1
|
Lavrova A, Pham NTT, Reid RI, Boeve BF, Knopman DS, Petersen RC, Nguyen AT, Ross Reichard R, Dickson DW, Jack CR, Whitwell JL, Josephs KA. Relation of Alzheimer's disease-related TDP-43 proteinopathy to metrics from diffusion tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI). Neurobiol Aging 2025; 150:97-108. [PMID: 40088623 DOI: 10.1016/j.neurobiolaging.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025]
Abstract
Transactive response DNA-binding protein 43 kDa (TDP-43) deposition is linked to regional brain atrophy in Alzheimer's disease (AD), but diffusion changes associated with AD-related TDP-43 proteinopathy remain underexplored. This study evaluates the potential of diffusion tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI) as in vivo markers for detecting TDP-43 proteinopathy in AD. We analyzed DTI and NODDI metrics in 49 cases with AD neuropathologic changes, categorized by postmortem TDP-43 status. Diffusion metrics from the temporal lobe gray and white matter regions and key white matter tracts were compared between TDP-43-positive and negative cases. Group differences were significant in the left hippocampus, amygdala, and uncinate fasciculus after adjusting for age, Braak neurofibrillary tangle (NFT) stage and APOE ε4 status. TDP-43-positive cases showed increased mean diffusivity (MD) and altered neurite density index (NDI) and orientation dispersion index (ODI). Area under the receiver operating characteristic curve (AUROC) analysis revealed high predictive accuracy for amygdala ODI (AUC = 0.809, sensitivity = 0.81, specificity = 0.76), hippocampal MD (AUC = 0.763, sensitivity = 0.81, specificity = 0.67), and uncinate fasciculus MD (AUC = 0.782, sensitivity = 0.88, specificity = 0.61). Combined, DTI/NODDI predictors demonstrated stronger discriminative ability (AUC = 0.856, sensitivity = 0.88, specificity = 0.76). These findings suggest that AD-related TDP-43 proteinopathy is associated with specific diffusion changes in the left temporal lobe. DTI and NODDI metrics, particularly MD, NDI, and ODI, may improve the antemortem detection of TDP-43 pathology in AD.
Collapse
Affiliation(s)
- Anna Lavrova
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Robert I Reid
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | - R Ross Reichard
- Department of Laboratory Medicine and Pathology, Rochester, MN, USA
| | | | | | | | | |
Collapse
|
2
|
Pottier C, Küçükali F, Baker M, Batzler A, Jenkins GD, van Blitterswijk M, Vicente CT, De Coster W, Wynants S, Van de Walle P, Ross OA, Murray ME, Faura J, Haggarty SJ, van Rooij JG, Mol MO, Hsiung GYR, Graff C, Öijerstedt L, Neumann M, Asmann Y, McDonnell SK, Baheti S, Josephs KA, Whitwell JL, Bieniek KF, Forsberg L, Heuer H, Lago AL, Geier EG, Yokoyama JS, Oddi AP, Flanagan M, Mao Q, Hodges JR, Kwok JB, Domoto-Reilly K, Synofzik M, Wilke C, Onyike C, Dickerson BC, Evers BM, Dugger BN, Munoz DG, Keith J, Zinman L, Rogaeva E, Suh E, Gefen T, Geula C, Weintraub S, Diehl-Schmid J, Farlow MR, Edbauer D, Woodruff BK, Caselli RJ, Donker Kaat LL, Huey ED, Reiman EM, Mead S, King A, Roeber S, Nana AL, Ertekin-Taner N, Knopman DS, Petersen RC, Petrucelli L, Uitti RJ, Wszolek ZK, Ramos EM, Grinberg LT, Tempini MLG, Rosen HJ, Spina S, Piguet O, Grossman M, Trojanowski JQ, Keene CD, Jin LW, Prudlo J, Geschwind DH, Rissman RA, Cruchaga C, Ghetti B, Halliday GM, Beach TG, Serrano GE, Arzberger T, Herms J, Boxer AL, Honig LS, Vonsattel JP, Lopez OL, Kofler J, White CL, Gearing M, Glass J, Rohrer JD, Irwin DJ, Lee EB, et alPottier C, Küçükali F, Baker M, Batzler A, Jenkins GD, van Blitterswijk M, Vicente CT, De Coster W, Wynants S, Van de Walle P, Ross OA, Murray ME, Faura J, Haggarty SJ, van Rooij JG, Mol MO, Hsiung GYR, Graff C, Öijerstedt L, Neumann M, Asmann Y, McDonnell SK, Baheti S, Josephs KA, Whitwell JL, Bieniek KF, Forsberg L, Heuer H, Lago AL, Geier EG, Yokoyama JS, Oddi AP, Flanagan M, Mao Q, Hodges JR, Kwok JB, Domoto-Reilly K, Synofzik M, Wilke C, Onyike C, Dickerson BC, Evers BM, Dugger BN, Munoz DG, Keith J, Zinman L, Rogaeva E, Suh E, Gefen T, Geula C, Weintraub S, Diehl-Schmid J, Farlow MR, Edbauer D, Woodruff BK, Caselli RJ, Donker Kaat LL, Huey ED, Reiman EM, Mead S, King A, Roeber S, Nana AL, Ertekin-Taner N, Knopman DS, Petersen RC, Petrucelli L, Uitti RJ, Wszolek ZK, Ramos EM, Grinberg LT, Tempini MLG, Rosen HJ, Spina S, Piguet O, Grossman M, Trojanowski JQ, Keene CD, Jin LW, Prudlo J, Geschwind DH, Rissman RA, Cruchaga C, Ghetti B, Halliday GM, Beach TG, Serrano GE, Arzberger T, Herms J, Boxer AL, Honig LS, Vonsattel JP, Lopez OL, Kofler J, White CL, Gearing M, Glass J, Rohrer JD, Irwin DJ, Lee EB, Van Deerlin V, Castellani R, Mesulam MM, Tartaglia MC, Finger EC, Troakes C, Al-Sarraj S, Dalgard CL, Miller BL, Seelaar H, Graff-Radford NR, Boeve BF, Mackenzie IR, van Swieten JC, Seeley WW, Sleegers K, Dickson DW, Biernacka JM, Rademakers R. Deciphering distinct genetic risk factors for FTLD-TDP pathological subtypes via whole-genome sequencing. Nat Commun 2025; 16:3914. [PMID: 40280976 PMCID: PMC12032271 DOI: 10.1038/s41467-025-59216-0] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Frontotemporal lobar degeneration with neuronal inclusions of the TAR DNA-binding protein 43 (FTLD-TDP) is a fatal neurodegenerative disorder with only a limited number of risk loci identified. We report our comprehensive genome-wide association study as part of the International FTLD-TDP Whole-Genome Sequencing Consortium, including 985 patients and 3,153 controls compiled from 26 institutions/brain banks in North America, Europe and Australia, and meta-analysis with the Dementia-seq cohort. We confirm UNC13A as the strongest overall FTLD-TDP risk factor and identify TNIP1 as a novel FTLD-TDP risk factor. In subgroup analyzes, we further identify genome-wide significant loci specific to each of the three main FTLD-TDP pathological subtypes (A, B and C), as well as enrichment of risk loci in distinct tissues, brain regions, and neuronal subtypes, suggesting distinct disease aetiologies in each of the subtypes. Rare variant analysis confirmed TBK1 and identified C3AR1, SMG8, VIPR1, RBPJL, L3MBTL1 and ANO9, as novel subtype-specific FTLD-TDP risk genes, further highlighting the role of innate and adaptive immunity and notch signaling pathway in FTLD-TDP, with potential diagnostic and novel therapeutic implications.
Collapse
Affiliation(s)
- Cyril Pottier
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA.
| | - Fahri Küçükali
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Matt Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Anthony Batzler
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Gregory D Jenkins
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Cristina T Vicente
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Wouter De Coster
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Sarah Wynants
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Pieter Van de Walle
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Júlia Faura
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Stephen J Haggarty
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Merel O Mol
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ging-Yuek R Hsiung
- Department of Medicine, Division of Neurology, University of British Columbia, Vancouver, BC, Canada
| | - Caroline Graff
- Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Karolinska University Hospital, Solna, Sweden
| | - Linn Öijerstedt
- Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Karolinska University Hospital, Solna, Sweden
| | - Manuela Neumann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neuropathology, University of Tübingen, Tübingen, Germany
| | - Yan Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | | - Saurabh Baheti
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Kevin F Bieniek
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Leah Forsberg
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Hilary Heuer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Argentina Lario Lago
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Ethan G Geier
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Jennifer S Yokoyama
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Alexis P Oddi
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Margaret Flanagan
- University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Qinwen Mao
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - John R Hodges
- Central Clinical School and Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - John B Kwok
- University of Sydney, Sydney, NSW, Australia
- NeuRA, University of New South Wales, Randwick, NSW, Australia
| | | | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Carlo Wilke
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Chiadi Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | | | - Bret M Evers
- Division of Neuropathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, University of California, Davis Medical Center, Sacramento, CA, USA
| | - David G Munoz
- St. Michael's Hospital, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Julia Keith
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Lorne Zinman
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Ekaterina Rogaeva
- Krembil Discovery Tower, Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada
| | - EunRan Suh
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Technical University of Munich, Munich, Germany
- kbo-Inn-Salzach-Klinikum, Clinical Center for Psychiatry, Psychotherapy, Psychosomatic Medicine, Geriatrics and Neurology, Wasserburg/Inn, Germany
| | - Martin R Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | | | | - Laura L Donker Kaat
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Edward D Huey
- Department of Psychiatry and Human Behavior, Brown Alpert Medical School, Brown University, Providence, RI, USA
| | | | - Simon Mead
- MRC Prion Unit at University College London, Institute of Prion Diseases, London, UK
| | - Andrew King
- Department of Basic and Clinical Neuroscience, London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, London, UK
| | - Sigrun Roeber
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Alissa L Nana
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Nilufer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | - Ryan J Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Eliana Marisa Ramos
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Lea T Grinberg
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Maria Luisa Gorno Tempini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Howard J Rosen
- Department of Pathology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Salvatore Spina
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Olivier Piguet
- School of Psychology and Brain and Mind Centre, University of Sydney, Sydney, SWA, Australia
| | - Murray Grossman
- Department of Neurology, Penn Frontotemporal Degeneration Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - C Dirk Keene
- University of Washington BioRepository and Integrated Neuropathology (BRaIN) lab, Harborview Medical Center, Seattle, WA, USA
| | - Lee-Way Jin
- M.I.N.D. Institute Laboratory, University of California, Davis Medical Center, Sacramento, CA, USA
| | - Johannes Prudlo
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Daniel H Geschwind
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Robert A Rissman
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Thomas Arzberger
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Centre for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Adam L Boxer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Lawrence S Honig
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Jean P Vonsattel
- Department of Pathology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles L White
- Division of Neuropathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marla Gearing
- Department of Pathology and Laboratory Medicine and Department of Neurology, Emory University, Atlanta, GA, USA
| | - Jonathan Glass
- Department of Pathology and Laboratory Medicine and Department of Neurology, Emory University, Atlanta, GA, USA
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, University College London, Queen Square Institute of Neurology, London, UK
| | - David J Irwin
- Department of Neurology, Penn Frontotemporal Degeneration Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Vivianna Van Deerlin
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Rudolph Castellani
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marsel M Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Maria C Tartaglia
- Krembil Discovery Tower, Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada
| | - Elizabeth C Finger
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Claire Troakes
- Department of Basic and Clinical Neuroscience, London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Safa Al-Sarraj
- Department of Basic and Clinical Neuroscience, London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Bruce L Miller
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Ian Ra Mackenzie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - William W Seeley
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Kristel Sleegers
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | | | - Joanna M Biernacka
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
| |
Collapse
|
3
|
Hiya S, Maldonado-Díaz C, Rohde SK, Gonzales MM, Canbeldek L, Kulumani Mahadevan LS, Yokoda RT, Sullivan AC, Parker AS, White CL, Daoud EV, Flores-Almazan V, Crary JF, Farrell K, Walker JM, Richardson TE. Unraveling the clinical-pathological correlations of subjects with isolated and mixed neurodegenerative processes in the National Alzheimer's Coordinating Center dataset. J Neuropathol Exp Neurol 2025; 84:177-194. [PMID: 39728026 PMCID: PMC11842910 DOI: 10.1093/jnen/nlae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024] Open
Abstract
Although Alzheimer disease neuropathologic change (ADNC) is the most common pathology underlying clinical dementia, the presence of multiple comorbid neuropathologies is increasingly being recognized as a major contributor to the worldwide dementia burden. We analyzed 1051 subjects with specific combinations of isolated and mixed pathologies and conducted multivariate logistic regression analysis on a cohort of 4624 cases with mixed pathologies to systematically explore the independent cognitive contributions of each pathology. Alzheimer disease neuropathologic change and limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) were both associated with a primary clinical diagnosis of Alzheimer disease (AD) and were characterized by an amnestic dementia phenotype, while only ADNC associated with logopenic variant primary progressive aphasia (PPA). In subjects with ADNC and comorbid LATE-NC, Lewy body disease, and/or cerebrovascular disease, the clinical phenotype was usually diagnosed during life as "Probable AD." Conversely, the combination of ADNC with frontotemporal lobar degeneration with TDP-43, progressive supranuclear palsy (PSP), or corticobasal degeneration (CBD) resulted in a mixed clinical picture, with variable features of amnestic dementia, PPA subtypes, behavioral variant FTD, PSP syndrome, and CBD syndrome. These findings elucidate the cumulative effects of mixed pathologies and provide insights into interactions between neurodegenerative pathologies contributing to a variety of clinical dementia presentations.
Collapse
Affiliation(s)
- Satomi Hiya
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Carolina Maldonado-Díaz
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Susan K Rohde
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Human Genetics, Genomics of Neurodegenerative Diseases and Aging, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Neurology, Alzheimer Center Amsterdam, Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mitzi M Gonzales
- Department of Neurology, Cedars Sinai Medical Center, Los Angeles, CA, United States
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Leyla Canbeldek
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lakshmi S Kulumani Mahadevan
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Raquel T Yokoda
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - A Campbell Sullivan
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alicia S Parker
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Charles L White
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Elena V Daoud
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Victoria Flores-Almazan
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - John F Crary
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kurt Farrell
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jamie M Walker
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Neuropathology Brain Bank & Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Timothy E Richardson
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
4
|
Youssef H, Gatto RG, Pham NTT, Jones D, Petersen RC, Machulda MM, Whitwell JL, Josephs KA. Multiple Neuropathologies Underly Hippocampal Subfield Atrophy in a Case With a Slowly Progressive Amnestic Syndrome: Challenging the Notion of Pure LATE-NC. Neuropathology 2025. [PMID: 39973236 DOI: 10.1111/neup.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/21/2025]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly, marked by abnormal protein buildup (beta-amyloid and tau) resulting in neuronal loss, especially in the medial temporal lobe and other limbic regions. The presence of transactive response DNA binding protein 43 (TDP-43) immunoreactive inclusions in medial temporal lobe regions has also been associated with neuroimaging changes in limbic regions. It has been proposed that hypometabolism in limbic regions on [18F] fluorodeoxyglucose positron emission tomography (FDG-PET) in a patient with a slowly evolving amnestic syndrome may be a signature of the presence of TDP-43. In this context, we observed an 86-year-old Caucasian female with dementia characterized by a slowly evolving amnestic syndrome, along with focal medial temporal atrophy evident on MRI and hypometabolism in limbic regions on FDG-PET. The patient subsequently died and underwent an autopsy. We performed detailed neuroimaging and digital neuropathological analyses of the hippocampal subfields to better understand the relationship between clinico-imaging findings and histopathology. In addition to TDP-43, we identified three other pathological processes in the medial temporal lobe: sequestosome-1/p62, argyrophilic grain disease (AGD), and primary age-related tauopathy (PART). Hippocampal subfield volumes and rates of atrophy were no different from those of matched healthy controls, except for the atrophy rate in cornu ammonis 1 (CA1). Digital histopathology revealed the relative highest burden of pathology for p62, followed by TDP-43, AGD, and PART in CA1. Multiple pathological processes appear to have contributed to the hippocampal atrophy and hypometabolism in our patient with a slowly progressive amnestic syndrome.
