1
|
Hojjati SH, Chiang GC, Butler TA, Chen K, Khalafi M, Yazdi BG, Foldi N, Nayak S, de Leon M, Li Y, Stern Y, Luchsinger JA, Razlighi QR. Heterogeneous tau deposition patterns in the preclinical stage link to domain-specific cognitive deficits. Alzheimers Dement 2025; 21:e70153. [PMID: 40355988 PMCID: PMC12069010 DOI: 10.1002/alz.70153] [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: 12/27/2024] [Revised: 02/24/2025] [Accepted: 03/11/2025] [Indexed: 05/15/2025]
Abstract
INTRODUCTION The spatial heterogeneity of tau deposition is closely linked to clinical variants of Alzheimer's disease (AD). Detecting these patterns in the preclinical stage is challenging, but second-generation tau tracers provide a unique opportunity to do so. METHODS We used independent component analysis (ICA) and tau positron emission tomography (PET) imaging with the 18F-MK6240 tracer in 590 cognitively healthy adults (mean age 66.58 ± 5.13 years, 340 females) to identify tau patterns in the preclinical stage. RESULTS Using all individuals, seven distinct patterns emerged, with medial temporal lobe (MTL) involvement associated with age, Aβ burden, apolipoprotein E (APOE) genotype, and plasma total tau. Bilateral amygdala-hippocampus tau deposition was associated negatively with memory (t = -2.64, p < 0.01), while broader neocortical patterns, especially asymmetric ones, were linked to deficits in language (t < -3.13, p < 0.002) and reasoning (t < -2.63, p < 0.01). DISCUSSION These findings advance our understanding of preclinical tau heterogeneity, offering new insights for early AD intervention. HIGHLIGHTS Seven tau deposition patterns were identified in preclinical stages of AD, including medial temporal lobe and asymmetric neocortical patterns. Medial temporal lobe patterns were strongly linked to age, APOE genotype, Aβ burden, and plasma total tau levels. Neocortical patterns, especially asymmetric ones, were linked to domain-specific cognitive deficits, notably in language and reasoning. This research highlights the potential of using tau deposition patterns for early detection and tailoring interventions in preclinical AD.
Collapse
Affiliation(s)
- Seyed Hani Hojjati
- Department of RadiologyBrain Health Imaging Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Gloria C. Chiang
- Department of RadiologyBrain Health Imaging Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Tracy A. Butler
- Department of RadiologyBrain Health Imaging Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Kewei Chen
- College of Health SolutionsArizona State UniversityPhoenixArizonaUSA
- School of Mathematics and StatisticsArizona State UniversityPhoenixArizonaUSA
- Department of NeurologyUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Mohammad Khalafi
- Department of RadiologyBrain Health Imaging Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Bardiya Ghaderi Yazdi
- Department of RadiologyBrain Health Imaging Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Nancy Foldi
- Department of RadiologyBrain Health Imaging Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Siddharth Nayak
- Department of NeurologyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Mony de Leon
- Department of RadiologyBrain Health Imaging Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Yi Li
- Department of RadiologyBrain Health Imaging Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Yaakov Stern
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainVagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyVagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Gertrude H. Sergievsky CenterVagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of PsychiatryVagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - José A. Luchsinger
- Department of MedicineColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Department of EpidemiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Qolamreza R. Razlighi
- Department of RadiologyBrain Health Imaging Institute, Weill Cornell MedicineNew YorkNew YorkUSA
| |
Collapse
|
2
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated Appropriate Use Criteria for Amyloid and Tau PET: A Report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. J Nucl Med 2025:jnumed.124.268756. [PMID: 39778970 DOI: 10.2967/jnumed.124.268756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 01/11/2025] Open
Abstract
The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. Methods: The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. Results: For amyloid PET, 7 scenarios were rated as appropriate, 2 as uncertain, and 8 as rarely appropriate. For tau PET, 5 scenarios were rated as appropriate, 6 as uncertain, and 6 as rarely appropriate. Conclusion: AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Gil D Rabinovici
- Department of Neurology and Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California;
| | - David S Knopman
- Mayo Clinic Neurology and Neurosurgery, Rochester, Minnesota
| | - Javier Arbizu
- Department of Nuclear Medicine, University of Navarra Clinic, Pamplona, Spain
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri; Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Kevin J Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Peter Herscovitch
- Positron Emission Tomography Department, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Phillip H Kuo
- Medical Imaging, Medicine, and Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Jennifer H Lingler
- Department of Health and Community Systems, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| | | | - Julie C Price
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Stephen P Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine, Brown University, Providence, Rhode Island
