | | | | | | | | | | | | | | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología Y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | | | - Masato Hasegawa
- Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | | | | | | | | | | | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Laboratory Medicine Program, University Health Network, Toronto, Canada
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | | | | | | | - Qinwen Mao
- University of Utah, Salt Lake City, UT, USA
| | | | | | | | | | - Liisa Myllykangas
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sukriti Nag
- Rush University Medical Center, Chicago, IL, USA
| | - Janna H Neltner
- University of Kentucky, Rm 575 Todd Building, Lexington, KY, USA
| | | | | | - Yuko Saito
- Tokyo Metropolitan Geriatric Hospital & Institute of Gerontology, Tokyo, Japan
| | | | | | | | - Dietmar R Thal
- Laboratory for Neuropathology, Department of Imaging and Pathoogy, and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Sandra O Tomé
- Laboratory for Neuropathology, Department of Imaging and Pathoogy, and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | | | | | - Charles L White
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Hyun-Sik Yang
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, BostonBoston, MAMA, USA
| | | | | | | |
Collapse
|
15
|
Sekar D, Tusubira D, Ross K. TDP-43 and NEAT long non-coding RNA: Roles in neurodegenerative disease. Front Cell Neurosci 2022; 16:954912. [PMID: 36385948 PMCID: PMC9650703 DOI: 10.3389/fncel.2022.954912] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/11/2022] [Indexed: 01/03/2023] Open
Abstract
Understanding and ameliorating neurodegenerative diseases represents a key challenge for supporting the health span of the aging population. Diverse protein aggregates have been implicated in such neurodegenerative disorders, including amyloid-β, α-synuclein, tau, fused in sarcoma (FUS), and transactivation response element (TAR) DNA-binding protein 43 (TDP-43). Recent years have seen significant growth in our mechanistic knowledge of relationships between these proteins and some of the membrane-less nuclear structures that fulfill key roles in the cell function. These include the nucleolus, nuclear speckles, and paraspeckles. The ability of macromolecular protein:RNA complexes to partition these nuclear condensates through biophysical processes that involve liquid-liquid phase separation (LLPS) has also gained attention recently. The paraspeckle, which is scaffolded by the architectural long-non-coding RNA nuclear enriched abundant transcript 1 (NEAT1) plays central roles in RNA processing and metabolism and has been linked dynamically to TDP-43. In this mini-review, we outline essential early and recent insights in relation to TDP-43 proteinopathies. We then appraise the relationships between TDP-43 and NEAT1 in the context of neuronal paraspeckles and neuronal stress. We highlight key areas for investigation based on recent advances in our understanding of how TDP-43 affects neuronal function, especially in relation to messenger ribosomal nucleic acid (mRNA) splicing. Finally, we offer perspectives that should be considered for translational pipelines in order to improve health outcomes for the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Durairaj Sekar
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Deusdedit Tusubira
- Department of Biochemistry, Mbarara University of Science and Technology, Mbarara, Uganda,*Correspondence: Deusdedit Tusubira, ; orcid.org/0000-0002-4698-424X
| | - Kehinde Ross
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom,Institute for Health Research, Liverpool John Moores University, Liverpool, United Kingdom,Kehinde Ross, ; orcid.org/0000-0003-0252-1152
| |
Collapse
|
16
|
Laudanski K, Liu D, Okeke T, Restrepo M, Szeto WY. Persistent Depletion of Neuroprotective Factors Accompanies Neuroinflammatory, Neurodegenerative, and Vascular Remodeling Spectra in Serum Three Months after Non-Emergent Cardiac Surgery. Biomedicines 2022; 10:2364. [PMID: 36289630 PMCID: PMC9598177 DOI: 10.3390/biomedicines10102364] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/24/2022] Open
Abstract
We hypothesized that the persistent depletion of neuroprotective markers accompanies neuroinflammation and neurodegeneration in patients after cardiac surgery. A total of 158 patients underwent elective heart surgery with their blood collected before surgery (tbaseline) and 24 h (t24hr), seven days (t7d), and three months (t3m) post-surgery. The patients' serum was measured for markers of neurodegeneration (τau, τaup181-183, amyloid β1-40/β2-42, and S100), atypical neurodegeneration (KLK6 and NRGN), neuro-injury (neurofilament light/heavy, UC-HL, and GFAP), neuroinflammation (YKL-40 and TDP-43), peripheral nerve damage (NCAM-1), neuroprotection (apoE4, BDNF, fetuin, and clusterin), and vascular smoldering inflammation (C-reactive protein, CCL-28 IL-6, and IL-8). The mortality at 28 days, incidence of cerebrovascular accidents (CVA), and functional status were followed for three months. The levels of amyloid β1-40/β1-42 and NF-L were significantly elevated at all time points. The levels of τau, S100, KLK6, NRGN, and NCAM-1 were significantly elevated at 24 h. A cluster analysis demonstrated groupings around amyloids, KLK6, and NCAM-1. YKL-40, but not TDP-43, was significantly elevated across all time points. BDNF, apoE4, fetuin, and clusterin levels were significantly diminished long-term. IL-6 and IL-8 levles returned to baseline at t3m. The levels of CRP, CCL-28, and Hsp-70 remained elevated. At 3 months, 8.2% of the patients experienced a stroke, with transfusion volume being a significant variable. Cardiac-surgery patients exhibited persistent peripheral and neuronal inflammation, blood vessel remodeling, and the depletion of neuroprotective factors 3 months post-procedure.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Leonard Davis Institute for Health Economics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Da Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110055, China
| | - Tony Okeke
- Department of Bioengineering, Drexel University, Philadelphia, PA 19104, USA
| | - Mariana Restrepo
- College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wilson Y. Szeto
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
17
|
Chiang WC, Fang YS, Lye YS, Weng TY, Ganesan K, Huang SH, Chang LY, Chou SC, Chen YR. Hyperphosphorylation-Mimetic TDP-43 Drives Amyloid Formation and Possesses Neuronal Toxicity at the Oligomeric Stage. ACS Chem Neurosci 2022; 13:2599-2612. [PMID: 36007056 DOI: 10.1021/acschemneuro.1c00873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
TDP-43 proteinopathies cover a range of neurodegenerative diseases, including frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Hyperphosphorylated TDP-43 was found within the inclusion bodies in disease lesions; however, the role of hyperphosphorylation and the toxic species are still ambiguous. To characterize the hyperphosphorylation effect of TDP-43, here, we employed five serine mutations implicated in the diseases at serine locations 379, 403, 404, 409, and 410 in the C-terminus to aspartate (S5D) and to alanine (S5A). We systematically characterized the conformation, liquid-liquid phase separation, oligomerization, and fibrillization of TDP-43 variants. Results revealed that the recombinant TDP-43 variants readily formed structurally similar spherical oligomers, as evidenced by circular dichroism spectroscopy, fluorescence spectroscopy, the TDP-43 oligomer-specific antibody assay, dynamic light scattering, and transmission electron microscopy. After incubation, only the phosphor-mimic S5D TDP-43 formed thioflavin-positive amyloid fibrils, whereas wild-type and S5A TDP-43 formed amorphous aggregates. We also examined membrane disruption, the cytotoxicity of human neuroblastoma, and the synaptic loss of primary neurons induced by oligomers and large aggregates of TDP-43. The results showed that all oligomeric TDP-43 variants were toxic regardless of hyperphosphorylation, but the fibrils and amorphous aggregates were not. Overall, our results demonstrated the hyperphosphorylation effect on fibril formation and the toxicity attributed from TDP-43 oligomers. This study facilitates the understanding and therapeutic development for TDP-43 proteinopathies.
Collapse
Affiliation(s)
- Wan-Chin Chiang
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 11529, Taiwan
| | - Yu-Sheng Fang
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 11529, Taiwan.,Institute of Bioinformatics and Structural Biology, National Tsing Hua University, 101, Kuang-Fu Rd., Sec. 2., Hsinchu 30013, Taiwan
| | - Yuh Shen Lye
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 11529, Taiwan.,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei 11529, Taiwan
| | - Tzu-Yu Weng
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 11529, Taiwan
| | - Kiruthika Ganesan
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 11529, Taiwan
| | - Shih-Han Huang
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 11529, Taiwan
| | - Lan-Yun Chang
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 11529, Taiwan
| | - Shih-Chieh Chou
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 11529, Taiwan
| | - Yun-Ru Chen
- Genomics Research Center, Academia Sinica, 128, Academia Rd., Sec. 2, Nankang District, Taipei 11529, Taiwan.,Institute of Bioinformatics and Structural Biology, National Tsing Hua University, 101, Kuang-Fu Rd., Sec. 2., Hsinchu 30013, Taiwan.,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
18
|
The Rab11-regulated endocytic pathway and BDNF/TrkB signaling: Roles in plasticity changes and neurodegenerative diseases. Neurobiol Dis 2022; 171:105796. [PMID: 35728773 DOI: 10.1016/j.nbd.2022.105796] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/10/2022] [Accepted: 06/14/2022] [Indexed: 02/08/2023] Open
Abstract
Neurons are highly polarized cells that rely on the intracellular transport of organelles. This process is regulated by molecular motors such as dynein and kinesins and the Rab family of monomeric GTPases that together help move cargo along microtubules in dendrites, somas, and axons. Rab5-Rab11 GTPases regulate receptor trafficking along early-recycling endosomes, which is a process that determines the intracellular signaling output of different signaling pathways, including those triggered by BDNF binding to its tyrosine kinase receptor TrkB. BDNF is a well-recognized neurotrophic factor that regulates experience-dependent plasticity in different circuits in the brain. The internalization of the BDNF/TrkB complex results in signaling endosomes that allow local signaling in dendrites and presynaptic terminals, nuclear signaling in somas and dynein-mediated long-distance signaling from axons to cell bodies. In this review, we briefly discuss the organization of the endocytic pathway and how Rab11-recycling endosomes interact with other endomembrane systems. We further expand upon the roles of the Rab11-recycling pathway in neuronal plasticity. Then, we discuss the BDNF/TrkB signaling pathways and their functional relationships with the postendocytic trafficking of BDNF, including axonal transport, emphasizing the role of BDNF signaling endosomes, particularly Rab5-Rab11 endosomes, in neuronal plasticity. Finally, we discuss the evidence indicating that the dysfunction of the early-recycling pathway impairs BDNF signaling, contributing to several neurodegenerative diseases.