Collapse
Affiliation(s)
- Hossam Youssef
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rodolfo G Gatto
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - David Jones
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Mary M Machulda
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Woodworth DC, Nguyen KM, Sordo L, Scambray KA, Head E, Kawas CH, Corrada MM, Nelson PT, Sajjadi SA. Evaluating the updated LATE-NC staging criteria using data from NACC. Alzheimers Dement 2024; 20:8359-8373. [PMID: 39352226 PMCID: PMC11667507 DOI: 10.1002/alz.14262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Limbic-predominant age-related TAR DNA-binding protein of 43 kDa encephalopathy neuropathologic change (LATE-NC) staging criteria were updated in 2023. We evaluated this updated staging using National Alzheimer's Coordinating Center data. METHODS We examined associations of LATE-NC stages with cognition and other neuropathologic changes (NCs), and with cognition while accounting for other NCs, using multilevel regression models. RESULTS Of 1352 participants, 502 (37%) had LATE-NC (23% stage 1a, 6% stage 1b, 58% stage 2, 13% stage 3). LATE-NC stages were associated with cognition, hippocampal sclerosis of aging (HS-A), Alzheimer's disease NC (ADNC), Lewy bodies (LBs), and hippocampal atrophy. While stage 1b was associated with cognition and HS-A consistent with other stages, it was not associated with ADNC or LBs. All LATE-NC stages remained significantly associated with worse cognition when accounting for other NCs. DISCUSSION The updated LATE-NC staging criteria capture variations in early TDP-43 pathology spread which are consequential for cognition and associations with other NCs. HIGHLIGHTS We applied the updated limbic-predominant age-related TAR DNA-binding protein of 43 kDa encephalopathy neuropathologic change (LATE-NC) staging criteria to data from the National Alzheimer's Coordinating Center. LATE-NC stage 1b was identified in 22% of participants with stage 1. In contrast to other LATE-NC stages, stage 1b was not associated with Alzheimer's disease neuropathologic change (ADNC) or Lewy bodies. Stages 1a and 1b were significantly associated with dementia and memory impairment. Stages 1b+ were more strongly tied to dementia than all other neuropathologic changes except high likelihood ADNC.
Collapse
Affiliation(s)
- Davis C. Woodworth
- Department of NeurologyUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Katelynn M. Nguyen
- Department of NeurologyUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Lorena Sordo
- Department of NeurologyUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Kiana A. Scambray
- Department of NeurologyUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Elizabeth Head
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| | - Claudia H. Kawas
- Department of NeurologyUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Neurobiology and BehaviorUniversity of CaliforniaIrvineCaliforniaUSA
| | - María M. Corrada
- Department of NeurologyUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Epidemiology and BiostatisticsUniversity of CaliforniaIrvineCaliforniaUSA
| | - Peter T. Nelson
- Department of Pathology and Laboratory MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - S. Ahmad Sajjadi
- Department of NeurologyUniversity of CaliforniaIrvineCaliforniaUSA
- Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
- Department of Pathology and Laboratory MedicineUniversity of CaliforniaIrvineCaliforniaUSA
| |
Collapse
|
6
|
Denning AE, Ittyerah R, Levorse LM, Sadeghpour N, Athalye C, Chung E, Ravikumar S, Dong M, Duong MT, Li Y, Ilesanmi A, Sreepada LP, Sabatini P, Lowe M, Bahena A, Zablah J, Spencer BE, Watanabe R, Kim B, Sørensen MH, Khandelwal P, Brown C, Hrybouski S, Xie SX, de Flores R, Robinson JL, Schuck T, Ohm DT, Arezoumandan S, Porta S, Detre JA, Insausti R, Wisse LEM, Das SR, Irwin DJ, Lee EB, Wolk DA, Yushkevich PA. Association of quantitative histopathology measurements with antemortem medial temporal lobe cortical thickness in the Alzheimer's disease continuum. Acta Neuropathol 2024; 148:37. [PMID: 39227502 PMCID: PMC11371872 DOI: 10.1007/s00401-024-02789-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/07/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024]
Abstract
The medial temporal lobe (MTL) is a hotspot for neuropathology, and measurements of MTL atrophy are often used as a biomarker for cognitive decline associated with neurodegenerative disease. Due to the aggregation of multiple proteinopathies in this region, the specific relationship of MTL atrophy to distinct neuropathologies is not well understood. Here, we develop two quantitative algorithms using deep learning to measure phosphorylated tau (p-tau) and TDP-43 (pTDP-43) pathology, which are both known to accumulate in the MTL and are associated with MTL neurodegeneration. We focus on these pathologies in the context of Alzheimer's disease (AD) and limbic predominant age-related TDP-43 encephalopathy (LATE) and apply our deep learning algorithms to distinct histology sections, on which MTL subregions were digitally annotated. We demonstrate that both quantitative pathology measures show high agreement with expert visual ratings of pathology and discriminate well between pathology stages. In 140 cases with antemortem MR imaging, we compare the association of semi-quantitative and quantitative postmortem measures of these pathologies in the hippocampus with in vivo structural measures of the MTL and its subregions. We find widespread associations of p-tau pathology with MTL subregional structural measures, whereas pTDP-43 pathology had more limited associations with the hippocampus and entorhinal cortex. Quantitative measurements of p-tau pathology resulted in a significantly better model of antemortem structural measures than semi-quantitative ratings and showed strong associations with cortical thickness and volume. By providing a more granular measure of pathology, the quantitative p-tau measures also showed a significant negative association with structure in a severe AD subgroup where semi-quantitative ratings displayed a ceiling effect. Our findings demonstrate the advantages of using quantitative neuropathology to understand the relationship of pathology to structure, particularly for p-tau, and motivate the use of quantitative pathology measurements in future studies.
Collapse
Affiliation(s)
- Amanda E Denning
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA.
| | - Ranjit Ittyerah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lisa M Levorse
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Chinmayee Athalye
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Eunice Chung
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sadhana Ravikumar
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Mengjin Dong
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Tran Duong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ademola Ilesanmi
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lasya P Sreepada
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Philip Sabatini
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - MaKayla Lowe
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Alejandra Bahena
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jamila Zablah
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Barbara E Spencer
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryohei Watanabe
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Boram Kim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Maja Højvang Sørensen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Pulkit Khandelwal
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher Brown
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Sharon X Xie
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Robin de Flores
- UMR-S U1237, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, INSERM, Caen-Normandie University, GIP Cyceron, Caen, France
| | - John L Robinson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Theresa Schuck
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel T Ohm
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sanaz Arezoumandan
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sílvia Porta
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - John A Detre
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ricardo Insausti
- Human Neuroanatomy Lab, University of Castilla La Mancha, Albacete, Spain
| | - Laura E M Wisse
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sandhitsu R Das
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - David J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Neurodegenerative Disease Research, Institute On Aging, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A Yushkevich
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Lang R, Hodgson RE, Shelkovnikova TA. TDP-43 in nuclear condensates: where, how, and why. Biochem Soc Trans 2024; 52:1809-1825. [PMID: 38958608 PMCID: PMC11668305 DOI: 10.1042/bst20231447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
TDP-43 is an abundant and ubiquitously expressed nuclear protein that becomes dysfunctional in a spectrum of neurodegenerative diseases. TDP-43's ability to phase separate and form/enter biomolecular condensates of varying size and composition is critical for its functionality. Despite the high density of phase-separated assemblies in the nucleus and the nuclear abundance of TDP-43, our understanding of the condensate-TDP-43 relationship in this cellular compartment is only emerging. Recent studies have also suggested that misregulation of nuclear TDP-43 condensation is an early event in the neurodegenerative disease amyotrophic lateral sclerosis. This review aims to draw attention to the nuclear facet of functional and aberrant TDP-43 condensation. We will summarise the current knowledge on how TDP-43 containing nuclear condensates form and function and how their homeostasis is affected in disease.
Collapse
Affiliation(s)
- Ruaridh Lang
- Sheffield Institute for Translational Neuroscience (SITraN) and Neuroscience Institute, University of Sheffield, Sheffield, U.K
| | - Rachel E. Hodgson
- Sheffield Institute for Translational Neuroscience (SITraN) and Neuroscience Institute, University of Sheffield, Sheffield, U.K
| | - Tatyana A. Shelkovnikova
- Sheffield Institute for Translational Neuroscience (SITraN) and Neuroscience Institute, University of Sheffield, Sheffield, U.K
| |
Collapse
|
8
|
Zheng X, Wang M, He Q, Chen S, Simayi D, Ma X, Zhao J, Sun X, Yang P, Mao Q, Xia H. Production and characterization of novel monoclonal antibodies against pathological human TDP-43 proteins. J Neuropathol Exp Neurol 2024; 83:655-669. [PMID: 38728009 PMCID: PMC11258413 DOI: 10.1093/jnen/nlae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024] Open
Abstract
The RNA/DNA-binding protein TDP-43 plays a pivotal role in the ubiquitinated inclusions characteristic of TDP-43 proteinopathies, including most cases of frontotemporal lobar degeneration (FTLD-TDP) and Alzheimer disease (AD). To understand the mechanisms of pathological TDP-43 processing and identify potential biomarkers, we generated novel phosphorylation-independent monoclonal antibodies (MAbs) using bacteria-expressed human full-length recombinant TDP-43. Remarkably, we identified a distinctive MAb, No. 9, targeting an epitope in amino acid (aa) region 311-360 of the C-terminus. This antibody showed preferential reactivity for pathological TDP-43 inclusions, with only mild reactivity for normal nuclear TDP-43. MAb No. 9 revealed more pathology in FTLD-TDP type A and type B brains and in AD brains compared to the commercial p409/410 MAb. Using synthetic phosphorylated peptides, we also obtained MAbs targeting the p409/410 epitope. Interestingly, MAb No. 14 was found to reveal additional pathology in AD compared to the commercial p409/410 MAb, specifically, TDP-43-immunopositive deposits with amyloid plaques in AD brains. These unique immunopositivities observed with MAbs No. 9 and No. 14 are likely attributed to their conformation-dependent binding to TDP-43 inclusions. We expect that this novel set of MAbs will prove valuable as tools for future patient-oriented investigations into TDP-43 proteinopathies.
Collapse
Affiliation(s)
- Xiaojing Zheng
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Shaanxi, P.R. China
| | - Mengtian Wang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Shaanxi, P.R. China
| | - Qiongyan He
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Shaanxi, P.R. China
| | - Shuyu Chen
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Shaanxi, P.R. China
| | - Dilihumaer Simayi
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Shaanxi, P.R. China
| | - Xia Ma
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Shaanxi, P.R. China
| | - Junli Zhao
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Shaanxi, P.R. China
| | - Xiaohong Sun
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Shaanxi, P.R. China
| | - Peiyan Yang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Shaanxi, P.R. China
| | - Qinwen Mao
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Shaanxi, P.R. China
| |
Collapse
|
9
|
Tomé SO, Gawor K, Thal DR. LATE-NC in Alzheimer's disease: Molecular aspects and synergies. Brain Pathol 2024; 34:e13213. [PMID: 37793659 PMCID: PMC11189776 DOI: 10.1111/bpa.13213] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 10/06/2023] Open
Abstract
Alzheimer's disease (AD) is classically characterized by senile plaques and neurofibrillary tangles (NFTs). However, multiple copathologies can be observed in the AD brain and contribute to the development of cognitive decline. Limbic-predominant age-related TDP-43 encephalopathy neuropathological changes (LATE-NC) accumulates in the majority of AD cases and leads to more severe cognitive decline compared with AD pathology alone. In this review, we focus on the synergistic relationship between LATE-NC and tau in AD, highlighting the aggravating role of TDP-43 aggregates on tau pathogenesis and its impact on the clinical picture and therapeutic strategies. Additionally, we discuss to what extent the molecular patterns of LATE-NC in AD differ from frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP) neuropathological changes. Thus, we highlight the importance of tau and TDP-43 synergies for subtyping AD patients, which may respond differently to therapeutic interventions depending on the presence of comorbid LATE-NC.
Collapse
Grants
- 10810 Alzheimer Forschung Initiative (Germany)
- 13803 Alzheimer Forschung Initiative (Germany)
- 22-AAIIA-963171 Alzheimer's Association (USA)
- A2022019F BrightFocus Foundation (USA)
- TH-624-4-1 Deutsche Forschungsgemeinschaft (DFG, Germany)
- 4-2 Deutsche Forschungsgemeinschaft (DFG, Germany)
- 6-1 Deutsche Forschungsgemeinschaft (DFG, Germany)
- G065721N Fonds Wetenschappelijk Onderzoek (FWO, Belgium)
- G0F8516N Fonds Wetenschappelijk Onderzoek (FWO, Belgium)
- 2020/017 Stichting Alzheimer Onderzoek (SAO/FRA, Belgium)
- C3/20/057 Onderzoeksraad, KU Leuven (Belgium)
- PDMT2/21/069 Onderzoeksraad, KU Leuven (Belgium)
- IWT 135043 Vlaamse Impulsfinanciering voor Netwerken voor Dementie-onderzoek (Belgium)
- Alzheimer Forschung Initiative (Germany)
- Alzheimer's Association (USA)
- BrightFocus Foundation (USA)
- Deutsche Forschungsgemeinschaft (DFG, Germany)
- Fonds Wetenschappelijk Onderzoek (FWO, Belgium)
- Onderzoeksraad, KU Leuven (Belgium)
Collapse
Affiliation(s)
- Sandra O. Tomé
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain InstituteKU LeuvenLeuvenBelgium
| | - Klara Gawor
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain InstituteKU LeuvenLeuvenBelgium
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain InstituteKU LeuvenLeuvenBelgium
- Department of PathologyUniversity Hospitals of LeuvenLeuvenBelgium
| |
Collapse
|
10
|
Pottier C, Küçükali F, Baker M, Batzler A, Jenkins GD, van Blitterswijk M, Vicente CT, De Coster W, Wynants S, Van de Walle P, Ross OA, Murray ME, Faura J, Haggarty SJ, van Rooij JG, Mol MO, Hsiung GYR, Graff C, Öijerstedt L, Neumann M, Asmann Y, McDonnell SK, Baheti S, Josephs KA, Whitwell JL, Bieniek KF, Forsberg L, Heuer H, Lago AL, Geier EG, Yokoyama JS, Oddi AP, Flanagan M, Mao Q, Hodges JR, Kwok JB, Domoto-Reilly K, Synofzik M, Wilke C, Onyike C, Dickerson BC, Evers BM, Dugger BN, Munoz DG, Keith J, Zinman L, Rogaeva E, Suh E, Gefen T, Geula C, Weintraub S, Diehl-Schmid J, Farlow MR, Edbauer D, Woodruff BK, Caselli RJ, Donker Kaat LL, Huey ED, Reiman EM, Mead S, King A, Roeber S, Nana AL, Ertekin-Taner N, Knopman DS, Petersen RC, Petrucelli L, Uitti RJ, Wszolek ZK, Ramos EM, Grinberg LT, Gorno Tempini ML, Rosen HJ, Spina S, Piguet O, Grossman M, Trojanowski JQ, Keene DC, Lee-Way J, Prudlo J, Geschwind DH, Rissman RA, Cruchaga C, Ghetti B, Halliday GM, Beach TG, Serrano GE, Arzberger T, Herms J, Boxer AL, Honig LS, Vonsattel JP, Lopez OL, Kofler J, White CL, Gearing M, Glass J, Rohrer JD, Irwin DJ, Lee EB, et alPottier C, Küçükali F, Baker M, Batzler A, Jenkins GD, van Blitterswijk M, Vicente CT, De Coster W, Wynants S, Van de Walle P, Ross OA, Murray ME, Faura J, Haggarty SJ, van Rooij JG, Mol MO, Hsiung GYR, Graff C, Öijerstedt L, Neumann M, Asmann Y, McDonnell SK, Baheti S, Josephs KA, Whitwell JL, Bieniek KF, Forsberg L, Heuer H, Lago AL, Geier EG, Yokoyama JS, Oddi AP, Flanagan M, Mao Q, Hodges JR, Kwok JB, Domoto-Reilly K, Synofzik M, Wilke C, Onyike C, Dickerson BC, Evers BM, Dugger BN, Munoz DG, Keith J, Zinman L, Rogaeva E, Suh E, Gefen T, Geula C, Weintraub S, Diehl-Schmid J, Farlow MR, Edbauer D, Woodruff BK, Caselli RJ, Donker Kaat LL, Huey ED, Reiman EM, Mead S, King A, Roeber S, Nana AL, Ertekin-Taner N, Knopman DS, Petersen RC, Petrucelli L, Uitti RJ, Wszolek ZK, Ramos EM, Grinberg LT, Gorno Tempini ML, Rosen HJ, Spina S, Piguet O, Grossman M, Trojanowski JQ, Keene DC, Lee-Way J, Prudlo J, Geschwind DH, Rissman RA, Cruchaga C, Ghetti B, Halliday GM, Beach TG, Serrano GE, Arzberger T, Herms J, Boxer AL, Honig LS, Vonsattel JP, Lopez OL, Kofler J, White CL, Gearing M, Glass J, Rohrer JD, Irwin DJ, Lee EB, Van Deerlin V, Castellani R, Mesulam MM, Tartaglia MC, Finger EC, Troakes C, Al-Sarraj S, Miller BL, Seelaar H, Graff-Radford NR, Boeve BF, Mackenzie IR, van Swieten JC, Seeley WW, Sleegers K, Dickson DW, Biernacka JM, Rademakers R. Deciphering Distinct Genetic Risk Factors for FTLD-TDP Pathological Subtypes via Whole-Genome Sequencing. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.24.24309088. [PMID: 38978643 PMCID: PMC11230325 DOI: 10.1101/2024.06.24.24309088] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Frontotemporal lobar degeneration with neuronal inclusions of the TAR DNA-binding protein 43 (FTLD-TDP) is a fatal neurodegenerative disorder with only a limited number of risk loci identified. We report our comprehensive genome-wide association study as part of the International FTLD-TDP Whole-Genome Sequencing Consortium, including 985 cases and 3,153 controls, and meta-analysis with the Dementia-seq cohort, compiled from 26 institutions/brain banks in the United States, Europe and Australia. We confirm UNC13A as the strongest overall FTLD-TDP risk factor and identify TNIP1 as a novel FTLD-TDP risk factor. In subgroup analyses, we further identify for the first time genome-wide significant loci specific to each of the three main FTLD-TDP pathological subtypes (A, B and C), as well as enrichment of risk loci in distinct tissues, brain regions, and neuronal subtypes, suggesting distinct disease aetiologies in each of the subtypes. Rare variant analysis confirmed TBK1 and identified VIPR1 , RBPJL , and L3MBTL1 as novel subtype specific FTLD-TDP risk genes, further highlighting the role of innate and adaptive immunity and notch signalling pathway in FTLD-TDP, with potential diagnostic and novel therapeutic implications.