- Butler Hospital Memory and Aging Program, Providence, Rhode Island
| | | | | | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
- Molecular Neuroimaging, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
- Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
3
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated appropriate use criteria for amyloid and tau PET: A report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. Alzheimers Dement 2025; 21:e14338. [PMID: 39776249 PMCID: PMC11772739 DOI: 10.1002/alz.14338] [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: 07/19/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. METHODS The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. RESULTS For amyloid PET, seven scenarios were rated as appropriate, two as uncertain, and eight as rarely appropriate. For tau PET, five scenarios were rated as appropriate, six as uncertain, and six as rarely appropriate. DISCUSSION AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease. HIGHLIGHTS A multidisciplinary workgroup convened by the Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging updated the appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. The goal of these updated AUC is to assist clinicians in identifying clinical scenarios in which amyloid or tau PET may be useful for guiding the diagnosis and management of patients who have, or are at risk for, cognitive decline These updated AUC are intended for dementia specialists who spend a significant proportion of their clinical effort caring for patients with cognitive complaints, as well as serve as a general reference for a broader audience interested in implementation of amyloid and tau PET in clinical practice.
Collapse
Affiliation(s)
- Gil D. Rabinovici
- Department of Neurology and Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Javier Arbizu
- Department of Nuclear MedicineUniversity of Navarra ClinicPamplonaSpain
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
- Knight Alzheimer's Disease Research CenterWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Kevin J. Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Oskar Hansson
- Department of Clinical Sciences MalmöClinical Memory Research UnitFaculty of MedicineLund UniversityLundSweden
- Memory Clinic, Skåne University HospitalSkånes universitetssjukhusMalmöSweden
| | - Peter Herscovitch
- Positron Emission Tomography DepartmentNational Institutes of Health Clinical CenterBethesdaMarylandUSA
| | - Phillip H. Kuo
- Medical Imaging, Medicine, and Biomedical EngineeringUniversity of ArizonaTucsonArizonaUSA
| | - Jennifer H. Lingler
- Department of Health and Community SystemsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Satoshi Minoshima
- Department of Radiology and Imaging SciencesUniversity of UtahSalt Lake CityUtahUSA
| | | | - Julie C. Price
- Department of RadiologyMassachusetts General Hospital, BostonCharlestownMassachusettsUSA
| | - Stephen P. Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine at Brown UniversityProvidenceRhode IslandUSA
- Butler Hospital Memory and Aging ProgramProvidenceRhode IslandUSA
| | | | - Maria C. Carrillo
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Keith A. Johnson
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Molecular Neuroimaging, Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Departments of Neurology and RadiologyMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
4
|
Lee J, Burkett BJ, Min HK, Senjem ML, Dicks E, Corriveau-Lecavalier N, Mester CT, Wiste HJ, Lundt ES, Murray ME, Nguyen AT, Reichard RR, Botha H, Graff-Radford J, Barnard LR, Gunter JL, Schwarz CG, Kantarci K, Knopman DS, Boeve BF, Lowe VJ, Petersen RC, Jack CR, Jones DT. Synthesizing images of tau pathology from cross-modal neuroimaging using deep learning. Brain 2024; 147:980-995. [PMID: 37804318 PMCID: PMC10907092 DOI: 10.1093/brain/awad346] [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: 02/20/2023] [Revised: 08/30/2023] [Accepted: 09/24/2023] [Indexed: 10/09/2023] Open
Abstract
Given the prevalence of dementia and the development of pathology-specific disease-modifying therapies, high-value biomarker strategies to inform medical decision-making are critical. In vivo tau-PET is an ideal target as a biomarker for Alzheimer's disease diagnosis and treatment outcome measure. However, tau-PET is not currently widely accessible to patients compared to other neuroimaging methods. In this study, we present a convolutional neural network (CNN) model that imputes tau-PET images from more widely available cross-modality imaging inputs. Participants (n = 1192) with brain T1-weighted MRI (T1w), fluorodeoxyglucose (FDG)-PET, amyloid-PET and tau-PET were included. We found that a CNN model can impute tau-PET images with high accuracy, the highest being for the FDG-based model followed by amyloid-PET and T1w. In testing implications of artificial intelligence-imputed tau-PET, only the FDG-based model showed a significant improvement of performance in classifying tau positivity and diagnostic groups compared to the original input data, suggesting that application of the model could enhance the utility of the metabolic images. The interpretability experiment revealed that the FDG- and T1w-based models utilized the non-local input from physically remote regions of interest to estimate the tau-PET, but this was not the case for the Pittsburgh compound B-based model. This implies that the model can learn the distinct biological relationship between FDG-PET, T1w and tau-PET from the relationship between amyloid-PET and tau-PET. Our study suggests that extending neuroimaging's use with artificial intelligence to predict protein specific pathologies has great potential to inform emerging care models.