Collapse
|
19
|
Farrell C, Mumford P, Wiseman FK. Rodent Modeling of Alzheimer's Disease in Down Syndrome: In vivo and ex vivo Approaches. Front Neurosci 2022; 16:909669. [PMID: 35747206 PMCID: PMC9209729 DOI: 10.3389/fnins.2022.909669] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2022] [Indexed: 12/30/2022] Open
Abstract
There are an estimated 6 million people with Down syndrome (DS) worldwide. In developed countries, the vast majority of these individuals will develop Alzheimer's disease neuropathology characterized by the accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles within the brain, which leads to the early onset of dementia (AD-DS) and reduced life-expectancy. The mean age of onset of clinical dementia is ~55 years and by the age of 80, approaching 100% of individuals with DS will have a dementia diagnosis. DS is caused by trisomy of chromosome 21 (Hsa21) thus an additional copy of a gene(s) on the chromosome must cause the development of AD neuropathology and dementia. Indeed, triplication of the gene APP which encodes the amyloid precursor protein is sufficient and necessary for early onset AD (EOAD), both in people who have and do not have DS. However, triplication of other genes on Hsa21 leads to profound differences in neurodevelopment resulting in intellectual disability, elevated incidence of epilepsy and perturbations to the immune system. This different biology may impact on how AD neuropathology and dementia develops in people who have DS. Indeed, genes on Hsa21 other than APP when in three-copies can modulate AD-pathogenesis in mouse preclinical models. Understanding this biology better is critical to inform drug selection for AD prevention and therapy trials for people who have DS. Here we will review rodent preclinical models of AD-DS and how these can be used for both in vivo and ex vivo (cultured cells and organotypic slice cultures) studies to understand the mechanisms that contribute to the early development of AD in people who have DS and test the utility of treatments to prevent or delay the development of disease.
Collapse
|
20
|
Sirkis DW, Bonham LW, Johnson TP, La Joie R, Yokoyama JS. Dissecting the clinical heterogeneity of early-onset Alzheimer's disease. Mol Psychiatry 2022; 27:2674-2688. [PMID: 35393555 PMCID: PMC9156414 DOI: 10.1038/s41380-022-01531-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 12/14/2022]
Abstract
Early-onset Alzheimer's disease (EOAD) is a rare but particularly devastating form of AD. Though notable for its high degree of clinical heterogeneity, EOAD is defined by the same neuropathological hallmarks underlying the more common, late-onset form of AD. In this review, we describe the various clinical syndromes associated with EOAD, including the typical amnestic phenotype as well as atypical variants affecting visuospatial, language, executive, behavioral, and motor functions. We go on to highlight advances in fluid biomarker research and describe how molecular, structural, and functional neuroimaging can be used not only to improve EOAD diagnostic acumen but also enhance our understanding of fundamental pathobiological changes occurring years (and even decades) before the onset of symptoms. In addition, we discuss genetic variation underlying EOAD, including pathogenic variants responsible for the well-known mendelian forms of EOAD as well as variants that may increase risk for the much more common forms of EOAD that are either considered to be sporadic or lack a clear autosomal-dominant inheritance pattern. Intriguingly, specific pathogenic variants in PRNP and MAPT-genes which are more commonly associated with other neurodegenerative diseases-may provide unexpectedly important insights into the formation of AD tau pathology. Genetic analysis of the atypical clinical syndromes associated with EOAD will continue to be challenging given their rarity, but integration of fluid biomarker data, multimodal imaging, and various 'omics techniques and their application to the study of large, multicenter cohorts will enable future discoveries of fundamental mechanisms underlying the development of EOAD and its varied clinical presentations.
Collapse
Affiliation(s)
- Daniel W Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Luke W Bonham
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Taylor P Johnson
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA.
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
21
|
Abstract
Senile plaques have been studied in postmortem brains for more than 120 years and the resultant knowledge has not only helped us understand the etiology and pathogenesis of Alzheimer disease (AD), but has also pointed to possible modes of prevention and treatment. Within the last 15 years, it has become possible to image plaques in living subjects. This is arguably the single greatest advance in AD research since the identification of the Aβ peptide as the major plaque constituent. The limitations and potentialities of amyloid imaging are still not completely clear but are perhaps best glimpsed through the perspective gained from the accumulated postmortem histological studies. The basic morphological classification of plaques into neuritic, cored and diffuse has been supplemented by sophisticated immunohistochemical and biochemical analyses and increasingly detailed mapping of plaque brain distribution. Changes in plaque classification and staging have in turn contributed to changes in the definition and diagnostic criteria for AD. All of this information continues to be tested by clinicopathological correlations and it is through the insights thereby gained that we will best be able to employ the powerful tool of amyloid imaging.
Collapse
Affiliation(s)
- Thomas G Beach
- From the Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, USA
| |
Collapse
|
22
|
Harutyunyan A, Jones NC, Kwan P, Anderson A. Network Preservation Analysis Reveals Dysregulated Synaptic Modules and Regulatory Hubs Shared Between Alzheimer’s Disease and Temporal Lobe Epilepsy. Front Genet 2022; 13:821343. [PMID: 35309145 PMCID: PMC8926077 DOI: 10.3389/fgene.2022.821343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/20/2022] [Indexed: 01/08/2023] Open
Abstract
Background: There is increased prevalence of epilepsy in patients with Alzheimer’s disease (AD). Although shared pathological and clinical features have been identified, the underlying pathophysiology and cause-effect relationships are poorly understood. We aimed to identify commonly dysregulated groups of genes between these two disorders. Methods: Using publicly available transcriptomic data from hippocampal tissue of patients with temporal lobe epilepsy (TLE), late onset AD and non-AD controls, we constructed gene coexpression networks representing all three states. We then employed network preservation statistics to compare the density and connectivity-based preservation of functional gene modules between TLE, AD and controls and used the difference in significance scores as a surrogate quantifier of module preservation. Results: The majority (>90%) of functional gene modules were highly preserved between all coexpression networks, however several modules identified in the TLE network showed various degrees of preservation in the AD network compared to that of control. Of note, two synaptic signalling-associated modules and two metabolic modules showed substantial gain of preservation, while myelination and immune system-associated modules showed significant loss of preservation. The genes SCN3B and EPHA4 were identified as central regulatory hubs of the highly preserved synaptic signalling-associated module. GABRB3 and SCN2A were identified as central regulatory hubs of a smaller neurogenesis-associated module, which was enriched for multiple epileptic activity and seizure-related human phenotype ontologies. Conclusion: We conclude that these hubs and their downstream signalling pathways are common modulators of synaptic activity in the setting of AD and TLE, and may play a critical role in epileptogenesis in AD.
Collapse
Affiliation(s)
- Anna Harutyunyan
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Nigel C. Jones
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Patrick Kwan
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Alison Anderson
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- *Correspondence: Alison Anderson,
| |
Collapse
|
23
|
Shakir MN, Dugger BN. Advances in Deep Neuropathological Phenotyping of Alzheimer Disease: Past, Present, and Future. J Neuropathol Exp Neurol 2022; 81:2-15. [PMID: 34981115 PMCID: PMC8825756 DOI: 10.1093/jnen/nlab122] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is a neurodegenerative disorder characterized pathologically by the presence of neurofibrillary tangles and amyloid beta (Aβ) plaques in the brain. The disease was first described in 1906 by Alois Alzheimer, and since then, there have been many advancements in technologies that have aided in unlocking the secrets of this devastating disease. Such advancements include improving microscopy and staining techniques, refining diagnostic criteria for the disease, and increased appreciation for disease heterogeneity both in neuroanatomic location of abnormalities as well as overlap with other brain diseases; for example, Lewy body disease and vascular dementia. Despite numerous advancements, there is still much to achieve as there is not a cure for AD and postmortem histological analyses is still the gold standard for appreciating AD neuropathologic changes. Recent technological advances such as in-vivo biomarkers and machine learning algorithms permit great strides in disease understanding, and pave the way for potential new therapies and precision medicine approaches. Here, we review the history of human AD neuropathology research to include the notable advancements in understanding common co-pathologies in the setting of AD, and microscopy and staining methods. We also discuss future approaches with a specific focus on deep phenotyping using machine learning.
Collapse
Affiliation(s)
- Mustafa N Shakir
- From the Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA (MNS, BND)
| | - Brittany N Dugger
- From the Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA (MNS, BND)
| |
Collapse
|
24
|
Wegiel J, Flory M, Kuchna I, Nowicki K, Wegiel J, Ma SY, Zhong N, Bobrowicz TW, de Leon M, Lai F, Silverman WP, Wisniewski T. Developmental deficits and staging of dynamics of age associated Alzheimer's disease neurodegeneration and neuronal loss in subjects with Down syndrome. Acta Neuropathol Commun 2022; 10:2. [PMID: 34983655 PMCID: PMC8728914 DOI: 10.1186/s40478-021-01300-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
The increased life expectancy of individuals with Down syndrome (DS) is associated with increased prevalence of trisomy 21-linked early-onset Alzheimer's disease (EOAD) and dementia. The aims of this study of 14 brain regions including the entorhinal cortex, hippocampus, basal ganglia, and cerebellum in 33 adults with DS 26-72 years of age were to identify the magnitude of brain region-specific developmental neuronal deficits contributing to intellectual deficits, to apply this baseline to identification of the topography and magnitude of neurodegeneration and neuronal and volume losses caused by EOAD, and to establish age-based staging of the pattern of genetically driven neuropathology in DS. Both DS subject age and stage of dementia, themselves very strongly correlated, were strong predictors of an AD-associated decrease of the number of neurons, considered a major contributor to dementia. The DS cohort was subclassified by age as pre-AD stage, with 26-41-year-old subjects with a full spectrum of developmental deficit but with very limited incipient AD pathology, and 43-49, 51-59, and 61-72-year-old groups with predominant prevalence of mild, moderately severe, and severe dementia respectively. This multiregional study revealed a 28.1% developmental neuronal deficit in DS subjects 26-41 years of age and 11.9% AD-associated neuronal loss in DS subjects 43-49 years of age; a 28.0% maximum neuronal loss at 51-59 years of age; and a 11.0% minimum neuronal loss at 61-72 years of age. A total developmental neuronal deficit of 40.8 million neurons and AD-associated neuronal loss of 41.6 million neurons reflect a comparable magnitude of developmental neuronal deficit contributing to intellectual deficits, and AD-associated neuronal loss contributing to dementia. This highly predictable pattern of pathology indicates that successful treatment of DS subjects in the fourth decade of life may prevent AD pathology and functional decline.