Collapse
|
11
|
Nelson PT, Fardo DW, Wu X, Aung KZ, Cykowski MD, Katsumata Y. Limbic-predominant age-related TDP-43 encephalopathy (LATE-NC): Co-pathologies and genetic risk factors provide clues about pathogenesis. J Neuropathol Exp Neurol 2024; 83:396-415. [PMID: 38613823 PMCID: PMC11110076 DOI: 10.1093/jnen/nlae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2024] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is detectable at autopsy in more than one-third of people beyond age 85 years and is robustly associated with dementia independent of other pathologies. Although LATE-NC has a large impact on public health, there remain uncertainties about the underlying biologic mechanisms. Here, we review the literature from human studies that may shed light on pathogenetic mechanisms. It is increasingly clear that certain combinations of pathologic changes tend to coexist in aging brains. Although "pure" LATE-NC is not rare, LATE-NC often coexists in the same brains with Alzheimer disease neuropathologic change, brain arteriolosclerosis, hippocampal sclerosis of aging, and/or age-related tau astrogliopathy (ARTAG). The patterns of pathologic comorbidities provide circumstantial evidence of mechanistic interactions ("synergies") between the pathologies, and also suggest common upstream influences. As to primary mediators of vulnerability to neuropathologic changes, genetics may play key roles. Genes associated with LATE-NC include TMEM106B, GRN, APOE, SORL1, ABCC9, and others. Although the anatomic distribution of TDP-43 pathology defines the condition, important cofactors for LATE-NC may include Tau pathology, endolysosomal pathways, and blood-brain barrier dysfunction. A review of the human phenomenology offers insights into disease-driving mechanisms, and may provide clues for diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Peter T Nelson
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky, USA
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - David W Fardo
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Xian Wu
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Khine Zin Aung
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Yuriko Katsumata
- Department of Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
12
|
Carlos AF, Koga S, Graff-Radford NR, Baker MC, Rademakers R, Ross OA, Dickson DW, Josephs KA. Senile plaque-associated transactive response DNA-binding protein 43 in Alzheimer's disease: A case report spanning 16 years of memory loss. Neuropathology 2024; 44:115-125. [PMID: 37525358 PMCID: PMC10828111 DOI: 10.1111/neup.12938] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/15/2023] [Accepted: 07/10/2023] [Indexed: 08/02/2023]
Abstract
Transactive response DNA-binding protein 43 (TDP-43) pathological inclusions are found in frontotemporal lobar degeneration (FTLD-TDP) and Alzheimer's disease (AD-TDP). While clinically different, TDP-43 inclusions in FTLD-TDP and AD can have similar morphological characteristics. However, TDP-43 colocalizing with tau and forming "apple-bite" or "flame-shaped" neuronal cytoplasmic inclusions (NCI) are only found in AD-TDP. Here, we describe a case with AD and neuritic plaque-associated TDP-43. The patient was a 96-year-old right-handed Caucasian woman who had developed a slowly progressive amnestic syndrome compatible with typical AD at age 80. Genetic testing revealed APOE ε3/ε4, GRN r5848 CT, and MAPT H1/H2 genotype. Consistent with the old age at onset and long disease duration, limbic-predominant AD was found at autopsy, with high hippocampal yet low cortical neurofibrillary tangle (NFT) counts. Hippocampal and amygdala sclerosis were present. Immunohistochemistry for phospho-TDP-43 showed NCIs, dystrophic neurites, and rare neuronal intranuclear inclusions consistent with FTLD-TDP type A, as well as tau NFT-associated TDP-43 inclusions. These were frequent in the amygdala, entorhinal cortex, hippocampus, occipitotemporal gyrus, and inferior temporal gyrus but sparse in the mid-frontal cortex. Additionally, there were TDP-43-immunoreactive inclusions forming plaque-like structures in the molecular layer of the dentate fascia of the hippocampus. The presence of neuritic plaques in the same region was confirmed using thioflavin-S fluorescent microscopy and immunohistochemistry for phospho-tau. Double labeling immunofluorescence showed colocalization of TDP-43 and tau within neuritic plaques. Other pathologies included mild Lewy body pathology predominantly affecting the amygdala and olfactory bulb, aging-related tau astrogliopathy, and mixed small vessel disease (arteriolosclerosis and amyloid angiopathy) with several cortical microinfarcts. In conclusion, we have identified TDP-43 colocalizing with tau in neuritic plaques in AD, which expands the association of TDP-43 and tau in AD beyond NFTs. The clinical correlate of this plaque-associated TDP-43 appears to be a slowly progressive amnestic syndrome.
Collapse
Affiliation(s)
- Arenn F. Carlos
- Department of Neurology, Mayo Clinic, Rochester, MN 55905 USA
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32225 USA
| | | | - Matthew C. Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32225 USA
| | - Rosa Rademakers
- VIB Center for Molecular Neurology, VIB, Antwerp, Flanders 2000, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Flanders 2000, Belgium
| | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32225 USA
| | | | | |
Collapse
|
13
|
Youssef H, Gatto RG, Pham NTT, Petersen RC, Machulda MM, Reichard RR, Dickson DW, Jack CR, Whitwell JL, Josephs KA. TDP-43 Is Associated with Subiculum and Cornu Ammonis 1 Hippocampal Subfield Atrophy in Primary Age-Related Tauopathy. J Alzheimers Dis 2024; 99:1023-1032. [PMID: 38728190 PMCID: PMC11265226 DOI: 10.3233/jad-240136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
Background TAR DNA binding protein 43 (TDP-43) has been shown to be associated with whole hippocampal atrophy in primary age-related tauopathy (PART). It is currently unknown which subregions of the hippocampus are contributing to TDP-43 associated whole hippocampal atrophy in PART. Objective To identify which specific hippocampal subfield regions are contributing to TDP-43-associated whole hippocampal atrophy in PART. Methods A total of 115 autopsied cases from the Mayo Clinic Alzheimer Disease Research Center, Neurodegenerative Research Group, and the Mayo Clinic Study of Aging were analyzed. All cases underwent antemortem brain volumetric MRI, neuropathological assessment of the distribution of Aβ (Thal phase), and neurofibrillary tangle (Braak stage) to diagnose PART, as well as assessment of TDP-43 presence/absence in the amygdala, hippocampus and beyond. Hippocampal subfield segmentation was performed using FreeSurfer version 7.4.1. Statistical analyses using logistic regression were performed to assess for associations between TDP-43 and hippocampal subfield volumes, accounting for potential confounders. Results TDP-43 positive patients (n = 37, 32%), of which 15/15 were type-α, had significantly smaller whole hippocampal volumes, and smaller volumes of the body and tail of the hippocampus compared to TDP-43 negative patients. Subfield analyses revealed an association between TDP-43 and the molecular layer of hippocampal body and the body of cornu ammonis 1 (CA1), subiculum, and presubiculum regions. There was no association between TDP-43 stage and subfield volumes. Conclusions Whole hippocampal volume loss linked to TDP-43 in PART is mainly due to volume loss occurring in the molecular layer, CA1, subiculum and presubiculum of the hippocampal body.
Collapse
Affiliation(s)
- Hossam Youssef
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Mary M. Machulda
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - R. Ross Reichard
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Dennis W. Dickson
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL, USA
| | | | | | | |
Collapse
|
14
|
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: 16] [Impact Index Per Article: 8.0] [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
|
15
|
Carlos AF, Sekiya H, Koga S, Gatto RG, Casey MC, Pham NTT, Sintini I, Machulda MM, Jack CR, Lowe VJ, Whitwell JL, Petrucelli L, Reichard RR, Petersen RC, Dickson DW, Josephs KA. Clinicopathologic features of a novel star-shaped transactive response DNA-binding protein 43 (TDP-43) pathology in the oldest old. J Neuropathol Exp Neurol 2023; 83:36-52. [PMID: 38086178 PMCID: PMC10746697 DOI: 10.1093/jnen/nlad105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023] Open
Abstract
Transactive response DNA-binding protein 43 (TDP-43) pathology is categorized as type A-E in frontotemporal lobar degeneration and as type α-β in Alzheimer disease (AD) based on inclusion type. We screened amygdala slides of 131 cases with varying ages at death, clinical/neuroimaging findings, and AD neuropathologic changes for TDP-43 pathology using anti-phospho-TDP-43 antibodies. Seven cases (5%) only showed atypical TDP-43 inclusions that could not be typed. Immunohistochemistry and immunofluorescence assessed the atypical star-shaped TDP-43 pathology including its distribution, species, cellular localization, and colocalization with tau. All 7 had died at an extremely old age (median: 100 years [IQR: 94-101]) from nonneurological causes and none had dementia (4 cognitively unimpaired, 3 with amnestic mild cognitive impairment). Neuroimaging showed mild medial temporal involvement. Pathologically, the star-shaped TDP-43-positive inclusions were found in medial (subpial) amygdala and, occasionally, in basolateral regions. Hippocampus only showed TDP-43-positive neurites in the fimbria and subiculum while the frontal lobe was free of TDP-43 inclusions. The star-shaped inclusions were better detected with antibodies against N-terminal than C-terminal TDP-43. Double-labeling studies confirmed deposition of TDP-43 within astrocytes and colocalization with tau. We have identified a novel TDP-43 pathology with star-shaped morphology associated with superaging, with a homogeneous clinicopathologic picture, possibly representing a novel, true aging-related TDP-43 pathology.
Collapse
Affiliation(s)
- Arenn F Carlos
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Hiroaki Sekiya
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Rodolfo G Gatto
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Irene Sintini
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mary M Machulda
- Department of Psychiatry (Psychology), Mayo Clinic, Rochester, Minnesota, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - R Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
16
|
Minogue G, Kawles A, Zouridakis A, Keszycki R, Macomber A, Lubbat V, Gill N, Mao Q, Flanagan ME, Zhang H, Castellani R, Bigio EH, Mesulam MM, Geula C, Gefen T. Distinct Patterns of Hippocampal Pathology in Alzheimer's Disease with Transactive Response DNA-binding Protein 43. Ann Neurol 2023; 94:1036-1047. [PMID: 37592884 PMCID: PMC10872839 DOI: 10.1002/ana.26762] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/19/2023]
Abstract
OBJECTIVE Age-related dementia syndromes are often not related to a single pathophysiological process, leading to multiple neuropathologies found at autopsy. An amnestic dementia syndrome can be associated with Alzheimer's disease (AD) with comorbid transactive response DNA-binding protein 43 (TDP-43) pathology (AD/TDP). Here, we investigated neuronal integrity and pathological burden of TDP-43 and tau, along the well-charted trisynaptic hippocampal circuit (dentate gyrus [DG], CA3, and CA1) in participants with amnestic dementia due to AD/TDP, amnestic dementia due to AD alone, or non-amnestic dementia due to TDP-43 proteinopathy associated with frontotemporal lobar degeneration (FTLD-TDP). METHODS A total of 48 extensively characterized cases (14 AD, 16 AD/TDP, 18 FTLD-TDP) were analyzed using digital HALO software (Indica Labs, Albuquerque, NM, USA) to quantify pathological burden and neuronal loss. RESULTS In AD/TDP and FTLD-TDP, TDP-43 immunoreactivity was greatest in the DG. Tau immunoreactivity was significantly greater in DG and CA3 in AD/TDP compared with pure AD. All clinical groups showed the highest amounts of neurons in DG, followed by CA3, then CA1. The AD and AD/TDP groups showed lower neuronal counts compared with the FTLD-TDP group across all hippocampal subregions consistent with the salience of the amnestic phenotype. INTERPRETATION We conclude that AD/TDP can be distinguished from AD and FTLD-TDP based on differential regional distributions of hippocampal tau and TDP-43. Findings suggest that tau aggregation in AD/TDP might be enhanced by TDP-43. ANN NEUROL 2023;94:1036-1047.