Collapse
Affiliation(s)
- Jeyeon Lee
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biomedical Engineering, Hanyang University, Seoul 04763, Korea
| | - Brian J Burkett
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Hoon-Ki Min
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ellen Dicks
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Carly T Mester
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Heather J Wiste
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Emily S Lundt
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ross R Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - David T Jones
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Whitwell JL. Atypical clinical variants of Alzheimer's disease: are they really atypical? Front Neurosci 2024; 18:1352822. [PMID: 38482142 PMCID: PMC10933030 DOI: 10.3389/fnins.2024.1352822] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/15/2024] [Indexed: 02/12/2025] Open
Abstract
Alzheimer's disease (AD) is a neuropathological disorder defined by the deposition of the proteins, tau and β-amyloid. Alzheimer's disease is commonly thought of as a disease of the elderly that is associated with episodic memory loss. However, the very first patient described with AD was in her 50's with impairments in multiple cognitive domains. It is now clear that AD can present with multiple different non-amnestic clinical variants which have been labeled as atypical variants of AD. Instead of these variants of AD being considered "atypical," I propose that they provide an excellent disease model of AD and reflect the true clinical heterogeneity of AD. The atypical variants of AD usually have a relatively young age at onset, and they show striking cortical tau deposition on molecular PET imaging which relates strongly with patterns of neurodegeneration and clinical outcomes. In contrast, elderly patients with AD show less tau deposition on PET, and neuroimaging and clinical outcomes are confounded by other age-related pathologies, including TDP-43 and vascular pathology. There is also considerable clinical and anatomical heterogeneity across atypical and young-onset amnestic variants of AD which reflects the fact that AD is a disease that causes impairments in multiple cognitive domains. Future studies should focus on careful characterization of cognitive impairment in AD and consider the full clinical spectrum of AD, including atypical AD, in the design of research studies investigating disease mechanisms in AD and clinical treatment trials, particularly with therapeutics targeting tau.