Collapse
Affiliation(s)
- Jerzy Wegiel
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Michael Flory
- New York State Institute for Basic Research in Developmental Disabilities (IBR), Staten Island, NY USA
| | - Izabela Kuchna
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Krzysztof Nowicki
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Jarek Wegiel
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Shuang Yong Ma
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Nanbert Zhong
- New York State Institute for Basic Research in Developmental Disabilities (IBR), Staten Island, NY USA
| | | | - Mony de Leon
- Department of Radiology, Weill Cornell Medicine, New York, NY USA
| | - Florence Lai
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Wayne P. Silverman
- Department of Pediatrics, Irvine Medical Center, University of California, Irvine, CA USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Pathology and Psychiatry, NYU Grossman School of Medicine, New York, NY 10016 USA
| |
Collapse
|
25
|
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
|
26
|
Laudanski K, Hajj J, Restrepo M, Siddiq K, Okeke T, Rader DJ. Dynamic Changes in Central and Peripheral Neuro-Injury vs. Neuroprotective Serum Markers in COVID-19 Are Modulated by Different Types of Anti-Viral Treatments but Do Not Affect the Incidence of Late and Early Strokes. Biomedicines 2021; 9:1791. [PMID: 34944606 PMCID: PMC8698659 DOI: 10.3390/biomedicines9121791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 01/07/2023] Open
Abstract
The balance between neurodegeneration, neuroinflammation, neuroprotection, and COVID-19-directed therapy may underly the heterogeneity of SARS-CoV-2's neurological outcomes. A total of 105 patients hospitalized with a diagnosis of COVID-19 had serum collected over a 6 month period to assess neuroinflammatory (MIF, CCL23, MCP-1), neuro-injury (NFL, NCAM-1), neurodegenerative (KLK6, τ, phospho τ, amyloids, TDP43, YKL40), and neuroprotective (clusterin, fetuin, TREM-2) proteins. These were compared to markers of nonspecific inflammatory responses (IL-6, D-dimer, CRP) and of the overall viral burden (spike protein). Data regarding treatment (steroids, convalescent plasma, remdasavir), pre-existing conditions, and incidences of strokes were collected. Amyloid β42, TDP43, NF-L, and KLK6 serum levels declined 2-3 days post-admission, yet recovered to admission baseline levels by 7 days. YKL-40 and NCAM-1 levels remained elevated over time, with clusters of differential responses identified among TREM-2, TDP43, and YKL40. Fetuin was elevated after the onset of COVID-19 while TREM-2 initially declined before significantly increasing over time. MIF serum level was increased 3-7 days after admission. Ferritin correlated with TDP-43 and KLK6. No treatment with remdesivir coincided with elevations in Amyloid-β40. A lack of convalescent plasma resulted in increased NCAM-1 and total tau, and steroidal treatments did not significantly affect any markers. A total of 11 incidences of stroke were registered up to six months after initial admission for COVID-19. Elevated D-dimer, platelet counts, IL-6, and leukopenia were observed. Variable MIF serum levels differentiated patients with CVA from those who did not have a stroke during the acute phase of COVID-19. This study demonstrated concomitant and opposite changes in neurodegenerative and neuroprotective markers persisting well into recovery.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- The Department of Anesthesiology and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jihane Hajj
- School of Nursing, Widener University, Philadelphia, PA 19013, USA;
| | - Mariana Restrepo
- College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Kumal Siddiq
- College of Arts and Sciences, Drexel University, Philadelphia, PA 19104, USA;
| | - Tony Okeke
- School of Biomedical Engineering, Drexel University, Philadelphia, PA 19104, USA;
| | - Daniel J. Rader
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
27
|
Fortea J, Zaman SH, Hartley S, Rafii MS, Head E, Carmona-Iragui M. Alzheimer's disease associated with Down syndrome: a genetic form of dementia. Lancet Neurol 2021; 20:930-942. [PMID: 34687637 PMCID: PMC9387748 DOI: 10.1016/s1474-4422(21)00245-3] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 02/03/2023]
Abstract
Adults with Down syndrome develop the neuropathological hallmarks of Alzheimer's disease and are at very high risk of developing early-onset dementia, which is now the leading cause of death in this population. Diagnosis of dementia remains a clinical challenge because of the lack of validated diagnostic criteria in this population, and because symptoms are overshadowed by the intellectual disability associated with Down syndrome. In people with Down syndrome, fluid and imaging biomarkers have shown good diagnostic performances and a strikingly similar temporality of changes with respect to sporadic and autosomal dominant Alzheimer's disease. Most importantly, there are no treatments to prevent Alzheimer's disease, even though adults with Down syndrome could be an optimal population in whom to conduct Alzheimer's disease prevention trials. Unprecedented research activity in Down syndrome is rapidly changing this bleak scenario that will translate into disease-modifying therapies that could benefit other populations.
Collapse
Affiliation(s)
- Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu y Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Madrid, Spain.
| | - Shahid H Zaman
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK; Cambridgeshire & Peterborough NHS Foundation Trust, Cambridge, UK
| | - Sigan Hartley
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael S Rafii
- Alzheimer's Therapeutic Research Institute (ATRI), Keck School of Medicine, University of Southern California, San Diego, CA, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Maria Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu y Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Madrid, Spain
| |
Collapse
|
28
|
Rujeedawa T, Carrillo Félez E, Clare ICH, Fortea J, Strydom A, Rebillat AS, Coppus A, Levin J, Zaman SH. The Clinical and Neuropathological Features of Sporadic (Late-Onset) and Genetic Forms of Alzheimer's Disease. J Clin Med 2021; 10:4582. [PMID: 34640600 PMCID: PMC8509365 DOI: 10.3390/jcm10194582] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022] Open
Abstract
The purpose of this review is to compare and highlight the clinical and pathological aspects of genetic versus acquired Alzheimer's disease: Down syndrome-associated Alzheimer's disease in (DSAD) and Autosomal Dominant Alzheimer's disease (ADAD) are compared with the late-onset form of the disease (LOAD). DSAD and ADAD present in a younger population and are more likely to manifest with non-amnestic (such as dysexecutive function features) in the prodromal phase or neurological features (such as seizures and paralysis) especially in ADAD. The very large variety of mutations associated with ADAD explains the wider range of phenotypes. In the LOAD, age-associated comorbidities explain many of the phenotypic differences.
Collapse
Affiliation(s)
- Tanzil Rujeedawa
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
| | - Eva Carrillo Félez
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
| | - Isabel C. H. Clare
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
- Cambridgeshire and Peterborough Foundation NHS Trust, Fulbourn CB21 5EF, UK
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, 08029 Barcelona, Spain
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK;
- South London and the Maudsley NHS Foundation Trust, The LonDowns Consortium, London SE5 8AZ, UK
| | | | - Antonia Coppus
- Department for Primary and Community Care, Department of Primary and Community Care (149 ELG), Radboud University Nijmegen Medical Center, P.O. Box 9101, 6525 GA Nijmegen, The Netherlands;
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, 80539 Munich, Germany;
- German Center for Neurodegenerative Diseases, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Shahid H. Zaman
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8PQ, UK; (T.R.); (E.C.F.); (I.C.H.C.)
- Cambridgeshire and Peterborough Foundation NHS Trust, Fulbourn CB21 5EF, UK
| |
Collapse
|
29
|
Patni D, Jha SK. Protonation-Deprotonation Switch Controls the Amyloid-like Misfolding of Nucleic-Acid-Binding Domains of TDP-43. J Phys Chem B 2021; 125:8383-8394. [PMID: 34318672 DOI: 10.1021/acs.jpcb.1c03262] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Nutrient starvation stress acidifies the cytosol and leads to the formation of large protein assemblies and misfolded aggregates. However, how starvation stress is sensed at the molecular level and leads to protein misfolding is poorly understood. TDP-43 is a vital protein, which, under stress-like conditions, associates with stress granule proteins via its functional nucleic-acid-binding domains (TDP-43tRRM) and misfolds to form aberrant aggregates. Here, we show that the monomeric N form of TDP-43tRRM forms a misfolded amyloid-like protein assembly, β form, in a pH-dependent manner and identified the critical protein side-chain residue whose protonation triggers its misfolding. We systematically mutated the three buried ionizable residues, D105, H166, and H256, to neutral amino acids to block the pH-dependent protonation-deprotonation titration of their side chain and studied their effect on the N-to-β transition. We observed that D105A and H256Q resembled TDP-43tRRM in their pH-dependent misfolding behavior. However, H166Q retains the N-like secondary structure under low-pH conditions and does not show pH-dependent misfolding to the β form. These results indicate that H166 is the critical side-chain residue whose protonation triggers the misfolding of TDP-43tRRM and shed light on how stress-induced misfolding of proteins during neurodegeneration could begin from site-specific triggers.