Collapse
Affiliation(s)
- Grace Minogue
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Allegra Kawles
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Antonia Zouridakis
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Rachel Keszycki
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Alyssa Macomber
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Vivienne Lubbat
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Nathan Gill
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Qinwen Mao
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Margaret E. Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Hui Zhang
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Rudolph Castellani
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Eileen H. Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - M.-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
17
|
Niedowicz DM, Katsumata Y, Nelson PT. In severe ADNC, hippocampi with comorbid LATE-NC and hippocampal sclerosis have substantially more astrocytosis than those with LATE-NC or hippocampal sclerosis alone. J Neuropathol Exp Neurol 2023; 82:987-994. [PMID: 37935530 PMCID: PMC10658353 DOI: 10.1093/jnen/nlad085] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) and hippocampal sclerosis of aging (HS-A) pathologies are found together at autopsy in ∼20% of elderly demented persons. Although astrocytosis is known to occur in neurodegenerative diseases, it is currently unknown how the severity of astrocytosis is correlated with the common combinations of pathologies in aging brains. To address this knowledge gap, we analyzed a convenience sample of autopsied subjects from the University of Kentucky Alzheimer's Disease Research Center community-based autopsy cohort. The subjects were stratified into 5 groups (n = 51 total): pure ADNC, ADNC + LATE-NC, ADNC + HS-A, ADNC + LATE-NC + HS-A, and low-pathology controls. Following GFAP immunostaining and digital slide scanning with a ScanScope, we measured GFAP-immunoreactive astrocytosis. The severities of GFAP-immunoreactive astrocytosis in hippocampal subfield CA1 and subiculum were compared between groups. The group with ADNC + LATE-NC + HS-A had the most astrocytosis as operationalized by either any GFAP+ or strong GFAP+ immunoreactivity in both CA1 and subiculum. In comparison to that pathologic combination, ADNC + HS or ADNC + LATE-NC alone showed lower astrocytosis. Pure ADNC had only marginally increased astrocytosis in CA1 and subiculum, in comparison to low-pathology controls. We conclude that there appeared to be pathogenetic synergy such that ADNC + LATE-NC + HS-A cases had relatively high levels of astrocytosis in the hippocampal formation.
Collapse
|
18
|
Corriveau-Lecavalier N, Botha H, Graff-Radford J, Switzer AR, Przybelski SA, Wiste HJ, Murray ME, Reichard RR, Dickson DW, Nguyen AT, Ramanan VK, McCarter SJ, Boeve BF, Machulda MM, Fields JA, Stricker NH, Nelson PT, Grothe MJ, Knopman DS, Lowe VJ, Petersen RC, Jack CR, Jones DT. A limbic-predominant amnestic neurodegenerative syndrome associated with TDP-43 pathology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.19.23298314. [PMID: 38045300 PMCID: PMC10690340 DOI: 10.1101/2023.11.19.23298314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Limbic-predominant age-related TDP-43 encephalopathy (LATE) is a neuropathologically-defined disease that affects 40% of persons in advanced age, but its associated neurological syndrome is not defined. LATE neuropathological changes (LATE-NC) are frequently comorbid with Alzheimer's disease neuropathologic changes (ADNC). When seen in isolation, LATE-NC have been associated with a predominantly amnestic profile and slow clinical progression. We propose a set of clinical criteria for a limbic-predominant amnestic neurodegenerative syndrome (LANS) that is highly associated with LATE-NC but also other pathologic entities. The LANS criteria incorporate core, standard and advanced features that are measurable in vivo, including older age at evaluation, mild clinical syndrome, disproportionate hippocampal atrophy, impaired semantic memory, limbic hypometabolism, absence of neocortical degenerative patterns and low likelihood of neocortical tau, with degrees of certainty (highest, high, moderate, low). We operationalized this set of criteria using clinical, imaging and biomarker data to validate its associations with clinical and pathologic outcomes. We screened autopsied patients from Mayo Clinic (n = 922) and ADNI (n = 93) cohorts and applied the LANS criteria to those with an antemortem predominant amnestic syndrome (Mayo, n = 165; ADNI, n = 53). ADNC, ADNC/LATE-NC and LATE-NC accounted for 35%, 37% and 4% of cases in the Mayo cohort, respectively, and 30%, 22%, and 9% of cases in the ADNI cohort, respectively. The LANS criteria effectively categorized these cases, with ADNC having the lowest LANS likelihoods, LATE-NC patients having the highest likelihoods, and ADNC/LATE-NC patients having intermediate likelihoods. A logistic regression model using the LANS features as predictors of LATE-NC achieved a balanced accuracy of 74.6% in the Mayo cohort, and out-of-sample predictions in the ADNI cohort achieved a balanced accuracy of 73.3%. Patients with high LANS likelihoods had a milder and slower clinical course and more severe temporo-limbic degeneration compared to those with low likelihoods. Stratifying ADNC/LATE-NC patients from the Mayo cohort according to their LANS likelihood revealed that those with higher likelihoods had more temporo-limbic degeneration and a slower rate of cognitive decline, and those with lower likelihoods had more lateral temporo-parietal degeneration and a faster rate of cognitive decline. The implementation of LANS criteria has implications to disambiguate the different driving etiologies of progressive amnestic presentations in older age and guide prognosis, treatment, and clinical trials. The development of in vivo biomarkers specific to TDP-43 pathology are needed to refine molecular associations between LANS and LATE-NC and precise antemortem diagnoses of LATE.
Collapse
Affiliation(s)
- Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Heather J. Wiste
- Department of Quantitative Health Sciences, Mayo Clinic Rochester, MN, USA
| | | | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN, USA
| | | | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN, USA
| | | | | | | | - Mary M. Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Julie A. Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Nikki H. Stricker
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Peter T. Nelson
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Michel J. Grothe
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center, Madrid, Spain
- Wallenberg Center for Molecular and Translational Medicine and Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Clifford R. Jack
- Department of Neuroscience, Mayo Clinic Jacksonville, FL, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - David T. Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
19
|
Dijkstra AA, Morrema THJ, Hart de Ruyter FJ, Gami-Patel P, Verbraak FD, de Boer JF, Bouwman FH, Pijnenburg YAL, den Haan J, Rozemuller AJ, Hoozemans JJM. TDP-43 pathology in the retina of patients with frontotemporal lobar degeneration. Acta Neuropathol 2023; 146:767-770. [PMID: 37597044 PMCID: PMC10564657 DOI: 10.1007/s00401-023-02623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 08/21/2023]
Affiliation(s)
- Anke A Dijkstra
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands.
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
| | - Tjado H J Morrema
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Frederique J Hart de Ruyter
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Amsterdam UMC, Location VUmc, Alzheimer Center, Neurology, Amsterdam, The Netherlands
| | - Priya Gami-Patel
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Frank D Verbraak
- Ophthalmology Department, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Johannes F de Boer
- Department of Physics, Bio Laser Lab, VU University, Amsterdam, The Netherlands
| | - Femke H Bouwman
- Amsterdam UMC, Location VUmc, Alzheimer Center, Neurology, Amsterdam, The Netherlands
| | | | - Jurre den Haan
- Amsterdam UMC, Location VUmc, Alzheimer Center, Neurology, Amsterdam, The Netherlands
| | - Annemieke J Rozemuller
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Jeroen J M Hoozemans
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Oveisgharan S, Yu L, Agrawal S, Nag S, Bennett DA, Buchman AS, Schneider JA. Relation of Motor Impairments to Neuropathologic Changes of Limbic-Predominant Age-Related TDP-43 Encephalopathy in Older Adults. Neurology 2023; 101:e1542-e1553. [PMID: 37604667 PMCID: PMC10585698 DOI: 10.1212/wnl.0000000000207726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/14/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Limbic-predominant age-related transactive response DNA-binding protein 43 (TDP-43) encephalopathy neuropathologic change (LATE-NC) is common and is a major contributor to cognitive decline and Alzheimer dementia in older adults. The objective of the current study was to examine whether LATE-NC was also associated with declining motor function in older adults. METHODS Participants were from 2 longitudinal clinical pathologic studies of aging who did not have dementia at the time of enrollment. Postmortem pathologic examination included immunohistochemical staining for TDP-43 in 8 brain regions, which was summarized as a dichotomous variable indicating advanced LATE-NC stages at which TDP-43 pathology had accumulated in the hippocampus, entorhinal, or neocortical regions. Annual motor testing included maximal inspiratory and expiratory pressures (summarized as respiratory muscle strength), grip and pinch strength (summarized as hand strength), finger tapping speed and the Purdue Pegboard Test (summarized as hand dexterity), and walking 8 feet and turning 360° (summarized as gait function). The severity of parkinsonism was also assessed and summarized as a global parkinsonism score. Global cognition was a summary of standardized scores of 19 neuropsychological tests. We used linear mixed-effect models to examine the associations of LATE-NC with longitudinal changes of motor decline and used multivariate random coefficient models to simultaneously examine the associations of LATE-NC with cognitive and motor decline. RESULTS Among 1,483 participants (mean age at death 90.1 [SD = 6.4] years, 70% women, mean follow-up 7.4 [SD = 3.8] years), LATE-NC was present in 34.0% (n = 504). In separate linear mixed-effect models controlling for demographics and other brain pathologies, LATE-NC was associated with faster decline in respiratory muscle strength (estimate = -0.857, SE = 0.322, p = 0.008) and hand strength (estimate = -0.005, SE = 0.002, p = 0.005) but was not related to hand dexterity, gait function, or parkinsonism. In multivariate random coefficient models including respiratory muscle strength, hand strength, and global cognition as the outcomes, LATE-NC remained associated with a faster respiratory muscle strength decline rate (estimate = -0.021, SE = 0.009, p = 0.023), but the association with hand strength was no longer significant (estimate = -0.002, SE = 0.003, p = 0.390). DISCUSSION Motor impairment, specifically respiratory muscle weakness, may be an unrecognized comorbidity of LATE-NC that highlights the potential association of TDP-43 proteinopathy with noncognitive phenotypes in aging adults.
Collapse
Affiliation(s)
- Shahram Oveisgharan
- From the Rush Alzheimer's Disease Center (S.O., L.Y., S.A., S.N., D.A.B., A.S.B., J.A.S.), Department of Neurological Sciences (S.O., L.Y., D.A.B., A.S.B., J.A.S.), and Department of Pathology (S.A., S.N., J.A.S.), Rush University Medical Center, Chicago, IL.
| | - Lei Yu
- From the Rush Alzheimer's Disease Center (S.O., L.Y., S.A., S.N., D.A.B., A.S.B., J.A.S.), Department of Neurological Sciences (S.O., L.Y., D.A.B., A.S.B., J.A.S.), and Department of Pathology (S.A., S.N., J.A.S.), Rush University Medical Center, Chicago, IL
| | - Sonal Agrawal
- From the Rush Alzheimer's Disease Center (S.O., L.Y., S.A., S.N., D.A.B., A.S.B., J.A.S.), Department of Neurological Sciences (S.O., L.Y., D.A.B., A.S.B., J.A.S.), and Department of Pathology (S.A., S.N., J.A.S.), Rush University Medical Center, Chicago, IL
| | - Sukriti Nag
- From the Rush Alzheimer's Disease Center (S.O., L.Y., S.A., S.N., D.A.B., A.S.B., J.A.S.), Department of Neurological Sciences (S.O., L.Y., D.A.B., A.S.B., J.A.S.), and Department of Pathology (S.A., S.N., J.A.S.), Rush University Medical Center, Chicago, IL
| | - David A Bennett
- From the Rush Alzheimer's Disease Center (S.O., L.Y., S.A., S.N., D.A.B., A.S.B., J.A.S.), Department of Neurological Sciences (S.O., L.Y., D.A.B., A.S.B., J.A.S.), and Department of Pathology (S.A., S.N., J.A.S.), Rush University Medical Center, Chicago, IL
| | - Aron S Buchman
- From the Rush Alzheimer's Disease Center (S.O., L.Y., S.A., S.N., D.A.B., A.S.B., J.A.S.), Department of Neurological Sciences (S.O., L.Y., D.A.B., A.S.B., J.A.S.), and Department of Pathology (S.A., S.N., J.A.S.), Rush University Medical Center, Chicago, IL
| | - Julie A Schneider
- From the Rush Alzheimer's Disease Center (S.O., L.Y., S.A., S.N., D.A.B., A.S.B., J.A.S.), Department of Neurological Sciences (S.O., L.Y., D.A.B., A.S.B., J.A.S.), and Department of Pathology (S.A., S.N., J.A.S.), Rush University Medical Center, Chicago, IL
| |
Collapse
|
21
|
Torso M, Ridgway GR, Valotti M, Hardingham I, Chance SA. In vivo cortical diffusion imaging relates to Alzheimer's disease neuropathology. Alzheimers Res Ther 2023; 15:165. [PMID: 37794477 PMCID: PMC10548768 DOI: 10.1186/s13195-023-01309-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 09/17/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND There has been increasing interest in cortical microstructure as a complementary and earlier measure of neurodegeneration than macrostructural atrophy, but few papers have related cortical diffusion imaging to post-mortem neuropathology. This study aimed to characterise the associations between the main Alzheimer's disease (AD) neuropathological hallmarks and multiple cortical microstructural measures from in vivo diffusion MRI. Comorbidities and co-pathologies were also investigated. METHODS Forty-three autopsy cases (8 cognitively normal, 9 mild cognitive impairment, 26 AD) from the National Alzheimer's Coordinating Center and Alzheimer's Disease Neuroimaging Initiative databases were included. Structural and diffusion MRI scans were analysed to calculate cortical minicolumn-related measures (AngleR, PerpPD+, and ParlPD) and mean diffusivity (MD). Neuropathological hallmarks comprised Thal phase, Braak stage, neuritic plaques, and combined AD neuropathological changes (ADNC-the "ABC score" from NIA-AA recommendations). Regarding comorbidities, relationships between cortical microstructure and severity of white matter rarefaction (WMr), cerebral amyloid angiopathy (CAA), atherosclerosis of the circle of Willis (ACW), and locus coeruleus hypopigmentation (LCh) were investigated. Finally, the effect of coexistent pathologies-Lewy body disease and TAR DNA-binding protein 43 (TDP-43)-on cortical microstructure was assessed. RESULTS Cortical diffusivity measures were significantly associated with Thal phase, Braak stage, ADNC, and LCh. Thal phase was associated with AngleR in temporal areas, while Braak stage was associated with PerpPD+ in a wide cortical pattern, involving mainly temporal and limbic areas. A similar association was found between ADNC (ABC score) and PerpPD+. LCh was associated with PerpPD+, ParlPD, and MD. Co-existent neuropathologies of Lewy body disease and TDP-43 exhibited significantly reduced AngleR and MD compared to ADNC cases without co-pathology. CONCLUSIONS Cortical microstructural diffusion MRI is sensitive to AD neuropathology. The associations with the LCh suggest that cortical diffusion measures may indirectly reflect the severity of locus coeruleus neuron loss, perhaps mediated by the severity of microglial activation and tau spreading across the brain. Recognizing the impact of co-pathologies is important for diagnostic and therapeutic decision-making. Microstructural markers of neurodegeneration, sensitive to the range of histopathological features of amyloid, tau, and monoamine pathology, offer a more complete picture of cortical changes across AD than conventional structural atrophy.
Collapse
|
22
|
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: 18] [Impact Index Per Article: 9.0] [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
|
23
|
Estades Ayuso V, Pickles S, Todd T, Yue M, Jansen-West K, Song Y, González Bejarano J, Rawlinson B, DeTure M, Graff-Radford NR, Boeve BF, Knopman DS, Petersen RC, Dickson DW, Josephs KA, Petrucelli L, Prudencio M. TDP-43-regulated cryptic RNAs accumulate in Alzheimer's disease brains. Mol Neurodegener 2023; 18:57. [PMID: 37605276 PMCID: PMC10441763 DOI: 10.1186/s13024-023-00646-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/04/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Inclusions of TAR DNA-binding protein 43 kDa (TDP-43) has been designated limbic-predominant, age-related TDP-43 encephalopathy (LATE), with or without co-occurrence of Alzheimer's disease (AD). Approximately, 30-70% AD cases present TDP-43 proteinopathy (AD-TDP), and a greater disease severity compared to AD patients without TDP-43 pathology. However, it remains unclear to what extent TDP-43 dysfunction is involved in AD pathogenesis. METHODS To investigate whether TDP-43 dysfunction is a prominent feature in AD-TDP cases, we evaluated whether non-conserved cryptic exons, which serve as a marker of TDP-43 dysfunction in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD-TDP), accumulate in AD-TDP brains. We assessed a cohort of 192 post-mortem brains from three different brain regions: amygdala, hippocampus, and frontal cortex. Following RNA and protein extraction, qRT-PCR and immunoassays were performed to quantify the accumulation of cryptic RNA targets and phosphorylated TDP-43 pathology, respectively. RESULTS We detected the accumulation of misspliced cryptic or skiptic RNAs of STMN2, KCNQ2, UNC13A, CAMK2B, and SYT7 in the amygdala and hippocampus of AD-TDP cases. The topographic distribution of cryptic RNA accumulation mimicked that of phosphorylated TDP-43, regardless of TDP-43 subtype classification. Further, cryptic RNAs efficiently discriminated AD-TDP cases from controls. CONCLUSIONS Overall, our results indicate that cryptic RNAs may represent an intriguing new therapeutic and diagnostic target in AD, and that methods aimed at detecting and measuring these species in patient biofluids could be used as a reliable tool to assess TDP-43 pathology in AD. Our work also raises the possibility that TDP-43 dysfunction and related changes in cryptic splicing could represent a common molecular mechanism shared between AD-TDP and FTLD-TDP.