Collapse
Affiliation(s)
- Divya Patni
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Santosh Kumar Jha
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
30
|
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: 3.0] [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
|
31
|
Hunter S, Hokkanen SRK, Keage HAD, Fleming J, Minett T, Polvikoski T, Allinson K, Brayne C. TDP-43 Related Neuropathologies and Phosphorylation State: Associations with Age and Clinical Dementia in the Cambridge City over-75s Cohort. J Alzheimers Dis 2021; 75:337-350. [PMID: 32280087 DOI: 10.3233/jad-191093] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathologies associated with the Tar-DNA binding protein 43 KDa (TDP-43) are associated with neurodegenerative diseases and aging. Phosphorylation of cellular proteins is a well-accepted mechanism of biological control and can be associated with disease pathways. Phosphorylation state associated with TDP-43 associated pathology has not been investigated with respect to dementia status in a population representative sample. TDP-43 immunohistochemistry directed toward phosphorylated (TDP-43P) and unphosphorylated (TDP-43U) was assessed in sections of hippocampus and temporal cortex from 222 brains donated to the population representative Cambridge City over-75s Cohort. Relationships between dementia status and age at death for TDP-43 immunoreactive pathologies by phosphorylation state were investigated. TDP-43 pathologies are common in the oldest old in the population and often do not conform to MacKenzie classification. Increasing age is associated with glial (TDP-43P) and neuronal inclusions (TDP-43P and TDP-43U), neurites, and granulovacuolar degeneration (GVD). Dementia status is associated with GVD and glial (TDP-43 P) and neural inclusions (TDP-43 P and U). Dementia severity was associated with glial (TDP-43P) and neuronal inclusions (TDP-43U and TDP-43P), GVD, and neurites. The associations between dementia severity and both glial cytoplasmic inclusions and GVD were independent from other pathologies and TDP-43 neuronal cytoplasmic inclusions. TDP-43 pathology contributes to dementia status and progression in a variety of ways in different phosphorylation states involving both neurons and glia, independently from age and from classic Alzheimer-related pathologies. TDP-43 pathologies as cytoplasmic inclusions in neurons or glia or as GVD contribute independently to dementia.
Collapse
Affiliation(s)
- Sally Hunter
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Suvi R K Hokkanen
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Hannah A D Keage
- Cognitive Ageing and Impairment Neurosciences, School of Psychology, Social Work and Social Policy, University of South Australia, Adelaide, Australia
| | - Jane Fleming
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Thais Minett
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, Cambridge, UK.,Department of Radiology, University of Cambridge, Cambridge, UK
| | - Tuomo Polvikoski
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Kieren Allinson
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Carol Brayne
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
32
|
Tombini M, Assenza G, Ricci L, Lanzone J, Boscarino M, Vico C, Magliozzi A, Di Lazzaro V. Temporal Lobe Epilepsy and Alzheimer's Disease: From Preclinical to Clinical Evidence of a Strong Association. J Alzheimers Dis Rep 2021; 5:243-261. [PMID: 34113782 PMCID: PMC8150253 DOI: 10.3233/adr-200286] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Increasing evidence coming from both experimental and humans' studies strongly suggest the existence of a link between epilepsy, in particular temporal lobe epilepsy (TLE), and Alzheimer's disease (AD). Patients with mild cognitive impairment and AD are more prone to have seizures, and seizures seem to facilitate amyloid-β and tau deposits, thus promoting neurodegenerative processes. Consistent with this view, long-lasting drug-resistant TLE and AD have been shown to share several pathological and neuroimaging features. Even if studies addressing prevalence of interictal and subclinical epileptiform activity in these patients are not yet conclusive, their findings raise the possibility that epileptiform activity might negatively impact memory and hasten cognitive decline, either directly or by association with unrecognized silent seizures. In addition, data about detrimental effect of network hyperexcitability in temporal regions in the premorbid and early stages ofADopen up newtherapeutic opportunities for antiseizure medications and/or antiepileptic strategies that might complement or enhance existing therapies, and potentially modify disease progression. Here we provide a review of evidence linking epileptiform activity, network hyperexcitability, and AD, and their role promoting and accelerating neurodegenerative process. Finally, the effects of antiseizure medications on cognition and their optimal administration in patients with AD are summarized.
Collapse
Affiliation(s)
- Mario Tombini
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Giovanni Assenza
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Lorenzo Ricci
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Jacopo Lanzone
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Marilisa Boscarino
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Carlo Vico
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Alessandro Magliozzi
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| |
Collapse
|
33
|
Spina S, La Joie R, Petersen C, Nolan AL, Cuevas D, Cosme C, Hepker M, Hwang JH, Miller ZA, Huang EJ, Karydas AM, Grant H, Boxer AL, Gorno-Tempini ML, Rosen HJ, Kramer JH, Miller BL, Seeley WW, Rabinovici GD, Grinberg LT. Comorbid neuropathological diagnoses in early versus late-onset Alzheimer's disease. Brain 2021; 144:2186-2198. [PMID: 33693619 DOI: 10.1093/brain/awab099] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/21/2020] [Accepted: 01/03/2021] [Indexed: 12/14/2022] Open
Abstract
Copathologies play an important role in the expression of the AD clinical phenotype and may influence treatment efficacy. Early-onset AD (EOAD), defined as manifesting before age 65, is viewed as a relatively pure form of AD with a more homogeneous neuropathological substrate. We sought to compare the frequency of common neuropathological diagnoses in a consecutive autopsy series of 96 patients with EOAD (median age of onset = 55 years, 44 females) and 48 with late-onset AD (LOAD) (median age of onset = 73 years, 14 females). The UCSF Neurodegenerative Disease Brain Bank database was reviewed to identify patients with a primary pathological diagnosis of AD. Prevalence and stage of Lewy body disease (LBD), limbic age-related TDP-43 encephalopathy (LATE), argyrophilic grain disease (AGD), hippocampal sclerosis (HS), cerebral amyloid angiopathy (CAA), and vascular brain injury (VBI) were compared between the two cohorts. We found at least one non-AD pathological diagnosis in 98% of patients with EOAD (versus 100% of LOAD), and the number of comorbid diagnoses per patient was lower in EOAD than in LOAD (median=2 versus 3, Mann-Whitney Z = 3.00, p = 0.002). LBD and CAA were common in both EOAD and LOAD (CAA: 86% versus 79%, Fisher exact p = 0.33; LBD: 49% versus 42%, p = 0.48, respectively), although amygdala-predominant LBD was more commonly found in EOAD than LOAD (22% versus 6%, p = 0.02). In contrast, LATE (35% versus 8%, p < 0.001), HS (15% versus 3%, p = 0.02), AGD (58% versus 41%, p = 0.052), and VBI (65% versus 39%, p = 0.004) were more common in LOAD than EOAD, respectively. The number of copathologies predicted worse cognitive performance at the time of death on MMSE (1.4 points/pathology (95%CI [-2.5, -0.2]) and Clinical Dementia Rating - Sum of Boxes (1.15 point/pathology, 95%CI [0.45, 1.84]), across the EOAD and the LOAD cohorts. The effect of sex on the number of copathologies was not significant (p = 0.17). Prevalence of at least one APOE ε4 allele was similar across the two cohorts (52% and 54%) and was associated with a greater number of copathologies (+0.40, 95%CI [0.01, 0.79], p = 0.047), independent of age of symptom onset, sex, and disease duration. Females showed higher density of neurofibrillary tangles compared to men, controlling for age of onset, APOE ε4, and disease duration. Our findings suggest that non-AD pathological diagnoses play an important role in the clinical phenotype of EOAD with potentially significant implications for clinical practice and clinical trials design.
Collapse
Affiliation(s)
- Salvatore Spina
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Cathrine Petersen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Amber L Nolan
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Deion Cuevas
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Celica Cosme
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mackenzie Hepker
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Ji-Hye Hwang
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Zachary A Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Eric J Huang
- Department of Pathology; University of California, San Francisco, San Francisco, CA, USA
| | - Anna M Karydas
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Harli Grant
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Howard J Rosen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,Department of Pathology; University of California, San Francisco, San Francisco, CA, USA
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,Department of Radiology and Biomedical Imaging; University of California, San Francisco, San Francisco, CA, USA
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,Department of Pathology; University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
34
|
Laos V, Bishop D, Ganguly P, Schonfeld G, Trapp E, Cantrell KL, Buratto SK, Shea JE, Bowers MT. Catalytic Cross Talk between Key Peptide Fragments That Couple Alzheimer's Disease with Amyotrophic Lateral Sclerosis. J Am Chem Soc 2021; 143:3494-3502. [PMID: 33621087 DOI: 10.1021/jacs.0c12729] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Protein aggregation is a common feature in prominent neurodegenerative diseases, usually thought to be due to the assembly of a single peptide or protein. Recent studies have challenged this notion and suggested several proteins may be involved in promoting and amplifying disease. For example, the TDP-43 protein associated with Amyotrophic Lateral Sclerosis has been found in the brain along with Aβ assemblies associated with Alzheimer's disease, and those patients that show the presence of TDP-43 are 10 times more likely to demonstrate cognitive impairment compared to TDP-43-negative Alzheimer's patients. Here we examine the interactions between the amyloidogenic core of TDP-43, TDP-43307-319, and a neurotoxic physiologically observed fragment of Aβ, Aβ25-35. Utilizing ion mobility mass spectrometry in concert with atomic force microscopy and molecular dynamics simulations, we investigate which oligomers are involved in seeding aggregation across these two different protein systems and gain insight into which structures initiate and result from these interactions. Studies were conducted by mixing Aβ25-35 with the toxic wild type TDP-43307-319 peptide and with the nontoxic synthetic TDP-43307-319 mutant, G314V. Our findings identify a strong catalytic effect of TDP-43307-319 WT monomer in the acceleration of Aβ25-35 aggregation to its toxic cylindrin and β barrel forms. This observation is unprecedented in both its speed and specificity. Interestingly, the nontoxic G314V mutant of TDP-43307-319 and dimers or higher order oligomers of WT TDP-43307-319 do not promote aggregation of Aβ25-35 but rather dissociate preformed toxic higher order oligomers of Aβ25-35. Reasons for these very different behaviors are reported.
Collapse
Affiliation(s)
- Veronica Laos
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Dezmond Bishop
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Pritam Ganguly
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Grace Schonfeld
- Department of Chemistry, Westmont College, Santa Barbara, California 93108, United States
| | - Ellen Trapp
- Department of Chemistry, Westmont College, Santa Barbara, California 93108, United States
| | - Kristi Lazar Cantrell
- Department of Chemistry, Westmont College, Santa Barbara, California 93108, United States
| | - Steven K Buratto
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Joan-Emma Shea
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Michael T Bowers
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
35
|
Gicas KM, Honer WG, Wilson RS, Boyle PA, Leurgans SE, Schneider JA, Bennett DA. Association of serial position scores on memory tests and hippocampal-related neuropathologic outcomes. Neurology 2020; 95:e3303-e3312. [PMID: 33144516 PMCID: PMC7836661 DOI: 10.1212/wnl.0000000000010952] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 08/17/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether serial position scores in verbal memory differentiate hippocampal-related neuropathologic outcomes, we examined these associations in a sample of older adults without dementia who underwent autopsy. METHODS We used data from the Rush Memory and Aging Project, a longitudinal clinical-pathologic cohort study of community-dwelling adults. A total of 701 participants (mean age 82.7, 71.2% female) completed baseline cognitive evaluations and underwent brain autopsy to identify pathologic Alzheimer disease (AD), TDP-43 inclusions (defining limbic-predominant age-related TDP-43 encephalopathy [LATE]), and hippocampal sclerosis. The Consortium to Establish a Registry for Alzheimer's Disease word list memory test immediate recall trials provided serial position scores, which index the proportion of words recalled from the beginning (primacy scores) and end (recency scores) of a word list. Binary and ordinal logistic regressions examined associations between serial position scores and neuropathologic outcomes. Secondary outcomes included Alzheimer dementia and mild cognitive impairment proximate to death. RESULTS Primacy and recency scores were uncorrelated (r = 0.07). Each SD of better primacy score was associated with lower likelihood of neuropathologic changes (24% lower LATE, 31% lower pathologic AD, 37% lower hippocampal sclerosis). For pathologic AD, better baseline primacy scores were associated with a 36% lower likelihood of comorbidity with LATE or hippocampal sclerosis. Primacy scores better discriminated between clinical diagnoses proximate to death, including those with mild cognitive impairment compared to no impairment. Recency scores showed weaker or no associations. CONCLUSIONS Primacy scores may be particularly sensitive markers of AD and related hippocampal neuropathologies. The differential predictive value of serial position scores suggests they offer complementary information about disease outcomes in addition to the routinely used total recall scores.
Collapse
Affiliation(s)
- Kristina M Gicas
- From the Department of Psychology (K.M.G.), York University, Toronto; Department of Psychiatry (W.G.H.), University of British Columbia, Vancouver Canada; and Departments of Neurological Sciences (R.S.W., S.E.L., D.A.B.), Psychiatry and Behavioral Sciences (R.S.W., P.A.B.), and Pathology (J.A.S.), Rush University Medical Center, Chicago, IL.
| | - William G Honer
- From the Department of Psychology (K.M.G.), York University, Toronto; Department of Psychiatry (W.G.H.), University of British Columbia, Vancouver Canada; and Departments of Neurological Sciences (R.S.W., S.E.L., D.A.B.), Psychiatry and Behavioral Sciences (R.S.W., P.A.B.), and Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Robert S Wilson
- From the Department of Psychology (K.M.G.), York University, Toronto; Department of Psychiatry (W.G.H.), University of British Columbia, Vancouver Canada; and Departments of Neurological Sciences (R.S.W., S.E.L., D.A.B.), Psychiatry and Behavioral Sciences (R.S.W., P.A.B.), and Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Patricia A Boyle
- From the Department of Psychology (K.M.G.), York University, Toronto; Department of Psychiatry (W.G.H.), University of British Columbia, Vancouver Canada; and Departments of Neurological Sciences (R.S.W., S.E.L., D.A.B.), Psychiatry and Behavioral Sciences (R.S.W., P.A.B.), and Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Sue E Leurgans
- From the Department of Psychology (K.M.G.), York University, Toronto; Department of Psychiatry (W.G.H.), University of British Columbia, Vancouver Canada; and Departments of Neurological Sciences (R.S.W., S.E.L., D.A.B.), Psychiatry and Behavioral Sciences (R.S.W., P.A.B.), and Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - Julie A Schneider
- From the Department of Psychology (K.M.G.), York University, Toronto; Department of Psychiatry (W.G.H.), University of British Columbia, Vancouver Canada; and Departments of Neurological Sciences (R.S.W., S.E.L., D.A.B.), Psychiatry and Behavioral Sciences (R.S.W., P.A.B.), and Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| | - David A Bennett
- From the Department of Psychology (K.M.G.), York University, Toronto; Department of Psychiatry (W.G.H.), University of British Columbia, Vancouver Canada; and Departments of Neurological Sciences (R.S.W., S.E.L., D.A.B.), Psychiatry and Behavioral Sciences (R.S.W., P.A.B.), and Pathology (J.A.S.), Rush University Medical Center, Chicago, IL
| |
Collapse
|
36
|
Robinson AC, Chew-Graham S, Davidson YS, Horan MA, Roncaroli F, Minshull J, du Plessis D, Pal P, Payton A, Pendleton N, Mann DMA. A Comparative Study of Pathological Outcomes in The University of Manchester Longitudinal Study of Cognition in Normal Healthy Old Age and Brains for Dementia Research Cohorts. J Alzheimers Dis 2020; 73:619-632. [PMID: 31796669 PMCID: PMC7029329 DOI: 10.3233/jad-190580] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In the present study, we have characterized and compared individuals whose brains were donated as part of The University of Manchester Longitudinal Study of Cognition in Normal Healthy Old Age (UoM) with those donated through the Manchester arm of the UK Brains for Dementia Research (BDR) program. The aim of this study was to investigate how differences in study recruitment may affect final pathological composition of cohort studies. The UoM cohort was established as a longitudinal study of aging and cognition whereas the BDR program was established, prima facie, to collect brains from both demented and non-demented individuals for the purpose of building a tissue research resource. Consequently, the differences in recruitment patterns generated differences in demographic, clinical, and neuropathological characteristics. There was a higher proportion of recruits with dementia [mostly Alzheimer's disease (AD)] within the BDR cohort than in the UoM cohort. In pathological terms, the BDR cohort was more 'polarized', being more composed of demented cases with high Braak pathology scores and non-demented cases with low Braak scores, and fewer non-AD pathology cases, than the UoM cohort. In both cohorts, cerebral amyloid angiopathy tended to be greater in demented than non-demented individuals. Such observations partly reflect the recruitment of demented and non-demented individuals into the BDR cohort, and also that insufficient study time may have elapsed for disease onset and development in non-demented individuals to take place. Conversely, in the UoM cohort, where there had been nearly 30 years of study time, a broader spread of AD-type pathological changes had 'naturally' evolved in the brains of both demented and non-demented participants.
Collapse
Affiliation(s)
- Andrew C Robinson
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience & Experimental Psychology, The University of Manchester, Salford Royal Hospital, Salford, UK
| | - Stephen Chew-Graham
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience & Experimental Psychology, The University of Manchester, Salford Royal Hospital, Salford, UK
| | - Yvonne S Davidson
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience & Experimental Psychology, The University of Manchester, Salford Royal Hospital, Salford, UK
| | - Michael A Horan
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience & Experimental Psychology, The University of Manchester, Salford Royal Hospital, Salford, UK
| | - Federico Roncaroli
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience & Experimental Psychology, The University of Manchester, Salford Royal Hospital, Salford, UK.,Neuropathology Unit, Salford Royal Hospital, Greater Manchester, England, UK
| | - James Minshull
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience & Experimental Psychology, The University of Manchester, Salford Royal Hospital, Salford, UK
| | - Daniel du Plessis
- Neuropathology Unit, Salford Royal Hospital, Greater Manchester, England, UK
| | - Piyali Pal
- Department of Neuropathology, Walton Centre for Neurology and Neurosurgery, Liverpool, UK
| | - Antony Payton
- Faculty of Biology, Medicine and Health, School of Health Sciences, Division of Informatics, Imaging & Data Sciences, The University of Manchester, Manchester, UK
| | - Neil Pendleton
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience & Experimental Psychology, The University of Manchester, Salford Royal Hospital, Salford, UK
| | - David M A Mann
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience & Experimental Psychology, The University of Manchester, Salford Royal Hospital, Salford, UK
| |
Collapse
|
37
|
Deng Y, Zhang J, Sun X, Ma G, Luo G, Miao Z, Song L. miR-132 improves the cognitive function of rats with Alzheimer's disease by inhibiting the MAPK1 signal pathway. Exp Ther Med 2020; 20:159. [PMID: 33093897 PMCID: PMC7571341 DOI: 10.3892/etm.2020.9288] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a common worldwide progressive neurodegenerative disease. The dysregulation of miRNA is crucial in neurodegenerative diseases and neuron apoptosis during AD and is closely associated with the MAPK pathway. By bioinformatic website, we found that there was target inhibiting relationship between microRNA (miR)-132 and MAPK1. Therefore, the current study speculated that miR-132 could improve the cognitive function of rats with AD by inhibiting MAPK1 expression. To verify our hypothesis, 10 normal rats and 60 rats with AD were selected and divided into model, Ad-miR-132 negative control (NC), Ad-miR-132, Ad-small interfering (si)MAPK1 NC, Ad-siMAPK1 and Ad-miR-132 + Ad-MAPK1 groups. Rats were evaluated for learning by performing morris water maze tests and pathological changes of the hippocampus were assessed via HE staining. Additionally, hippocampus cell apoptosis was determined using a TUNEL assay and levels of acetylcholinesterase (AChE), reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were evaluated in sera via ELISA. The mRNA and protein expression of miR-132, iNOS, MAPK1 and phosphorylated (p)-MAPK1 was determined in hippocampus tissues via reverse transcription-quantitative PCR and western blotting, respectively. Compared with normal mice, rats with AD had significantly decreased learning abilities, increased cell apoptosis rates, increased levels of AChE, iNOS, ROS, MDA, MAPK1 and p-MAPK1 and decreased levels of SOD, GSH-Px and miR-132. Upregulation of miR-132 group improved the above indictors and silencing MAKP1 worsened the condition of rats. miR-132 upregulation therefore reversed the negative effects caused by MAPK1 silencing in rats with AD. In conclusion, miR-132 inhibited hippocampal iNOS expression and oxidative stress by inhibiting MAPK1expression to improve the cognitive function of rats with AD.