Collapse
Affiliation(s)
- Virginia Estades Ayuso
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Sarah Pickles
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Tiffany Todd
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Mei Yue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Yuping Song
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | | | | | | | | | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | | | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Mercedes Prudencio
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA.
- Department of Research, Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
| |
Collapse
|
24
|
Asken BM, Tanner JA, Gaynor LS, VandeVrede L, Mantyh WG, Casaletto KB, Staffaroni AM, Fonseca C, Shankar R, Grant H, Smith K, Lago AL, Xu H, La Joie R, Cobigo Y, Rosen H, Perry DC, Rojas JC, Miller BL, Gardner RC, Wang KKW, Kramer JH, Rabinovici GD. Alzheimer's pathology is associated with altered cognition, brain volume, and plasma biomarker patterns in traumatic encephalopathy syndrome. Alzheimers Res Ther 2023; 15:126. [PMID: 37480088 PMCID: PMC10360257 DOI: 10.1186/s13195-023-01275-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 07/12/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Traumatic encephalopathy syndrome (TES) is a clinical phenotype sensitive but non-specific to underlying chronic traumatic encephalopathy (CTE) neuropathology. However, cognitive symptoms of TES overlap with Alzheimer's disease (AD), and features of AD pathology like beta-amyloid (Aβ) plaques often co-occur with CTE, making clinical-to-pathological conclusions of TES diagnoses challenging. We investigated how Alzheimer's neuropathological changes associated with cognition, brain volume, and plasma biomarkers in patients with repetitive head impacts (RHI)/TES, clinical AD, or typically aging controls. METHODS We studied 154 participants including 33 with RHI/TES (age 61.5 ± 11.5, 100% male, 11/33 Aβ[ +]), 62 with AD and no known prior RHI (age 67.1 ± 10.2, 48% male, 62/62 Aβ[ +]), and 59 healthy controls without RHI (HC; age 73.0 ± 6.2, 40% male, 0/59 Aβ[ +]). Patients completed neuropsychological testing (memory, executive functioning, language, visuospatial) and structural MRI (voxel-based morphometry analysis), and provided plasma samples analyzed for GFAP, NfL, IL-6, IFN-γ, and YKL-40. For cognition and plasma biomarkers, patients with RHI/TES were stratified as Aβ[ +] or Aβ[ -] and compared to each other plus the AD and HC groups (ANCOVA adjusting for age and sex). Differences with at least a medium effect size (Cohen's d > 0.50) were interpreted as potentially meaningful. RESULTS Cognitively, within the TES group, Aβ[ +] RHI/TES performed worse than Aβ[-] RHI/TES on visuospatial (p = .04, d = 0.86) and memory testing (p = .07, d = 0.74). Comparing voxel-wise brain volume, both Aβ[ +] and Aβ[ -] RHI/TES had lower medial and anterior temporal lobe volume than HC and did not significantly differ from AD. Comparing plasma biomarkers, Aβ[ +] RHI/TES had higher plasma GFAP than HC (p = .01, d = 0.88) and did not significantly differ from AD. Conversely, Aβ[ -] RHI/TES had higher NfL than HC (p = .004, d = 0.93) and higher IL-6 than all other groups (p's ≤ .004, d's > 1.0). CONCLUSIONS Presence of Alzheimer's pathology in patients with RHI/TES is associated with altered cognitive and biomarker profiles. Patients with RHI/TES and positive Aβ-PET have cognitive and plasma biomarker changes that are more like patients with AD than patients with Aβ[ -] RHI/TES. Measuring well-validated Alzheimer's biomarkers in patients with RHI/TES could improve interpretation of research findings and heighten precision in clinical management.
Collapse
Affiliation(s)
- Breton M Asken
- Department of Clinical & Health Psychology, 1Florida Alzheimer's Disease Research Center, University of Florida, 1225 Center Drive, Gainesville, FL, 32610, USA.
| | - Jeremy A Tanner
- Department of Neurology, Biggs Institute for Alzheimer's and Neurodegenerative Diseases, South Texas Alzheimer's Disease Research Center, University of Texas Health - San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Leslie S Gaynor
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Lawren VandeVrede
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - William G Mantyh
- Department of Neurology, University of Minnesota, PWB 12-100, 516 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Kaitlin B Casaletto
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Adam M Staffaroni
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Corrina Fonseca
- Department of Neuroscience, Helen Wills Neuroscience Institute, University of California, 132 Barker Hall MC#3190, Berkeley, CA, 94720, USA
| | - Ranjani Shankar
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Harli Grant
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Karen Smith
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Argentina Lario Lago
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Haiyan Xu
- Department of Surgery, University of Florida, PO Box 100128, Gainesville, FL, 32610, USA
| | - Renaud La Joie
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Yann Cobigo
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Howie Rosen
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - David C Perry
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Julio C Rojas
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Bruce L Miller
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Raquel C Gardner
- Sheba Medical Center, Tel Hashomer City of Health, Tel Aviv District, Derech Sheba 2, Ramat Gan, Israel
| | - Kevin K W Wang
- Department of Neurobiology, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA
| | - Joel H Kramer
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Gil D Rabinovici
- Department of Neurology, Weill Institute for Neurosciences, Memory and Aging Center, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| |
Collapse
|
25
|
Nelson RS, Abner EL, Jicha GA, Schmitt FA, Di J, Wilcock DM, Barber JM, Van Eldik LJ, Katsumata Y, Fardo DW, Nelson PT. Neurodegenerative pathologies associated with behavioral and psychological symptoms of dementia in a community-based autopsy cohort. Acta Neuropathol Commun 2023; 11:89. [PMID: 37269007 PMCID: PMC10236713 DOI: 10.1186/s40478-023-01576-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 06/04/2023] Open
Abstract
In addition to the memory disorders and global cognitive impairment that accompany neurodegenerative diseases, behavioral and psychological symptoms of dementia (BPSD) commonly impair quality of life and complicate clinical management. To investigate clinical-pathological correlations of BPSD, we analyzed data from autopsied participants from the community-based University of Kentucky Alzheimer's Disease Research Center longitudinal cohort (n = 368 research volunteers met inclusion criteria, average age at death 85.4 years). Data assessing BPSD were obtained approximately annually, including parameters for agitation, anxiety, apathy, appetite problems, delusions, depression, disinhibition, hallucinations, motor disturbance, and irritability. Each BPSD was scored on a severity scale (0-3) via the Neuropsychiatric Inventory Questionnaire (NPI-Q). Further, Clinical Dementia Rating (CDR)-Global and -Language evaluations (also scored on 0-3 scales) were used to indicate the degree of global cognitive and language impairment. The NPI-Q and CDR ratings were correlated with neuropathology findings at autopsy: Alzheimer's disease neuropathological changes (ADNC), neocortical and amygdala-only Lewy bodies (LBs), limbic predominant age-related TDP-43 encephalopathy neuropathologic changes (LATE-NC), primary age-related tauopathy (PART), hippocampal sclerosis, and cerebrovascular pathologies. Combinations of pathologies included the quadruple misfolding proteinopathy (QMP) phenotype with co-occurring ADNC, neocortical LBs, and LATE-NC. Statistical models were used to estimate the associations between BPSD subtypes and pathologic patterns. Individuals with severe ADNC (particularly those with Braak NFT stage VI) had more BPSD, and the QMP phenotype was associated with the highest mean number of BPSD symptoms: > 8 different BPSD subtypes per individual. Disinhibition and language problems were common in persons with severe ADNC but were not specific to any pathology. "Pure" LATE-NC was associated with global cognitive impairment, apathy, and motor disturbance, but again, these were not specific associations. In summary, Braak NFT stage VI ADNC was strongly associated with BPSD, but no tested BPSD subtype was a robust indicator of any particular "pure" or mixed pathological combination.
Collapse
Affiliation(s)
| | - Erin L Abner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Epidemiology and Environmental Health, University of Kentucky, Lexington, KY, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Jing Di
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, USA
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Justin M Barber
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - David W Fardo
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, USA.
- University of Kentucky, Rm 575 Todd Building, Lexington, KY, 40536, USA.
| |
Collapse
|
26
|
Carlos AF, Tosakulwong N, Weigand SD, Senjem ML, Schwarz CG, Knopman DS, Boeve BF, Petersen RC, Nguyen AT, Reichard RR, Dickson DW, Jack CR, Lowe V, Whitwell JL, Josephs KA. TDP-43 pathology effect on volume and flortaucipir uptake in Alzheimer's disease. Alzheimers Dement 2023; 19:2343-2354. [PMID: 36463537 PMCID: PMC10239529 DOI: 10.1002/alz.12878] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/18/2022] [Accepted: 10/21/2022] [Indexed: 12/07/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) patients ≥70 years show smaller medial temporal volumes despite less 18 F-flortaucipir-positron emission tomography (PET) uptake than younger counterparts. We investigated whether TAR DNA-binding protein 43 (TDP-43) was contributing to this volume-uptake mismatch. METHODS Seventy-seven participants with flortaucipir-PET and volumetric magnetic resonance imaging underwent postmortem AD and TDP-43 pathology assessments. Bivariate-response linear regression estimated the effect of age and TDP-43 pathology on volume and/or flortaucipir standardized uptake volume ratios of the hippocampus, amygdala, entorhinal, inferior temporal, and midfrontal cortices. RESULTS Older participants had lower hippocampal volumes and overall flortaucipir uptake. TDP-43-immunoreactivity correlated with reduced medial temporal volumes but was unrelated to flortaucipir uptake. TDP-43 effect size was consistent across the age spectrum. However, at older ages, the cohort mean volumes moved toward those of TDP-43-positives, reflecting the increasing TDP-43 pathology frequency with age. DISCUSSION TDP-43 pathology is a relevant contributor driving the volume-uptake mismatch in older AD participants. HIGHLIGHTS TDP-43 pathology affects medial temporal volume loss but not tau radiotracer uptake. Greater TDP-43 pathology effect is seen in old age due to its increasing frequency. TDP-43 pathology is a relevant driver of the volume-uptake mismatch in old AD patients.
Collapse
Affiliation(s)
- Arenn F. Carlos
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905 USA
| | - Nirubol Tosakulwong
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905 USA
| | - Stephen D. Weigand
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905 USA
| | - Matthew L. Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905 USA
- Department of Information Technology, Mayo Clinic, Rochester, MN, 55905 USA
| | | | | | | | | | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905 USA
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905 USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224 USA
| | | | - Val Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905 USA
| | | | | |
Collapse
|
27
|
Llamas-Rodríguez J, Oltmer J, Marshall M, Champion S, Frosch MP, Augustinack JC. TDP-43 and tau concurrence in the entorhinal subfields in primary age-related tauopathy and preclinical Alzheimer's disease. Brain Pathol 2023:e13159. [PMID: 37037195 DOI: 10.1111/bpa.13159] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023] Open
Abstract
Phosphorylated tau (p-tau) pathology correlates strongly with cognitive decline and is a pathological hallmark of Alzheimer's Disease (AD). In recent years, phosphorylated transactive response DNA-binding protein (pTDP-43) has emerged as a common comorbidity, found in up to 70% of all AD cases (Josephs et al., Acta Neuropathol, 131(4), 571-585; Josephs, Whitwell, et al., Acta Neuropathol, 127(6), 811-824). Current staging schemes for pTDP-43 in AD and primary age-related tauopathy (PART) track its progression throughout the brain, but the distribution of pTDP-43 within the entorhinal cortex (EC) at the earliest stages has not been studied. Moreover, the exact nature of p-tau and pTDP-43 co-localization is debated. We investigated the selective vulnerability of the entorhinal subfields to phosphorylated pTDP-43 pathology in preclinical AD and PART postmortem tissue. Within the EC, posterior-lateral subfields showed the highest semi-quantitative pTDP-43 density scores, while the anterior-medial subfields had the lowest. On the rostrocaudal axis, pTDP-43 scores were higher posteriorly than anteriorly (p < 0.010), peaking at the posterior-most level (p < 0.050). Further, we showed the relationship between pTDP-43 and p-tau in these regions at pathology-positive but clinically silent stages. P-tau and pTDP-43 presented a similar pattern of affected subregions (p < 0.0001) but differed in density magnitude (p < 0.0001). P-tau burden was consistently higher than pTDP-43 at every anterior-posterior level and in most EC subfields. These findings highlight pTDP-43 burden heterogeneity within the EC and the posterior-lateral subfields as the most vulnerable regions within stage II of the current pTDP-43 staging schemes for AD and PART. The EC is a point of convergence for p-tau and pTDP-43 and identifying its most vulnerable neuronal populations will prove key for early diagnosis and disease intervention.
Collapse
Affiliation(s)
- Josué Llamas-Rodríguez
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Jan Oltmer
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Michael Marshall
- Department of Neuropathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Samantha Champion
- Department of Neuropathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Matthew P Frosch
- Department of Neuropathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jean C Augustinack
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| |
Collapse
|
28
|
Santillo AF, Leuzy A, Honer M, Landqvist Waldö M, Tideman P, Harper L, Ohlsson T, Moes S, Giannini L, Jögi J, Groot C, Ossenkoppele R, Strandberg O, van Swieten J, Smith R, Hansson O. [ 18F]RO948 tau positron emission tomography in genetic and sporadic frontotemporal dementia syndromes. Eur J Nucl Med Mol Imaging 2023; 50:1371-1383. [PMID: 36513817 PMCID: PMC10027632 DOI: 10.1007/s00259-022-06065-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE To examine [18F]RO948 retention in FTD, sampling the underlying protein pathology heterogeneity. METHODS A total of 61 individuals with FTD (n = 35), matched cases of AD (n = 13) and Aβ-negative cognitively unimpaired individuals (n = 13) underwent [18F]RO948PET and MRI. FTD included 21 behavioral variant FTD (bvFTD) cases, 11 symptomatic C9orf72 mutation carriers, one patient with non-genetic bvFTD-ALS, one individual with bvFTD due to a GRN mutation, and one due to a MAPT mutation (R406W). Tracer retention was examined using a region-of-interest and voxel-wise approaches. Two individuals (bvFTD due to C9orf72) underwent postmortem neuropathological examination. Tracer binding was additionally assessed in vitro using [3H]RO948 autoradiography in six separate cases. RESULTS [18F]RO948 retention across ROIs was clearly lower than in AD and comparable to that in Aβ-negative cognitively unimpaired individuals. Only minor loci of tracer retention were seen in bvFTD; these did not overlap with the observed cortical atrophy in the cases, the expected pattern of atrophy, nor the expected or verified protein pathology distribution. Autoradiography analyses showed no specific [3H]RO948 binding. The R406W MAPT mutation carriers were clear exceptions with AD-like retention levels and specific in-vitro binding. CONCLUSION [18F]RO948 uptake is not significantly increased in the majority of FTD patients, with a clear exception being specific MAPT mutations.