Collapse
Affiliation(s)
- Yiming Deng
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China.,China National Clinical Research Center for Neurological Diseases, Beijing 100070, P.R. China
| | - Jingyu Zhang
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China.,China National Clinical Research Center for Neurological Diseases, Beijing 100070, P.R. China
| | - Xuan Sun
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China.,China National Clinical Research Center for Neurological Diseases, Beijing 100070, P.R. China
| | - Gaoting Ma
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China.,China National Clinical Research Center for Neurological Diseases, Beijing 100070, P.R. China
| | - Gang Luo
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China.,China National Clinical Research Center for Neurological Diseases, Beijing 100070, P.R. China
| | - Zhongrong Miao
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China.,China National Clinical Research Center for Neurological Diseases, Beijing 100070, P.R. China
| | - Ligang Song
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China.,China National Clinical Research Center for Neurological Diseases, Beijing 100070, P.R. China
| |
Collapse
|
38
|
McAleese KE, Walker L, Erskine D, Johnson M, Koss D, Thomas AJ, Attems J. Concomitant LATE-NC in Alzheimer's disease is not associated with increased tau or amyloid-β pathological burden. Neuropathol Appl Neurobiol 2020; 46:722-734. [PMID: 32896913 DOI: 10.1111/nan.12664] [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] [Received: 06/14/2020] [Revised: 07/23/2020] [Accepted: 08/22/2020] [Indexed: 12/14/2022]
Abstract
AIMS Limbic-predominant age-related TDP-43 encephalopathy neuropathological change (LATE-NC) is present in approximately 50% of Alzheimer's disease (AD) cases and is associated with accelerated cognitive decline. Studies indicate a potential synergistic relationship between LATE-NC and hyperphosphorylated tau. It is unknown if LATE-NC is an independent driver of cognitive impairment or exerts its influence through synergistic relationships with tau. This cliniconeuropathological study investigated the impact of LATE-NC on quantified measures of AD-associated pathology and its impact on clinical measures. METHODS A total of 61 AD cases underwent neuropathological assessment for LATE-NC and quantitative assessment [area covered by immunoreactivity (IR)] for early conformational tau (MC-1), late-stage hyperphosphorylated tau (AT8) and amyloid-β in the amygdala and five neocortical regions. Clinical measures included age of disease onset, final Mini-Mental State Examination (MMSE) score and rate of cognitive decline. RESULTS LATE-NC was present in 41 AD cases (AD/LATE-NC; 67.2%). No significant differences in MC-1-IR, AT8-IR or 4G8-IR were observed in any region between AD/LATE-NC and AD without LATE-NC, indicating no accelerated aggregation or hyperphosphorylation of tau proteins in the AD/LATE-NC cases. Final MMSE was significantly lower in AD/LATE-NC cases and was significantly associated with LATE-NC score even when controlled for the presence of both MC-1-IR and AT8-IR (P = 0.009). CONCLUSION The presence of LATE-NC in AD is not associated with an increase in the burden of early or late tau or Aβ pathology. LATE-NC is associated with a lower final MMSE score independent of tau pathology.
Collapse
Affiliation(s)
- K E McAleese
- Translation and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - L Walker
- Translation and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - D Erskine
- Translation and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - M Johnson
- Translation and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - D Koss
- Translation and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - A J Thomas
- Translation and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - J Attems
- Translation and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
39
|
Thomas DX, Bajaj S, McRae-McKee K, Hadjichrysanthou C, Anderson RM, Collinge J. Association of TDP-43 proteinopathy, cerebral amyloid angiopathy, and Lewy bodies with cognitive impairment in individuals with or without Alzheimer's disease neuropathology. Sci Rep 2020; 10:14579. [PMID: 32883971 PMCID: PMC7471113 DOI: 10.1038/s41598-020-71305-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 07/14/2020] [Indexed: 11/08/2022] Open
Abstract
Alzheimer's disease patients typically present with multiple co-morbid neuropathologies at autopsy, but the impact of these pathologies on cognitive impairment during life is poorly understood. In this study, we developed cognitive trajectories for patients with common co-pathologies in the presence and absence of Alzheimer's disease neuropathology. Cognitive trajectories were modelled in a Bayesian hierarchical regression framework to estimate the effects of each neuropathology on cognitive decline as assessed by the mini-mental state examination and the clinical dementia rating scale sum of boxes scores. We show that both TDP-43 proteinopathy and cerebral amyloid angiopathy associate with cognitive impairment of similar magnitude to that associated with Alzheimer's disease neuropathology. Within our study population, 63% of individuals given the 'gold-standard' neuropathological diagnosis of Alzheimer's disease in fact possessed either TDP-43 proteinopathy or cerebral amyloid angiopathy of sufficient severity to independently explain the majority of their cognitive impairment. This suggests that many individuals diagnosed with Alzheimer's disease may actually suffer from a mixed dementia, and therapeutics targeting only Alzheimer's disease-related processes may have severely limited efficacy in these co-morbid populations.
Collapse
Affiliation(s)
- David X Thomas
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, W1W 7FF, UK.
| | - Sumali Bajaj
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, W2 1PG, UK
| | - Kevin McRae-McKee
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, W2 1PG, UK
| | - Christoforos Hadjichrysanthou
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, W2 1PG, UK
| | - Roy M Anderson
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, London, W2 1PG, UK
| | - John Collinge
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, W1W 7FF, UK
| |
Collapse
|
40
|
Jamerlan A, An SSA. The influence of Aβ-dependent and independent pathways on TDP-43 proteinopathy in Alzheimer's disease: a possible connection to LATE-NC. Neurobiol Aging 2020; 95:161-167. [PMID: 32814257 DOI: 10.1016/j.neurobiolaging.2020.06.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that results from the accumulation of plaques by cleaved Aβ42 peptides as well as neurofibrillary tangles of tau proteins. This accumulation triggers a complex cascade of cytotoxic, neuroinflammatory, and oxidative stresses that lead to neuronal death throughout the progression of the disease. Much of research in AD focused on the 2 pathologic proteins. Interestingly, another form of dementia with similar clinical manifestations of AD, but preferentially affected much older individuals, was termed as limbic-predominant age-related transactive response DNA-binding protein 43 (TDP-43) encephalopathy (LATE) and involved the cytotoxic intraneuronal deposition of phosphorylated TDP-43. TDP-43 proteinopathy was also found to be involved in AD pathology leading to the possibility that AD and LATE may share a common upstream etiology. This paper discusses the roles molecular pathways known in AD may have on influencing TDP-43 proteinopathy and the development of AD, LATE, or the 2 being comorbid with each other.
Collapse
Affiliation(s)
- Angelo Jamerlan
- Department of Bionano Technology, Gachon University, Seongnam-si, Republic of Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Seongnam-si, Republic of Korea.
| |
Collapse
|
41
|
Buciuc M, Botha H, Murray ME, Schwarz CG, Senjem ML, Jones DT, Knopman DS, Boeve BF, Petersen RC, Jack CR, Petrucelli L, Parisi JE, Dickson DW, Lowe V, Whitwell JL, Josephs KA. Utility of FDG-PET in diagnosis of Alzheimer-related TDP-43 proteinopathy. Neurology 2020; 95:e23-e34. [PMID: 32518145 DOI: 10.1212/wnl.0000000000009722] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 12/07/2019] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE To evaluate FDG-PET as an antemortem diagnostic tool for Alzheimer-related TAR DNA-binding protein of 43 kDa (TDP-43) proteinopathy. METHODS We conducted a cross-sectional neuroimaging-histologic analysis of patients with antemortem FDG-PET and postmortem brain tissue from the Mayo Clinic Alzheimer's Disease Research Center and Study of Aging with Alzheimer spectrum pathology. TDP-43-positive status was assigned when TDP-43-immunoreactive inclusions were identified in the amygdala. Statistical parametric mapping (SPM) analyses compared TDP-43-positive (TDP-43[+]) with TDP-43-negative cases (TDP-43[-]), correcting for field strength, sex, Braak neurofibrillary tangle, and neuritic plaque stages. Cross-validated logistic regression analyses were used to determine whether regional FDG-PET values predict TDP-43 status. We also assessed the ratio of inferior temporal to medial temporal (IMT) metabolism as this was proposed as a biomarker of hippocampal sclerosis. RESULTS Of 73 cases, 27 (37%) were TDP-43(+), of which 6 (8%) had hippocampal sclerosis. SPM analysis showed TDP-43(+) cases having greater hypometabolism of medial temporal, frontal superior medial, and frontal supraorbital (FSO) regions (p unc < 0.001). Logistic regression analysis showed only FSO and IMT to be associated with TDP-43(+) status, identifying up to 81% of TDP-43(+) cases (p < 0.001). An IMT/FSO ratio was superior to the IMT in discriminating TDP-43(+) cases: 78% vs 48%, respectively. CONCLUSIONS Alzheimer-related TDP-43 proteinopathy is associated with hypometabolism in the medial temporal and frontal regions. Combining FDG-PET measures from these regions may be useful for antemortem prediction of Alzheimer-related TDP-43 proteinopathy. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that hypometabolism in the medial temporal and frontal regions on FDG-PET is associated with Alzheimer-related TDP-43 proteinopathy.