Collapse
Affiliation(s)
- Alexander F Santillo
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden.
- Memory Clinic, Skåne University Hospital, SE-20502, Malmö, Sweden.
| | - Antoine Leuzy
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
| | - Michael Honer
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Maria Landqvist Waldö
- Clinical Sciences Helsingborg, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Pontus Tideman
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
| | - Luke Harper
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
| | - Tomas Ohlsson
- Radiation Physics, Skane University Hospital, Scania, Sweden
| | - Svenja Moes
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Lucia Giannini
- Alzheimer Center, Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jonas Jögi
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Colin Groot
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
| | - Rik Ossenkoppele
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Olof Strandberg
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
| | - John van Swieten
- Alzheimer Center, Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ruben Smith
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- Department of Clinical Sciences, Clinical Memory Research Unit, Faculty of Medicine, Lund University, Lund/Malmö, Sweden
- Memory Clinic, Skåne University Hospital, SE-20502, Malmö, Sweden
| |
Collapse
|
29
|
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: 81] [Impact Index Per Article: 40.5] [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
|
30
|
Josephs KA, Koga S, Tosakulwong N, Weigand SD, Nha Pham TT, Baker M, Whitwell JL, Rademakers R, Petrucelli L, Dickson DW. Molecular fragment characteristics and distribution of tangle associated TDP-43 (TATs) and other TDP-43 lesions in Alzheimer's disease. FREE NEUROPATHOLOGY 2023; 4:22. [PMID: 38093787 PMCID: PMC10716685 DOI: 10.17879/freeneuropathology-2023-5192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/03/2023] [Indexed: 03/07/2024]
Abstract
TAR DNA binding protein 43 (TDP-43) pathology is a defining feature of frontotemporal lobar degeneration (FTLD). In FTLD-TDP there is a moderate-to-high burden of morphologically distinctive TDP-43 immunoreactive inclusions distributed throughout the brain. In Alzheimer's disease (AD), similar TDP-43 immunoreactive inclusions are observed. In AD, however, there is a unique phenomenon of neurofibrillary tangle-associated TDP-43 (TATs) whereby TDP-43 intermingles with neurofibrillary tangles. Little is known about the characteristics and distribution of TATs, or how burden and distribution of TATs compares to burden and distribution of other FTLD-TDP-like lesions observed in AD. Here we characterize molecular fragment characteristics, burden and distribution of TATs and assess how these features compare to features of other TDP-43 lesions. We performed TDP-43 immunohistochemistry with anti-phosphorylated, C- and N-terminal TDP-43 antibodies in 20 high-probability AD cases and semi-quantitative burden of seven inclusion types within five brain regions (entorhinal cortex, subiculum, CA1 and dentate gyrus of hippocampus, occipitotemporal cortex). Hierarchical cluster analysis was used to analyze the dataset that consisted of 75 different combinations of neuropathological features. TATs were nonspherical with heterogeneous staining patterns and present in all regions except hippocampal dentate. All three antibodies detected TATs although N-terminal antibody sensitivity was low. Three clusters were identified: Cluster-1 had mild-moderate TATs, moderate-frequent neuronal cytoplasmic inclusions, dystrophic neurites, neuronal intranuclear inclusions and fine neurites, and perivascular and granular inclusions identified only with the N-terminal antibody throughout the brain; Cluster-2 had scant TATs in limbic regions and Cluster-3 mild-moderate TATs and mild-moderate neuronal cytoplasmic inclusions and dystrophic neurites throughout the brain and moderate fine neurites. Only 17% of cluster 1 cases had the TMEM106b GG (protective) haplotype and 83% had hippocampal sclerosis. Both features differed across clusters (p=0.03 & p=0.01). TATs have molecular characteristics, distribution and burden, and genetic and pathologic associations like FTLD-TDP lesions.
Collapse
Affiliation(s)
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Nirubol Tosakulwong
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen D Weigand
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Matt Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | | |
Collapse
|
31
|
Carlos AF, Machulda MM, Rutledge MH, Nguyen AT, Reichard RR, Baker MC, Rademakers R, Dickson DW, Petersen RC, Josephs KA. Comparison of Clinical, Genetic, and Pathologic Features of Limbic and Diffuse Transactive Response DNA-Binding Protein 43 Pathology in Alzheimer's Disease Neuropathologic Spectrum. J Alzheimers Dis 2023; 93:1521-1535. [PMID: 37182869 PMCID: PMC10923399 DOI: 10.3233/jad-221094] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Increasing evidence suggests that TAR DNA-binding protein 43 (TDP-43) pathology in Alzheimer's disease (AD), or AD-TDP, can be diffuse or limbic-predominant. Understanding whether diffuse AD-TDP has genetic, clinical, and pathological features that differ from limbic AD-TDP could have clinical and research implications. OBJECTIVE To better characterize the clinical and pathologic features of diffuse AD-TDP and differentiate it from limbic AD-TDP. METHODS 363 participants from the Mayo Clinic Study of Aging, Alzheimer's Disease Research Center, and Neurodegenerative Research Group with autopsy confirmed AD and TDP-43 pathology were included. All underwent genetic, clinical, neuropsychologic, and neuropathologic evaluations. AD-TDP pathology distribution was assessed using the Josephs 6-stage scale. Stages 1-3 were classified as Limbic, those 4-6 as Diffuse. Multivariable logistic regression was used to identify clinicopathologic features that independently predicted diffuse pathology. RESULTS The cohort was 61% female and old at onset (median: 76 years [IQR:70-82]) and death (median: 88 years [IQR:82-92]). Fifty-four percent were Limbic and 46% Diffuse. Clinically, ∼10-20% increases in odds of being Diffuse associated with 5-year increments in age at onset (p = 0.04), 1-year longer disease duration (p = 0.02), and higher Neuropsychiatric Inventory scores (p = 0.03), while 15-second longer Trailmaking Test-B times (p = 0.02) and higher Block Design Test scores (p = 0.02) independently decreased the odds by ~ 10-15%. There was evidence for association of APOEɛ4 allele with limbic AD-TDP and of TMEM106B rs3173615 C allele with diffuse AD-TDP. Pathologically, widespread amyloid-β plaques (Thal phases: 3-5) decreased the odds of diffuse TDP-43 pathology by 80-90%, while hippocampal sclerosis increased it sixfold (p < 0.001). CONCLUSION Diffuse AD-TDP shows clinicopathologic and genetic features different from limbic AD-TDP.
Collapse
Affiliation(s)
- Arenn F. Carlos
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mary M. Machulda
- Department of Psychology and Psychiatry, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew C. Baker
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rosa Rademakers
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL 32224, USA
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Flanders 2000, Belgium
| | - Dennis W. Dickson
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | |
Collapse
|
32
|
The central role of tau in Alzheimer’s disease: From neurofibrillary tangle maturation to the induction of cell death. Brain Res Bull 2022; 190:204-217. [DOI: 10.1016/j.brainresbull.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022]
|
33
|
Murakami A, Koga S, Sekiya H, Oskarsson B, Boylan K, Petrucelli L, Josephs KA, Dickson DW. Old age amyotrophic lateral sclerosis and limbic TDP-43 pathology. Brain Pathol 2022; 32:e13100. [PMID: 35715944 PMCID: PMC9616086 DOI: 10.1111/bpa.13100] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/01/2022] [Indexed: 01/01/2023] Open
Abstract
This study aimed to assess and compare the burden of transactive response DNA-binding protein of 43 kDa (TDP-43) pathology and clinical features of amyotrophic lateral sclerosis (ALS) in three age groups. All cases were from the Mayo Clinic brain bank for neurodegenerative disorders and most were followed longitudinally in the ALS Clinic. Cases with moderate-to-severe Alzheimer's disease neuropathological change were excluded. The 55 cases included in the study were divided into three groups by age at death: 75 years or older (old-ALS, n = 8), 64-74 years (middle-ALS, n = 23), and 63 years or younger (young-ALS, n = 24). Clinical features, including disease duration, initial symptoms, and ALS Cognitive Behavior Score (ALS-CBS), were summarized. Sections of paraffin-embedded tissue from the motor cortex, basal forebrain, medial temporal lobe, and middle frontal gyrus were processed for phospho-TDP-43 immunohistochemistry. The burden of TDP-43 pathology was analyzed using digital image analysis. The TDP-43 burden in the limbic system (i.e., amygdala, dentate gyrus and CA1 sector of the hippocampus, subiculum, and entorhinal cortex) was greater in old-ALS than in young-ALS and middle-ALS. TDP-43 burden in the middle frontal gyrus was sparse and did not differ between the three groups. The average of ALS-CBS was not different between the three groups. The present study shows that the amygdala and hippocampus are vulnerable to TDP-43 pathology in older patients with ALS. We discuss the evidence for and against this pathology being related to concurrent limbic-predominant, age-related TDP-43 encephalopathy neuropathologic change.
Collapse
Affiliation(s)
- Aya Murakami
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Shunsuke Koga
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Hiroaki Sekiya
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Kevin Boylan
- Department of NeurologyMayo ClinicJacksonvilleFloridaUSA
| | | | | | | |
Collapse
|
34
|
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: 1.7] [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
| | - 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
| |
Collapse
|
35
|
Koper MJ, Tomé SO, Gawor K, Belet A, Van Schoor E, Schaeverbeke J, Vandenberghe R, Vandenbulcke M, Ghebremedhin E, Otto M, von Arnim CAF, Balusu S, Blaschko MB, De Strooper B, Thal DR. LATE-NC aggravates GVD-mediated necroptosis in Alzheimer's disease. Acta Neuropathol Commun 2022; 10:128. [PMID: 36057624 PMCID: PMC9441100 DOI: 10.1186/s40478-022-01432-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/15/2022] [Indexed: 12/26/2022] Open
Abstract
It has become evident that Alzheimer's Disease (AD) is not only linked to its hallmark lesions-amyloid plaques and neurofibrillary tangles (NFTs)-but also to other co-occurring pathologies. This may lead to synergistic effects of the respective cellular and molecular players, resulting in neuronal death. One of these co-pathologies is the accumulation of phosphorylated transactive-response DNA binding protein 43 (pTDP-43) as neuronal cytoplasmic inclusions, currently considered to represent limbic-predominant age-related TDP-43 encephalopathy neuropathological changes (LATE-NC), in up to 70% of symptomatic AD cases. Granulovacuolar degeneration (GVD) is another AD co-pathology, which also contains TDP-43 and other AD-related proteins. Recently, we found that all proteins required for necroptosis execution, a previously defined programmed form of neuronal cell death, are present in GVD, such as the phosphorylated necroptosis executioner mixed-lineage kinase domain-like protein (pMLKL). Accordingly, this protein is a reliable marker for GVD lesions, similar to other known GVD proteins. Importantly, it is not yet known whether the presence of LATE-NC in symptomatic AD cases is associated with necroptosis pathway activation, presumably contributing to neuron loss by cell death execution. In this study, we investigated the impact of LATE-NC on the severity of necroptosis-associated GVD lesions, phosphorylated tau (pTau) pathology and neuronal density. First, we used 230 human post-mortem cases, including 82 controls without AD neuropathological changes (non-ADNC), 81 non-demented cases with ADNC, i.e.: pathologically-defined preclinical AD (p-preAD) and 67 demented cases with ADNC. We found that Braak NFT stage and LATE-NC stage were good predictors for GVD expansion and neuronal loss in the hippocampal CA1 region. Further, we compared the impact of TDP-43 accumulation on hippocampal expression of pMLKL-positive GVD, pTau as well as on neuronal density in a subset of nine non-ADNC controls, ten symptomatic AD cases with (ADTDP+) and eight without LATE-NC (ADTDP-). Here, we observed increased levels of pMLKL-positive, GVD-exhibiting neurons in ADTDP+ cases, compared to ADTDP- and controls, which was accompanied by augmented pTau pathology. Neuronal loss in the CA1 region was increased in ADTDP+ compared to ADTDP- cases. These data suggest that co-morbid LATE-NC in AD impacts not only pTau pathology but also GVD-mediated necroptosis pathway activation, which results in an accelerated neuronal demise. This further highlights the cumulative and synergistic effects of comorbid pathologies leading to neuronal loss in AD. Accordingly, protection against necroptotic neuronal death appears to be a promising therapeutic option for AD and LATE.
Collapse
Affiliation(s)
- Marta J Koper
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Sandra O Tomé
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Klara Gawor
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Annelies Belet
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Evelien Van Schoor
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
- Laboratory for Neurobiology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Jolien Schaeverbeke
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Laboratory for Translational Neuropsychiatry, Department of Neuroscience, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Laboratory for Translational Neuropsychiatry, Department of Neuroscience, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Department of Geriatric Psychiatry, UZ Leuven, Leuven, Belgium
| | - Estifanos Ghebremedhin
- Institute of Anatomy - Anatomy I, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
| | - Markus Otto
- Department of Neurology, Ulm University, Ulm, Germany
- Department of Neurology, University of Halle, Halle, Germany
| | - Christine A F von Arnim
- Department of Neurology, Ulm University, Ulm, Germany
- Department of Geriatrics, Göttingen University, Göttingen, Germany
| | - Sriram Balusu
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Matthew B Blaschko
- Department of Electronics, Center for Processing Speech and Images, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
- Department of Pathology, UZ Leuven, Leuven, Belgium.
| |
Collapse
|
36
|
Tazwar M, Evia AM, Tamhane AA, Ridwan AR, Leurgans SE, Bennett DA, Schneider JA, Arfanakis K. Limbic-predominant age-related TDP-43 encephalopathy neuropathological change (LATE-NC) is associated with lower R 2 relaxation rate: an ex-vivo MRI and pathology investigation. Neurobiol Aging 2022; 117:128-138. [PMID: 35728463 PMCID: PMC9667705 DOI: 10.1016/j.neurobiolaging.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 05/04/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022]
Abstract
Limbic predominant age-related transactive response DNA binding protein 43 (TDP-43) encephalopathy neuropathological change (LATE-NC) is common in persons older than 80 years of age and is associated with cognitive decline and increased likelihood of dementia. The MRI signature of LATE-NC has not been fully determined. In this study, the association of LATE-NC with the transverse relaxation rate, R2, was investigated in a large number of community-based older adults. Cerebral hemispheres from 738 participants of the Rush Memory and Aging Project, Religious Orders Study, and Minority Aging Research Study, were imaged ex-vivo with multi-echo spin-echo MRI and underwent detailed neuropathologic examination. Voxel-wise analysis revealed a novel spatial pattern of lower R2 for higher LATE-NC stage, controlling for other neuropathologies and demographics. This pattern was consistent with the distribution of LATE-NC in gray matter, and also involved white matter providing temporo-temporal, fronto-temporal, and temporo-basal ganglia connectivity. Furthermore, analysis at different LATE-NC stages showed that R2 imaging may capture the general progression of LATE-NC, but only when TDP-43 inclusions extend beyond the amygdala.
Collapse
Affiliation(s)
- Mahir Tazwar
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Arnold M Evia
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Ashish A Tamhane
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Abdur Raquib Ridwan
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Sue E Leurgans
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Diagnostic Radiology, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
37
|
Pottier C, Mateiu L, Baker MC, DeJesus-Hernandez M, Teixeira Vicente C, Finch NA, Tian S, van Blitterswijk M, Murray ME, Ren Y, Petrucelli L, Oskarsson B, Biernacka JM, Graff-Radford NR, Boeve BF, Petersen RC, Josephs KA, Asmann YW, Dickson DW, Rademakers R. Shared brain transcriptomic signature in TDP-43 type A FTLD patients with or without GRN mutations. Brain 2022; 145:2472-2485. [PMID: 34918030 PMCID: PMC9337811 DOI: 10.1093/brain/awab437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/24/2021] [Accepted: 10/24/2021] [Indexed: 11/28/2022] Open
Abstract
Frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP) is a complex heterogeneous neurodegenerative disorder for which mechanisms are poorly understood. To explore transcriptional changes underlying FTLD-TDP, we performed RNA-sequencing on 66 genetically unexplained FTLD-TDP patients, 24 FTLD-TDP patients with GRN mutations and 24 control participants. Using principal component analysis, hierarchical clustering, differential expression and coexpression network analyses, we showed that GRN mutation carriers and FTLD-TDP-A patients without a known mutation shared a common transcriptional signature that is independent of GRN loss-of-function. After combining both groups, differential expression as compared to the control group and coexpression analyses revealed alteration of processes related to immune response, synaptic transmission, RNA metabolism, angiogenesis and vesicle-mediated transport. Deconvolution of the data highlighted strong cellular alterations that were similar in FTLD-TDP-A and GRN mutation carriers with NSF as a potentially important player in both groups. We propose several potentially druggable pathways such as the GABAergic, GDNF and sphingolipid pathways. Our findings underline new disease mechanisms and strongly suggest that affected pathways in GRN mutation carriers extend beyond GRN and contribute to genetically unexplained forms of FTLD-TDP-A.
Collapse
Affiliation(s)
- Cyril Pottier
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Ligia Mateiu
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
| | - Matthew C Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Cristina Teixeira Vicente
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - NiCole A Finch
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Shulan Tian
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Joanna M Biernacka
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Yan W Asmann
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | | | - Rosa Rademakers
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
38
|
Wang SHJ, Guo Y, Ervin JF, Lusk JB, Luo S. Neuropathological associations of limbic-predominant age-related TDP-43 encephalopathy neuropathological change (LATE-NC) differ between the oldest-old and younger-old. Acta Neuropathol 2022; 144:45-57. [PMID: 35551470 PMCID: PMC9997084 DOI: 10.1007/s00401-022-02432-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/08/2023]
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathological change (LATE-NC) is most often seen in the oldest-old (≥ 90 years of age) but can also be present in the younger-old (< 90 years of age). In this study, we compared the neuropathological associations of LATE-NC and contribution of LATE-NC to cognitive impairment between the oldest-old and younger-old. We observed significant differences in the prevalence of LATE-NC and its association with other co-pathologies in these two age groups. LATE-NC was present in 30.9% (34/110) of the oldest-old but only 9.4% (19/203) of the younger-old. Participants of the oldest-old with LATE-NC were more likely to have hippocampal sclerosis (HS) (55.9% vs. 10.5%, p < 0.001) and moderate to severe arteriolosclerosis (82.4% vs. 50%, p = 0.007), but not intermediate to high Alzheimer's disease neuropathologic change (ADNC) (70.6% vs. 59.2%, p = 0.486) or Lewy body disease (LBD) (20.6% vs. 26.3%, p = 0.793). Participants of the younger-old with LATE-NC were more likely to have intermediate to high ADNC (94.7% vs. 55.4%, p < 0.001) and LBD (63.2% vs. 28.8%, p = 0.013) in addition to hippocampal sclerosis (42.1% vs. 6.5%, p < 0.001), and moderate to severe arteriolosclerosis (42.1% vs. 15.2%, p = 0.020). Of note, participants with LATE-NC and no to low ADNC were very rare in the younger-old (< 1%) but relatively common in the oldest-old (9.1%). Logistic regression modeling showed that in the oldest-old, both intermediate to high ADNC and LATE-NC were independently associated with higher odds of having dementia (OR: 5.09, 95% CI [1.99, 13.06], p < 0.001 for ADNC; OR: 3.28, 95% CI [1.25, 8.57], p = 0.015 for LATE-NC). In the younger-old, by contrast, intermediate to high ADNC and LBD were independently associated with higher odds of having dementia (OR: 4.43, 95% CI [2.27, 8.63], p < 0.001 for ADNC; OR: 2.55, 95% CI [1.21, 5.35], p < 0.014 for LBD), whereas LATE-NC did not show an independent association with dementia. Overall, LATE-NC is strongly associated with arteriolosclerosis and HS in both groups; however, in the younger-old, LATE-NC is associated with other neurodegenerative pathologies, such as ADNC and LBD; whereas in the oldest-old, LATE-NC can exist independent of significant ADNC.
Collapse
Affiliation(s)
- Shih-Hsiu J Wang
- Department of Pathology, Duke University Medical Center, 214MA Davison Bldg., 40 Duke Medicine Circle, Durham, NC, 27710, USA.
- Department of Neurology, Duke University Medical Center, Durham, USA.
| | - Yuanyuan Guo
- Department of Biostatics and Bioinformatics, Duke University Medical Center, Durham, USA
| | - John F Ervin
- Department of Neurology, Duke University Medical Center, Durham, USA
| | - Jay B Lusk
- Department of Pathology, Duke University Medical Center, 214MA Davison Bldg., 40 Duke Medicine Circle, Durham, NC, 27710, USA
- Department of Neurology, Duke University Medical Center, Durham, USA
| | - Sheng Luo
- Department of Biostatics and Bioinformatics, Duke University Medical Center, Durham, USA
| |
Collapse
|
39
|
Nelson PT, Brayne C, Flanagan ME, Abner EL, Agrawal S, Attems J, Castellani RJ, Corrada MM, Cykowski MD, Di J, Dickson DW, Dugger BN, Ervin JF, Fleming J, Graff-Radford J, Grinberg LT, Hokkanen SRK, Hunter S, Kapasi A, Kawas CH, Keage HAD, Keene CD, Kero M, Knopman DS, Kouri N, Kovacs GG, Labuzan SA, Larson EB, Latimer CS, Leite REP, Matchett BJ, Matthews FE, Merrick R, Montine TJ, Murray ME, Myllykangas L, Nag S, Nelson RS, Neltner JH, Nguyen AT, Petersen RC, Polvikoski T, Reichard RR, Rodriguez RD, Suemoto CK, Wang SHJ, Wharton SB, White L, Schneider JA. Frequency of LATE neuropathologic change across the spectrum of Alzheimer's disease neuropathology: combined data from 13 community-based or population-based autopsy cohorts. Acta Neuropathol 2022; 144:27-44. [PMID: 35697880 PMCID: PMC9552938 DOI: 10.1007/s00401-022-02444-1] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/04/2022] [Accepted: 05/22/2022] [Indexed: 02/02/2023]
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) and Alzheimer's disease neuropathologic change (ADNC) are each associated with substantial cognitive impairment in aging populations. However, the prevalence of LATE-NC across the full range of ADNC remains uncertain. To address this knowledge gap, neuropathologic, genetic, and clinical data were compiled from 13 high-quality community- and population-based longitudinal studies. Participants were recruited from United States (8 cohorts, including one focusing on Japanese-American men), United Kingdom (2 cohorts), Brazil, Austria, and Finland. The total number of participants included was 6196, and the average age of death was 88.1 years. Not all data were available on each individual and there were differences between the cohorts in study designs and the amount of missing data. Among those with known cognitive status before death (n = 5665), 43.0% were cognitively normal, 14.9% had MCI, and 42.4% had dementia-broadly consistent with epidemiologic data in this age group. Approximately 99% of participants (n = 6125) had available CERAD neuritic amyloid plaque score data. In this subsample, 39.4% had autopsy-confirmed LATE-NC of any stage. Among brains with "frequent" neuritic amyloid plaques, 54.9% had comorbid LATE-NC, whereas in brains with no detected neuritic amyloid plaques, 27.0% had LATE-NC. Data on LATE-NC stages were available for 3803 participants, of which 25% had LATE-NC stage > 1 (associated with cognitive impairment). In the subset of individuals with Thal Aβ phase = 0 (lacking detectable Aβ plaques), the brains with LATE-NC had relatively more severe primary age-related tauopathy (PART). A total of 3267 participants had available clinical data relevant to frontotemporal dementia (FTD), and none were given the clinical diagnosis of definite FTD nor the pathological diagnosis of frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP). In the 10 cohorts with detailed neurocognitive assessments proximal to death, cognition tended to be worse with LATE-NC across the full spectrum of ADNC severity. This study provided a credible estimate of the current prevalence of LATE-NC in advanced age. LATE-NC was seen in almost 40% of participants and often, but not always, coexisted with Alzheimer's disease neuropathology.
Collapse
Affiliation(s)
- Peter T Nelson
- University of Kentucky, Rm 311 Sanders-Brown Center on Aging, Lexington, KY, USA.
| | | | | | - Erin L Abner
- University of Kentucky, Rm 311 Sanders-Brown Center on Aging, Lexington, KY, USA
| | | | | | | | | | | | - Jing Di
- University of Kentucky, Rm 311 Sanders-Brown Center on Aging, Lexington, KY, USA
| | | | | | | | | | | | - Lea T Grinberg
- University of California, San Francisco, CA, USA
- University of Sao Paulo Medical School, Sao Paulo, Brazil
| | | | | | | | | | | | | | - Mia Kero
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | | | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Laboratory Medicine Program, University Health Network, Toronto, ON, Canada
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | | | - Eric B Larson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, 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 311 Sanders-Brown Center on Aging, Lexington, KY, USA
| | | | | | | | | | | | | | | | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Lon White
- Pacific Health Research and Education Institute, Honolulu, HI, USA
| | | |
Collapse
|
40
|
Carlos AF, Tosakulwong N, Weigand SD, Boeve BF, Knopman DS, Petersen RC, Nguyen A, Reichard RR, Murray ME, Dickson DW, Josephs KA. Frequency and distribution of TAR DNA-binding protein 43 (TDP-43) pathology increase linearly with age in a large cohort of older adults with and without dementia. Acta Neuropathol 2022; 144:159-160. [PMID: 35536384 PMCID: PMC9943023 DOI: 10.1007/s00401-022-02434-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 12/30/2022]
Affiliation(s)
| | | | - Stephen D. Weigand
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Aivi Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | | | | |
Collapse
|
41
|
Cykowski MD, Arumanayagam AS, Powell SZ, Rivera AL, Abner EL, Roman GC, Masdeu JC, Nelson PT. Patterns of amygdala region pathology in LATE-NC: subtypes that differ with regard to TDP-43 histopathology, genetic risk factors, and comorbid pathologies. Acta Neuropathol 2022; 143:531-545. [PMID: 35366087 PMCID: PMC9038848 DOI: 10.1007/s00401-022-02416-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 12/12/2022]
Abstract
Transactive response (TAR) DNA-binding protein 43 kDa (TDP-43) pathology is a hallmark of limbic-predominant age-related TDP-43 encephalopathy (LATE). The amygdala is affected early in the evolution of LATE neuropathologic change (LATE-NC), and heterogeneity of LATE-NC in amygdala has previously been observed. However, much remains to be learned about how LATE-NC originates and progresses in the brain. To address this, we assessed TDP-43 and other pathologies in the amygdala region of 184 autopsied subjects (median age = 85 years), blinded to clinical diagnoses, other neuropathologic diagnoses, and risk genotype information. As previously described, LATE-NC was associated with older age at death, cognitive impairment, and the TMEM106B risk allele. Pathologically, LATE-NC was associated with comorbid hippocampal sclerosis (HS), myelin loss, and vascular disease in white matter (WM). Unbiased hierarchical clustering of TDP-43 inclusion morphologies revealed discernable subtypes of LATE-NC with distinct clinical, genetic, and pathologic associations. The most common patterns were: Pattern 1, with lamina II TDP-43 + processes and preinclusion pathology in cortices of the amygdala region, and frequent LATE-NC Stage 3 with HS; Pattern 2, previously described as type-β, with neurofibrillary tangle-like TDP-43 neuronal cytoplasmic inclusions (NCIs), high Alzheimer's disease neuropathologic change (ADNC), frequent APOE ε4, and usually LATE-NC Stage 2; Pattern 3, with round NCIs and thick neurites in amygdala, younger age at death, and often comorbid Lewy body disease; and Pattern 4 (the most common pattern), with tortuous TDP-43 processes in subpial and WM regions, low ADNC, rare HS, and lower dementia probability. TDP-43 pathology with features of patterns 1 and 2 were often comorbid in the same brains. Early and mild TDP-43 pathology was often best described to be localized in the "amygdala region" rather than the amygdala proper. There were also important shared attributes across patterns. For example, all four patterns were associated with the TMEM106B risk allele. Each pattern also demonstrated the potential to progress to higher LATE-NC stages with confluent anatomical and pathological patterns, and to contribute to dementia. Although LATE-NC showed distinct patterns of initiation in amygdala region, there was also apparent shared genetic risk and convergent pathways of clinico-pathological evolution.
Collapse
Affiliation(s)
- Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, 77030, USA.
- Methodist Neurological Institute Department of Neurology, Houston Methodist Hospital, Weil Cornell Medicine, Houston, TX, 77030, USA.
| | | | - Suzanne Z Powell
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Andreana L Rivera
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Erin L Abner
- Sanders-Brown Center On Aging, University of Kentucky, University of Kentucky, Lexington, KY, 40536, USA
- Department of Epidemiology, University of Kentucky, Lexington, KY, 40536, USA
| | - Gustavo C Roman
- Methodist Neurological Institute Department of Neurology, Houston Methodist Hospital, Weil Cornell Medicine, Houston, TX, 77030, USA
- Nantz National Alzheimer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Joseph C Masdeu
- Methodist Neurological Institute Department of Neurology, Houston Methodist Hospital, Weil Cornell Medicine, Houston, TX, 77030, USA
- Nantz National Alzheimer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Peter T Nelson
- Sanders-Brown Center On Aging, University of Kentucky, University of Kentucky, Lexington, KY, 40536, USA
- Department of Pathology, University of Kentucky, Lexington, KY, 40536, USA
| |
Collapse
|
42
|
Stanley NE, Robinson LJ, Mao Q. A Novel, Heterozygous BSCL2 Variant in Association With Early-Onset Epileptic Encephalopathy. J Neuropathol Exp Neurol 2022; 81:377-380. [PMID: 35290466 DOI: 10.1093/jnen/nlac013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- Nicole E Stanley
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | | | - Qinwen Mao
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
43
|
Gauthreaux KM, Teylan MA, Katsumata Y, Mock C, Culhane JE, Chen YC, Chan KCG, Fardo DW, Dugan AJ, Cykowski MD, Jicha GA, Kukull WA, Nelson PT. Limbic-Predominant Age-Related TDP-43 Encephalopathy: Medical and Pathologic Factors Associated With Comorbid Hippocampal Sclerosis. Neurology 2022; 98:e1422-e1433. [PMID: 35121671 PMCID: PMC8992604 DOI: 10.1212/wnl.0000000000200001] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 01/03/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Limbic-predominant age-related Tar DNA binding protein 43 (TDP-43) encephalopathy neuropathologic change (LATE-NC) is present in ≈25% of older persons' brains and is strongly associated with cognitive impairment. Hippocampal sclerosis (HS) pathology is often comorbid with LATE-NC, but the clinical and pathologic correlates of HS in LATE-NC are not well understood. METHODS This retrospective autopsy cohort study used data derived from the National Alzheimer's Coordinating Center Neuropathology Data Set, which included neurologic status, medical histories, and neuropathologic results. All autopsies were performed in 2014 or later. Among participants with LATE-NC, those who also had HS pathology were compared with those without HS with regard to candidate risk factors or common underlying diseases. Statistical significance was set at nominal p < 0.05 in this exploratory study. RESULTS A total of 408 participants were included (n = 221 were LATE-NC+/HS-, n = 145 were LATE-NC+/HS+, and n = 42 were LATE-NC-/HS+). Most of the included LATE-NC+ participants were severely impaired cognitively (83.3% with dementia). Compared to HS- participants, LATE-NC+ participants with HS trended toward having worse cognitive status and scored lower on the Personal Care and Orientation domains (both p = 0.03). Among LATE-NC+ participants with Braak neurofibrillary tangle (NFT) stages 0 to IV (n = 88), HS+ participants were more impaired in the Memory and Orientation domains (both p = 0.02). There were no differences (HS+ compared with HS-) in the proportion with clinical histories of seizures, stroke, cardiac bypass procedures, diabetes, or hypertension. The HS+ group lacking TDP-43 proteinopathy (n = 42) was relatively likely to have had strokes (p = 0.03). When LATE-NC+ participants with or without HS were compared, there were no differences in Alzheimer disease neuropathologies (Thal β-amyloid phases or Braak NFT stages) or Lewy body pathologies. However, the HS+ group was less likely to have amygdala-restricted TDP-43 proteinopathy (LATE-NC stage 1) and more likely to have neocortical TDP-43 proteinopathy (LATE-NC stage 3) (p < 0.001). LATE-NC+ brains with HS also tended to have more severe circle of Willis atherosclerosis and arteriolosclerosis pathologies. DISCUSSION In this cohort skewed toward participants with severe dementia, LATE-NC+ HS pathology was not associated with seizures or with Alzheimer-type pathologies. Rather, the presence of comorbid HS pathology was associated with more widespread TDP-43 proteinopathy and with more severe non-β-amyloid vessel wall pathologies.