Collapse
Affiliation(s)
- Marina Buciuc
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Hugo Botha
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Melissa E Murray
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Christopher G Schwarz
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Matthew L Senjem
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - David T Jones
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - David S Knopman
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Bradley F Boeve
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Ronald C Petersen
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Clifford R Jack
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Leonard Petrucelli
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Joseph E Parisi
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Dennis W Dickson
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Val Lowe
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Jennifer L Whitwell
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL
| | - Keith A Josephs
- From the Departments of Neurology (M.B., H.B., D.T.J., D.S.K., B.F.B., R.C.P., K.A.J.), Radiology (C.G.S., M.L.S., C.R.J., V.L., J.L.W.), and Laboratory Medicine and Pathology (J.E.P.), Mayo Clinic, Rochester, MN; and Department of Neuroscience (M.E.M., L.P., D.W.D.), Mayo Clinic, Jacksonville, FL.
| |
Collapse
|
42
|
de Flores R, Wisse LE, Das SR, Xie L, McMillan CT, Trojanowski JQ, Robinson JL, Grossman M, Lee E, Irwin DJ, Yushkevich PA, Wolk DA. Contribution of mixed pathology to medial temporal lobe atrophy in Alzheimer's disease. Alzheimers Dement 2020; 16:843-852. [PMID: 32323446 PMCID: PMC7715004 DOI: 10.1002/alz.12079] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/13/2020] [Accepted: 02/15/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION It is unclear how different proteinopathies (tau, transactive response DNA-binding protein 43 [TDP-43], amyloid β [Aβ], and α-synuclein) contribute to atrophy within medial temporal lobe (MTL) subregions in Alzheimer's disease (AD). METHODS We utilized antemortem structural magnetic resonance imaging (MRI) data to measure MTL substructures and examined the relative contribution of tau, TDP-43, Aβ, and α-synuclein measured in post-mortem tissue from 92 individuals with intermediate to high AD neuropathology. Receiver-operating characteristic (ROC) curves were analyzed for each subregion in order to discriminate TDP-43-negative and TDP-43-positive patients. RESULTS TDP-43 was strongly associated with anterior MTL regions, whereas tau was relatively more associated with the posterior hippocampus. Among the MTL regions, the anterior hippocampus showed the highest area under the ROC curve (AUC). DISCUSSION We found specific contributions of different pathologies on MTL substructure in this population with AD neuropathology. The anterior hippocampus may be a relevant region to detect concomitant TDP-43 pathology in the MTL of patients with AD.
Collapse
Affiliation(s)
- Robin de Flores
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Image Computing and Science Laboratory (PICSL), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laura E.M. Wisse
- Penn Image Computing and Science Laboratory (PICSL), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sandhitsu R. Das
- Penn Image Computing and Science Laboratory (PICSL), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Long Xie
- Penn Image Computing and Science Laboratory (PICSL), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Corey T. McMillan
- Penn FTD Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Q. Trojanowski
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John L. Robinson
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn FTD Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward Lee
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David J. Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn FTD Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul A. Yushkevich
- Penn Image Computing and Science Laboratory (PICSL), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David A. Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
43
|
Lloyd GM, Trejo-Lopez JA, Xia Y, McFarland KN, Lincoln SJ, Ertekin-Taner N, Giasson BI, Yachnis AT, Prokop S. Prominent amyloid plaque pathology and cerebral amyloid angiopathy in APP V717I (London) carrier - phenotypic variability in autosomal dominant Alzheimer's disease. Acta Neuropathol Commun 2020; 8:31. [PMID: 32164763 PMCID: PMC7068954 DOI: 10.1186/s40478-020-0891-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 01/30/2020] [Indexed: 12/14/2022] Open
Abstract
The discovery of mutations associated with familial forms of Alzheimer's disease (AD), has brought imperative insights into basic mechanisms of disease pathogenesis and progression and has allowed researchers to create animal models that assist in the elucidation of the molecular pathways and development of therapeutic interventions. Position 717 in the amyloid precursor protein (APP) is a hotspot for mutations associated with autosomal dominant AD (ADAD) and the valine to isoleucine amino acid substitution (V717I) at this position was among the first ADAD mutations identified, spearheading the formulation of the amyloid cascade hypothesis of AD pathogenesis. While this mutation is well described in multiple kindreds and has served as the basis for the generation of widely used animal models of disease, neuropathologic data on patients carrying this mutation are scarce. Here we present the detailed clinical and neuropathologic characterization of an APP V717I carrier, which reveals important novel insights into the phenotypic variability of ADAD cases. While age at onset, clinical presentation and widespread parenchymal beta-amyloid (Aβ) deposition are in line with previous reports, our case also shows widespread and severe cerebral amyloid angiopathy (CAA). This patient also presented with TDP-43 pathology in the hippocampus and amygdala, consistent with limbic predominant age-related TDP-43 proteinopathy (LATE). The APOE ε2/ε3 genotype may have been a major driver of the prominent vascular pathology seen in our case. These findings highlight the importance of neuropathologic examinations of genetically determined AD cases and demonstrate striking phenotypic variability in ADAD cases.
Collapse
Affiliation(s)
- 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
| | - Jorge A Trejo-Lopez
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Pathology, University of Florida, Gainesville, FL, 32610, USA
| | - Yuxing Xia
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Karen N McFarland
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neurology, 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
| | - Sarah J Lincoln
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, 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
| | - Anthony T Yachnis
- Department of Pathology, University of Florida, Gainesville, FL, 32610, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.
- Department of Pathology, 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
|
44
|
Are comorbidities compatible with a molecular pathological classification of neurodegenerative diseases? Curr Opin Neurol 2020; 32:279-291. [PMID: 30672825 DOI: 10.1097/wco.0000000000000664] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an update on comorbidities in neurodegenerative conditions. The term comorbidity is used here to distinguish cases with overlapping pathogenic mechanisms, which includes combinations of neurodegenerative proteinopathies from cases with multimorbidity, which is defined as concomitant brain and systemic disorders with different pathogenic mechanisms. RECENT FINDINGS Comorbid proteinopathies are more frequent in both sporadic and hereditary neurodegenerative diseases than previously assumed. The most frequent additional proteinopathies are related to Alzheimer's disease, Lewy body disorder, and limbic predominant transactive response DNA-binding protein 43 proteinopathy, however, different forms of tau pathologies are also increasingly recognized. In addition to ageing, synergistic interaction of proteins, common disease pathways, and the influence of genetic variations are discussed as possible pathogenic players. SUMMARY Comorbid proteinopathies might influence the clinical course and have implications for biomarker and therapeutic development. As pure forms of proteinopathies are still observed, the notion of current molecular classification is justified. This corroborates elucidation of various pathogenic pathways leading to neurodegeneration. Assuming that single proteins and associated pathways are targeted in therapy trials, efforts are needed to better stratify patients and to select pure proteinopathy forms lacking unfavorable genetic constellations. Otherwise combined therapeutic strategies might be necessary for comorbid proteinopathies.
Collapse
|
45
|
Flanagan ME, Cholerton B, Latimer CS, Hemmy LS, Edland SD, Montine KS, White LR, Montine TJ. TDP-43 Neuropathologic Associations in the Nun Study and the Honolulu-Asia Aging Study. J Alzheimers Dis 2019; 66:1549-1558. [PMID: 30452409 DOI: 10.3233/jad-180162] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Transactive response binding protein-43 (TDP-43) cytoplasmic neuronal and glial aggregates (pathologic TDP-43) have been described in multiple brain diseases. We describe the associations between neuropathologically confirmed TDP-43 and cognition in two population-based cohorts: the Nun Study (NS) and the Honolulu-Asia Aging Study (HAAS). In the HAAS, there was a significant association between hippocampal sclerosis (HS) and TDP-43 (OR = 11.04, p < 0.0001, 95% CI 3.57-34.13). In the NS, there were significant associations between TDP-43 and HS (OR = 16.44, p > 0.001 95%, CI 7.10-38.00) and Alzheimer's disease (AD) severity (OR = 1.74, p = 0.009, 95% CI 1.15-2.64). When cognitive scores were added to the model, HS remained significant but the other variables were not. When HS was removed from the model, the overall model remained significant and the associations between cognitive performance and TDP-43 (OR = 2.11, p = 0.022, 95% CI 1.11-4.02) were significant. In the NS, there was a significant association between cognitive performance and TDP-43 (OR 1.94 p = 0.005, 95% CI 1.22-3.09) (HS remained significant, but AD did not). When HS was removed from the model, only CERAD was significant (OR = 2.43 p < 0.001, 95% CI 1.58-3.74). These results support a consistent association between pathologic TDP-43, HS, and the development of cognitive impairment in two large studies of brain aging, while the relationship between AD pathology and TDP-43 may vary according to cohort-specific features.
Collapse
Affiliation(s)
- Margaret E Flanagan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Laura S Hemmy
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA.,Geriatric Research, Education, and Clinical Center, VA Health Care System, Minneapolis, MN, USA
| | - Steven D Edland
- Department of Family Medicine and Public Health, University of California, San Diego, CA, USA
| | | | - Lon R White
- Pacific Health Research and Education Institute (PHREI), Honolulu, HI, USA.,Department of Geriatric Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA
| | | |
Collapse
|
46
|
Kero M, Raunio A, Polvikoski T, Tienari PJ, Paetau A, Myllykangas L. Hippocampal Sclerosis in the Oldest Old: A Finnish Population-Based Study. J Alzheimers Dis 2019; 63:263-272. [PMID: 29614661 PMCID: PMC5900558 DOI: 10.3233/jad-171068] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background: There are only few population-based studies that have systemically investigated the prevalence of hippocampal sclerosis (HS) in the very old. The frequency of unilateral versus bilateral HS has been rarely studied. Objective: We investigated the prevalence and laterality of HS and its association with other neurodegenerative and vascular pathologies in a population-based sample of very elderly. Furthermore, the concomitant presence of immunoreactivity for TDP-43, p62, and HPtau was studied. Methods: The population-based Vantaa 85+ study includes all inhabitants of the city of Vantaa, who were >85 years in 1991 (n = 601). Neuropathological assessment was possible in 302 subjects. Severity of neuronal loss of CA sectors and subiculum was determined bilaterally by HE- staining. Immunohistochemistry performed using antibodies for TDP-43, p62, and HPtau. Results: Neuronal loss and pathological changes in the hippocampus sector CA1 and subiculum were observed in 47 of the 302 individuals (16%), and 51% of these changes were bilateral. HS without comorbid neurodegenerative pathology was found in 1/47 subjects with HS (2%). Dementia (p < 0.001) and TDP-43 immunopositivity of the granular cell layer of the dentate fascia (p < 0.001) were strongly associated with HS. The CERAD score, immunopositivity for HPtau and p62 in the granular cell layer of the fascia dentate were also associated. Conclusion: HS is prevalent (16%) in the oldest old population, but HS without any comorbid neurodegenerative pathology is rare. The high frequency of unilateral HS (49%) implied that bilateral sampling of hippocampi should be routine practice in neuropathological examination.
Collapse
Affiliation(s)
- Mia Kero
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Anna Raunio
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Tuomo Polvikoski
- Institute of Neuroscience, University of Newcastle, Newcastle upon Tyne, United Kingdom
| | - Pentti J Tienari
- Department of Neurology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anders Paetau
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Liisa Myllykangas
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
47
|
Verwey NA, Teunissen CE, Hoozemans JJM, Rozemuller AJM, Scheltens P, Pijnenburg YAL. Cerebrospinal Fluid Amyloid-β Subtypes in Confirmed Frontotemporal Lobar Degeneration Cases: A Pilot Study. J Alzheimers Dis 2019; 71:15-20. [PMID: 31356209 DOI: 10.3233/jad-190344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
To investigate amyloid-β (Aβ) in frontotemporal dementia (FTD), cerebrospinal fluid (CSF) Aβ38, Aβ40, and Aβ42 in frontotemporal lobar degeneration (FTLD; N = 18 genetically and/or pathologically confirmed and N = 8 FTD with concomitant amyotrophic lateral sclerosis) were compared with Alzheimer's disease (AD; pathological or Pittsburgh-compound-B Positron-emission-tomography (PIB-PET) positive; N = 25) and controls (N = 24). For all the Aβ subtypes, group difference was seen and post-hoc analysis revealed lower levels in FTLD compared to controls (p≤0.05). Aβ42/40 ratio showed no difference between FTLD and controls; however, a difference was seen between AD versus FTLD (p < 0.01). This is an intriguing finding, suggesting a possible role of Aβ in FTLD pathogenesis.
Collapse
Affiliation(s)
- Nicolaas A Verwey
- Department of Neurology, Medisch Centrum Leeuwarden, The Netherlands
| | - Charlotte E Teunissen
- Departments of Clinical Chemistry, Amsterdam University Medical Center location VUmc, Alzheimer Center, Amsterdam, The Netherlands
| | - Jeroen J M Hoozemans
- Departments of Pathology, Amsterdam University Medical Center location VUmc, Alzheimer Center, Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Departments of Pathology, Amsterdam University Medical Center location VUmc, Alzheimer Center, Amsterdam, The Netherlands
| | - Philip Scheltens
- Departments of Neurology, Amsterdam University Medical Center location VUmc, Alzheimer Center, Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Departments of Neurology, Amsterdam University Medical Center location VUmc, Alzheimer Center, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Tau and TDP-43 proteinopathies: kindred pathologic cascades and genetic pleiotropy. J Transl Med 2019; 99:993-1007. [PMID: 30742063 PMCID: PMC6609463 DOI: 10.1038/s41374-019-0196-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 12/11/2022] Open
Abstract
We review the literature on Tau and TDP-43 proteinopathies in aged human brains and the relevant underlying pathogenetic cascades. Complex interacting pathways are implicated in Alzheimer's disease and related dementias (ADRD), wherein multiple proteins tend to misfold in a manner that is "reactive," but, subsequently, each proteinopathy may contribute strongly to the clinical symptoms. Tau proteinopathy exists in brains of individuals across a broad spectrum of primary underlying conditions-e.g., developmental, traumatic, and inflammatory/infectious diseases. TDP-43 proteinopathy is also expressed in a wide range of clinical disorders. Although TDP-43 proteinopathy was first described in the central nervous system of patients with amyotrophic lateral sclerosis (ALS) and in subtypes of frontotemporal dementia (FTD/FTLD), TDP-43 proteinopathy is also present in chronic traumatic encephalopathy, cognitively impaired persons in advanced age with hippocampal sclerosis, Huntington's disease, and other diseases. We list known Tau and TDP-43 proteinopathies. There is also evidence of cellular co-localization between Tau and TDP-43 misfolded proteins, suggesting common pathways or protein interactions facilitating misfolding in one protein by the other. Multiple pleiotropic gene variants can alter risk for Tau or TDP-43 pathologies, and certain gene variants (e.g., APOE ε4, Huntingtin triplet repeats) are associated with increases of both Tau and TDP-43 proteinopathies. Studies of genetic risk factors have provided insights into multiple nodes of the pathologic cascades involved in Tau and TDP-43 proteinopathies. Variants from a specific gene can be either a low-penetrant risk factor for a group of diseases, or alternatively, a different variant of the same gene may be a disease-driving allele that is associated with a relatively aggressive and early-onset version of a clinically and pathologically specific disease type. Overall, a complex but enlightening paradigm has emerged, wherein both Tau and TDP-43 proteinopathies are linked to numerous overlapping upstream influences, and both are associated with multiple downstream pathologically- and clinically-defined deleterious effects.
Collapse
|
49
|
Abstract
This chapter describes the main neuropathological features of the most common age associated neurodegenerative diseases including Alzheimer's disease, Lewy body diseases, vascular dementia and the various types of frontotemporal lobar degeneration. In addition, the more recent concepts of primary age-related tauopathy and ageing-related tau astrogliopathy as well as chronic traumatic encephalopathy are briefly described. One section is dedicated to cerebral multi-morbidity as it is becoming increasingly clear that the old brain is characterised by the presence of multiple pathologies (to varying extent) rather than by one single, disease specific pathology alone. The main aim of this chapter is to inform the reader about the neuropathological basics of age associated neurodegenerative diseases as we feel this is crucial to meaningfully interpret the vast literature that is published in the broad field of dementia research.
Collapse
Affiliation(s)
- Lauren Walker
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Kirsty E McAleese
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Daniel Erskine
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Johannes Attems
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
50
|
Nelson PT, Dickson DW, Trojanowski JQ, Jack CR, Boyle PA, Arfanakis K, Rademakers R, Alafuzoff I, Attems J, Brayne C, Coyle-Gilchrist ITS, Chui HC, Fardo DW, Flanagan ME, Halliday G, Hokkanen SRK, Hunter S, Jicha GA, Katsumata Y, Kawas CH, Keene CD, Kovacs GG, Kukull WA, Levey AI, Makkinejad N, Montine TJ, Murayama S, Murray ME, Nag S, Rissman RA, Seeley WW, Sperling RA, White III CL, Yu L, Schneider JA. Limbic-predominant age-related TDP-43 encephalopathy (LATE): consensus working group report. Brain 2019; 142:1503-1527. [PMID: 31039256 PMCID: PMC6536849 DOI: 10.1093/brain/awz099] [Citation(s) in RCA: 851] [Impact Index Per Article: 170.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/10/2019] [Accepted: 02/25/2019] [Indexed: 12/18/2022] Open
Abstract
We describe a recently recognized disease entity, limbic-predominant age-related TDP-43 encephalopathy (LATE). LATE neuropathological change (LATE-NC) is defined by a stereotypical TDP-43 proteinopathy in older adults, with or without coexisting hippocampal sclerosis pathology. LATE-NC is a common TDP-43 proteinopathy, associated with an amnestic dementia syndrome that mimicked Alzheimer's-type dementia in retrospective autopsy studies. LATE is distinguished from frontotemporal lobar degeneration with TDP-43 pathology based on its epidemiology (LATE generally affects older subjects), and relatively restricted neuroanatomical distribution of TDP-43 proteinopathy. In community-based autopsy cohorts, ∼25% of brains had sufficient burden of LATE-NC to be associated with discernible cognitive impairment. Many subjects with LATE-NC have comorbid brain pathologies, often including amyloid-β plaques and tauopathy. Given that the 'oldest-old' are at greatest risk for LATE-NC, and subjects of advanced age constitute a rapidly growing demographic group in many countries, LATE has an expanding but under-recognized impact on public health. For these reasons, a working group was convened to develop diagnostic criteria for LATE, aiming both to stimulate research and to promote awareness of this pathway to dementia. We report consensus-based recommendations including guidelines for diagnosis and staging of LATE-NC. For routine autopsy workup of LATE-NC, an anatomically-based preliminary staging scheme is proposed with TDP-43 immunohistochemistry on tissue from three brain areas, reflecting a hierarchical pattern of brain involvement: amygdala, hippocampus, and middle frontal gyrus. LATE-NC appears to affect the medial temporal lobe structures preferentially, but other areas also are impacted. Neuroimaging studies demonstrated that subjects with LATE-NC also had atrophy in the medial temporal lobes, frontal cortex, and other brain regions. Genetic studies have thus far indicated five genes with risk alleles for LATE-NC: GRN, TMEM106B, ABCC9, KCNMB2, and APOE. The discovery of these genetic risk variants indicate that LATE shares pathogenetic mechanisms with both frontotemporal lobar degeneration and Alzheimer's disease, but also suggests disease-specific underlying mechanisms. Large gaps remain in our understanding of LATE. For advances in prevention, diagnosis, and treatment, there is an urgent need for research focused on LATE, including in vitro and animal models. An obstacle to clinical progress is lack of diagnostic tools, such as biofluid or neuroimaging biomarkers, for ante-mortem detection of LATE. Development of a disease biomarker would augment observational studies seeking to further define the risk factors, natural history, and clinical features of LATE, as well as eventual subject recruitment for targeted therapies in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Konstantinos Arfanakis
- Rush University Medical Center, Chicago, IL, USA
- Illinois Institute of Technology, Chicago, IL, USA
| | | | | | | | | | | | - Helena C Chui
- University of Southern California, Los Angeles, CA, USA
| | | | | | - Glenda Halliday
- The University of Sydney Brain and Mind Centre and Central Clinical School Faculty of Medicine and Health, Sydney, Australia
| | | | | | | | | | | | | | - Gabor G Kovacs
- Institute of Neurology Medical University of Vienna, Vienna, Austria
| | | | | | | | | | - Shigeo Murayama
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | | | - Sukriti Nag
- Rush University Medical Center, Chicago, IL, USA
| | | | | | | | | | - Lei Yu
- Rush University Medical Center, Chicago, IL, USA
| | | |
Collapse
|