Collapse
Affiliation(s)
- Kathryn M Gauthreaux
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Merilee A Teylan
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Yuriko Katsumata
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Charles Mock
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Jessica E Culhane
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Yen-Chi Chen
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Kwun C G Chan
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - David W Fardo
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Adam J Dugan
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Matthew D Cykowski
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Gregory A Jicha
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Walter A Kukull
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington
| | - Peter T Nelson
- From the National Alzheimer's Coordinating Center (K.M.G., M.A.T., C.M., J.E.C., K.C.G.C., W.A.K.), Department of Epidemiology, and Department of Statistics (Y.-C.C.) University of Washington, Seattle; Houston Methodist Hospital (M.D.C.), TX; and Sanders-Brown Center on Aging (Y.K., D.W.F., G.A.J., P.T.N.), Department of Biostatistics (Y.K., D.W.F., A.J.D.), Department of Neurology (G.A.J.), and Department of Pathology (P.T.N.), Division of Neuropathology, University of Kentucky, Lexington.
| |
Collapse
|
44
|
Buciuc M, Martin PR, Tosakulwong N, Murray ME, Petrucelli L, Senjem ML, Spychalla AJ, Knopman DS, Boeve BF, Petersen RC, Parisi JE, Reichard RR, Dickson DW, Jack CR, Whitwell JL, Josephs KA. TDP-43-associated atrophy in brains with and without frontotemporal lobar degeneration. Neuroimage Clin 2022; 34:102954. [PMID: 35168140 PMCID: PMC8850800 DOI: 10.1016/j.nicl.2022.102954] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 11/29/2022]
Abstract
Transactive response DNA-binding protein of ∼43 kDa (TDP-43), a primary pathologic substrate in tau-negative frontotemporal lobar degeneration (FTLD), is also often found in the brains of elderly individuals without FTLD and is a key player in the process of neurodegeneration in brains with and without FTLD. It is unknown how rates and trajectories of TDP-43-associated brain atrophy compare between these two groups. Additionally, non-FTLD TDP-43 inclusions are not homogeneous and can be divided into two morphologic types: type-α and neurofibrillary tangle-associated type-β. Therefore, we explored whether neurodegeneration also varies due to the morphologic type. In this longitudinal retrospective study of 293 patients with 843 MRI scans spanning over ∼10 years, we used a Bayesian hierarchical linear model to quantify similarities and differences between the non-FTLD TDP-43 (type-α/type-β) and FTLD-TDP (n = 68) in both regional volume at various timepoints before death and annualized rate of atrophy. Since Alzheimer's disease (AD) is a frequent co-pathology in non-FTLD TDP-43, we further divided types α/β based on presence/absence of intermediate-high likelihood AD: AD-TDP type-β (n = 90), AD-TDP type-α (n = 104), and Pure-TDP (n = 31, all type-α). FTLD-TDP was associated with faster atrophy rates in the inferior temporal lobe and temporal pole compared to all non-FTLD TDP-43 groups. The atrophy rate in the frontal lobe was modulated by age with younger FTLD-TDP having the fastest rates. Older FTLD-TDP showed a limbic predominant pattern of neurodegeneration. AD-TDP type-α showed faster rates of hippocampal atrophy and smaller volumes of amygdala, temporal pole, and inferior temporal lobe compared to AD-TDP type-β. Pure-TDP was associated with slowest rates and less atrophy in all brain regions. The results suggest that there are differences and similarities in longitudinal brain volume loss between FTLD-TDP and non-FTLD TDP-43. Within FTLD-TDP age plays a role in which brain regions are the most affected. Additionally, brain atrophy regional rates also vary by non-FTLD TDP-43 type.
Collapse
Affiliation(s)
- Marina Buciuc
- Department of Neurology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Peter R Martin
- Department of Quantitative Health Sciences, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Nirubol Tosakulwong
- Department of Quantitative Health Sciences, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA.
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA.
| | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Anthony J Spychalla
- Department of Radiology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - David S Knopman
- Department of Neurology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Ronald C Petersen
- Department of Neurology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Joseph E Parisi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - R Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA.
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Jennifer L Whitwell
- Department of Radiology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| | - Keith A Josephs
- Department of Neurology, Mayo Clinic, 200 1(st) Street NW, Rochester, MN 55905, USA.
| |
Collapse
|
45
|
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: 30] [Impact Index Per Article: 10.0] [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
|
46
|
Meneses A, Koga S, O’Leary J, Dickson DW, Bu G, Zhao N. TDP-43 Pathology in Alzheimer's Disease. Mol Neurodegener 2021; 16:84. [PMID: 34930382 PMCID: PMC8691026 DOI: 10.1186/s13024-021-00503-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/21/2021] [Indexed: 12/05/2022] Open
Abstract
Transactive response DNA binding protein of 43 kDa (TDP-43) is an intranuclear protein encoded by the TARDBP gene that is involved in RNA splicing, trafficking, stabilization, and thus, the regulation of gene expression. Cytoplasmic inclusion bodies containing phosphorylated and truncated forms of TDP-43 are hallmarks of amyotrophic lateral sclerosis (ALS) and a subset of frontotemporal lobar degeneration (FTLD). Additionally, TDP-43 inclusions have been found in up to 57% of Alzheimer's disease (AD) cases, most often in a limbic distribution, with or without hippocampal sclerosis. In some cases, TDP-43 deposits are also found in neurons with neurofibrillary tangles. AD patients with TDP-43 pathology have increased severity of cognitive impairment compared to those without TDP-43 pathology. Furthermore, the most common genetic risk factor for AD, apolipoprotein E4 (APOE4), is associated with increased frequency of TDP-43 pathology. These findings provide strong evidence that TDP-43 pathology is an integral part of multiple neurodegenerative conditions, including AD. Here, we review the biology and pathobiology of TDP-43 with a focus on its role in AD. We emphasize the need for studies on the mechanisms that lead to TDP-43 pathology, especially in the setting of age-related disorders such as AD.
Collapse
Affiliation(s)
- Axel Meneses
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Justin O’Leary
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| |
Collapse
|
47
|
Dugan AJ, Nelson PT, Katsumata Y, Shade LMP, Boehme KL, Teylan MA, Cykowski MD, Mukherjee S, Kauwe JSK, Hohman TJ, Schneider JA, Fardo DW. Analysis of genes (TMEM106B, GRN, ABCC9, KCNMB2, and APOE) implicated in risk for LATE-NC and hippocampal sclerosis provides pathogenetic insights: a retrospective genetic association study. Acta Neuropathol Commun 2021; 9:152. [PMID: 34526147 PMCID: PMC8442328 DOI: 10.1186/s40478-021-01250-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022] Open
Abstract
Limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC) is the most prevalent subtype of TDP-43 proteinopathy, affecting up to 1/3rd of aged persons. LATE-NC often co-occurs with hippocampal sclerosis (HS) pathology. It is currently unknown why some individuals with LATE-NC develop HS while others do not, but genetics may play a role. Previous studies found associations between LATE-NC phenotypes and specific genes: TMEM106B, GRN, ABCC9, KCNMB2, and APOE. Data from research participants with genomic and autopsy measures from the National Alzheimer’s Coordinating Center (NACC; n = 631 subjects included) and the Religious Orders Study and Memory and the Rush Aging Project (ROSMAP; n = 780 included) were analyzed in the current study. Our goals were to reevaluate disease-associated genetic variants using newly collected data and to query whether the specific genotype/phenotype associations could provide new insights into disease-driving pathways. Research subjects included in prior LATE/HS genome-wide association studies (GWAS) were excluded. Single nucleotide variants (SNVs) within 10 kb of TMEM106B, GRN, ABCC9, KCNMB2, and APOE were tested for association with HS and LATE-NC, and separately for Alzheimer’s pathologies, i.e. amyloid plaques and neurofibrillary tangles. Significantly associated SNVs were identified. When results were meta-analyzed, TMEM106B, GRN, and APOE had significant gene-based associations with both LATE and HS, whereas ABCC9 had significant associations with HS only. In a sensitivity analysis limited to LATE-NC + cases, ABCC9 variants were again associated with HS. By contrast, the associations of TMEM106B, GRN, and APOE with HS were attenuated when adjusting for TDP-43 proteinopathy, indicating that these genes may be associated primarily with TDP-43 proteinopathy. None of these genes except APOE appeared to be associated with Alzheimer’s-type pathology. In summary, using data not included in prior studies of LATE or HS genomics, we replicated several previously reported gene-based associations and found novel evidence that specific risk alleles can differentially affect LATE-NC and HS.
Collapse
|
48
|
Wisse LEM, Ravikumar S, Ittyerah R, Lim S, Lane J, Bedard ML, Xie L, Das SR, Schuck T, Grossman M, Lee EB, Tisdall MD, Prabhakaran K, Detre JA, Mizsei G, Trojanowski JQ, Artacho-Pérula E, de Iñiguez de Onzono Martin MM, M Arroyo-Jiménez M, Muñoz Lopez M, Molina Romero FJ, P Marcos Rabal M, Cebada Sánchez S, Delgado González JC, de la Rosa Prieto C, Córcoles Parada M, Wolk DA, Irwin DJ, Insausti R, Yushkevich PA. Downstream effects of polypathology on neurodegeneration of medial temporal lobe subregions. Acta Neuropathol Commun 2021; 9:128. [PMID: 34289895 PMCID: PMC8293481 DOI: 10.1186/s40478-021-01225-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/06/2021] [Indexed: 12/14/2022] Open
Abstract
The medial temporal lobe (MTL) is a nidus for neurodegenerative pathologies and therefore an important region in which to study polypathology. We investigated associations between neurodegenerative pathologies and the thickness of different MTL subregions measured using high-resolution post-mortem MRI. Tau, TAR DNA-binding protein 43 (TDP-43), amyloid-β and α-synuclein pathology were rated on a scale of 0 (absent)-3 (severe) in the hippocampus and entorhinal cortex (ERC) of 58 individuals with and without neurodegenerative diseases (median age 75.0 years, 60.3% male). Thickness measurements in ERC, Brodmann Area (BA) 35 and 36, parahippocampal cortex, subiculum, cornu ammonis (CA)1 and the stratum radiatum lacunosum moleculare (SRLM) were derived from 0.2 × 0.2 × 0.2 mm3 post-mortem MRI scans of excised MTL specimens from the contralateral hemisphere using a semi-automated approach. Spearman's rank correlations were performed between neurodegenerative pathologies and thickness, correcting for age, sex and hemisphere, including all four proteinopathies in the model. We found significant associations of (1) TDP-43 with thickness in all subregions (r = - 0.27 to r = - 0.46), and (2) tau with BA35 (r = - 0.31) and SRLM thickness (r = - 0.33). In amyloid-β and TDP-43 negative cases, we found strong significant associations of tau with ERC (r = - 0.40), BA35 (r = - 0.55), subiculum (r = - 0.42) and CA1 thickness (r = - 0.47). This unique dataset shows widespread MTL atrophy in relation to TDP-43 pathology and atrophy in regions affected early in Braak stageing and tau pathology. Moreover, the strong association of tau with thickness in early Braak regions in the absence of amyloid-β suggests a role of Primary Age-Related Tauopathy in neurodegeneration.
Collapse
Affiliation(s)
- L E M Wisse
- Department of Diagnostic Radiology, Lund University, Klinikgatan 13b, Lund, Sweden.
- Department of Radiology, University of Pennsylvania, Philadelphia, USA.
| | - S Ravikumar
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - R Ittyerah
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - S Lim
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - J Lane
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - M L Bedard
- Department of Pharmacology, University of North Carolina At Chapel Hill, Chapel Hill, USA
| | - L Xie
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - S R Das
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - T Schuck
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, USA
| | - M Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - E B Lee
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, USA
| | - M D Tisdall
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - K Prabhakaran
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - J A Detre
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - G Mizsei
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - J Q Trojanowski
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, USA
| | - E Artacho-Pérula
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | | | - M M Arroyo-Jiménez
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - M Muñoz Lopez
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - F J Molina Romero
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - M P Marcos Rabal
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - S Cebada Sánchez
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - J C Delgado González
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - C de la Rosa Prieto
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - M Córcoles Parada
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - D A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - D J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, USA
| | - R Insausti
- Human Neuroanatomy Laboratory, Neuromax CSIC Associated Unit, University of Castilla La Mancha, Albacete, Spain
| | - P A Yushkevich
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
49
|
Nelson PT. What to do with unusual TDP-43 proteinopathy cases? Neuropathol Appl Neurobiol 2021; 48:e12745. [PMID: 34164840 DOI: 10.1111/nan.12745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/04/2021] [Accepted: 06/19/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Department of Pathology, Division of Neuropathology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
50
|
Woodworth DC, Scambray KA, Corrada MM, Kawas CH, Sajjadi SA. Neuroimaging in the Oldest-Old: A Review of the Literature. J Alzheimers Dis 2021; 82:129-147. [PMID: 33998539 DOI: 10.3233/jad-201578] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The oldest-old, those 85 years and older, are the fastest growing segment of the population and present with the highest prevalence of dementia. Given the importance of neuroimaging measures to understand aging and dementia, the objective of this study was to review neuroimaging studies performed in oldest-old participants. We used PubMed, Google Scholar, and Web of Science search engines to identify in vivo CT, MRI, and PET neuroimaging studies either performed in the oldest-old or that addressed the oldest-old as a distinct group in analyses. We identified 60 studies and summarized the main group characteristics and findings. Generally, oldest-old participants presented with greater atrophy compared to younger old participants, with most studies reporting a relatively stable constant decline in brain volumes over time. Oldest-old participants with greater global atrophy and atrophy in key brain structures such as the medial temporal lobe were more likely to have dementia or cognitive impairment. The oldest-old presented with a high burden of white matter lesions, which were associated with various lifestyle factors and some cognitive measures. Amyloid burden as assessed by PET, while high in the oldest-old compared to younger age groups, was still predictive of transition from normal to impaired cognition, especially when other adverse neuroimaging measures (atrophy and white matter lesions) were also present. While this review highlights past neuroimaging research in the oldest-old, it also highlights the dearth of studies in this important population. It is imperative to perform more neuroimaging studies in the oldest-old to better understand aging and dementia.
Collapse
Affiliation(s)
- Davis C Woodworth
- Department of Neurology, University of California, Irvine, CA, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Kiana A Scambray
- Department of Neurology, University of California, Irvine, CA, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - María M Corrada
- Department of Neurology, University of California, Irvine, CA, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA.,Department of Epidemiology, University of California, Irvine, CA, USA
| | - Claudia H Kawas
- Department of Neurology, University of California, Irvine, CA, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA.,Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - S Ahmad Sajjadi
- Department of Neurology, University of California, Irvine, CA, USA.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| |
Collapse
|