Collapse
|
6
|
Kawles A, Keszycki R, Minogue G, Zouridakis A, Ayala I, Gill N, Macomber A, Lubbat V, Coventry C, Rogalski E, Weintraub S, Mao Q, Flanagan ME, Zhang H, Castellani R, Bigio EH, Mesulam MM, Geula C, Gefen T. Phenotypically concordant distribution of pick bodies in aphasic versus behavioral dementias. Acta Neuropathol Commun 2024; 12:31. [PMID: 38389095 PMCID: PMC10885488 DOI: 10.1186/s40478-024-01738-7] [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: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
Pick's disease (PiD) is a subtype of the tauopathy form of frontotemporal lobar degeneration (FTLD-tau) characterized by intraneuronal 3R-tau inclusions. PiD can underly various dementia syndromes, including primary progressive aphasia (PPA), characterized by an isolated and progressive impairment of language and left-predominant atrophy, and behavioral variant frontotemporal dementia (bvFTD), characterized by progressive dysfunction in personality and bilateral frontotemporal atrophy. In this study, we investigated the neocortical and hippocampal distributions of Pick bodies in bvFTD and PPA to establish clinicopathologic concordance between PiD and the salience of the aphasic versus behavioral phenotype. Eighteen right-handed cases with PiD as the primary pathologic diagnosis were identified from the Northwestern University Alzheimer's Disease Research Center brain bank (bvFTD, N = 9; PPA, N = 9). Paraffin-embedded sections were stained immunohistochemically with AT8 to visualize Pick bodies, and unbiased stereological analysis was performed in up to six regions bilaterally [middle frontal gyrus (MFG), superior temporal gyrus (STG), inferior parietal lobule (IPL), anterior temporal lobe (ATL), dentate gyrus (DG) and CA1 of the hippocampus], and unilateral occipital cortex (OCC). In bvFTD, peak neocortical densities of Pick bodies were in the MFG, while the ATL was the most affected in PPA. Both the IPL and STG had greater leftward pathology in PPA, with the latter reaching significance (p < 0.01). In bvFTD, Pick body densities were significantly right-asymmetric in the STG (p < 0.05). Hippocampal burden was not clinicopathologically concordant, as both bvFTD and PPA cases demonstrated significant hippocampal pathology compared to neocortical densities (p < 0.0001). Inclusion-to-neuron analyses in a subset of PPA cases confirmed that neurons in the DG are disproportionately burdened with inclusions compared to neocortical areas. Overall, stereological quantitation suggests that the distribution of neocortical Pick body pathology is concordant with salient clinical features unique to PPA vs. bvFTD while raising intriguing questions about the selective vulnerability of the hippocampus to 3R-tauopathies.
Collapse
Affiliation(s)
- Allegra Kawles
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rachel Keszycki
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Grace Minogue
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Antonia Zouridakis
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ivan Ayala
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nathan Gill
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Alyssa Macomber
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Vivienne Lubbat
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emily Rogalski
- Department of Neurology, University of Chicago School of Medicine, Chicago, IL, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Qinwen Mao
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hui Zhang
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rudolph Castellani
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eileen H Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - M-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
7
|
Kim J, Kim J, Park YH, Yoo H, Kim JP, Jang H, Park H, Seo SW. Distinct spatiotemporal patterns of cortical thinning in Alzheimer's disease-type cognitive impairment and subcortical vascular cognitive impairment. Commun Biol 2024; 7:198. [PMID: 38368479 PMCID: PMC10874406 DOI: 10.1038/s42003-024-05787-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 01/03/2024] [Indexed: 02/19/2024] Open
Abstract
Previous studies on Alzheimer's disease-type cognitive impairment (ADCI) and subcortical vascular cognitive impairment (SVCI) has rarely explored spatiotemporal heterogeneity. This study aims to identify distinct spatiotemporal cortical atrophy patterns in ADCI and SVCI. 1,338 participants (713 ADCI, 208 SVCI, and 417 cognitively unimpaired elders) underwent brain magnetic resonance imaging (MRI), amyloid positron emission tomography, and neuropsychological tests. Using MRI, this study measures cortical thickness in five brain regions (medial temporal, inferior temporal, posterior medial parietal, lateral parietal, and frontal areas) and utilizes the Subtype and Stage Inference (SuStaIn) model to predict the most probable subtype and stage for each participant. SuStaIn identifies two distinct cortical thinning patterns in ADCI (medial temporal: 65.8%, diffuse: 34.2%) and SVCI (frontotemporal: 47.1%, parietal: 52.9%) patients. The medial temporal subtype of ADCI shows a faster decline in attention, visuospatial, visual memory, and frontal/executive domains than the diffuse subtype (p-value < 0.01). However, there are no significant differences in longitudinal cognitive outcomes between the two subtypes of SVCI. Our study provides valuable insights into the distinct spatiotemporal patterns of cortical thinning in patients with ADCI and SVCI, suggesting the potential for individualized therapeutic and preventive strategies to improve clinical outcomes.
Collapse
Affiliation(s)
- Jinhee Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Departments of Neurology, Severance Hospital, Yonsei University School of Medicine, Seoul, Korea
| | - Jonghoon Kim
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Yu-Hyun Park
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University of Medicine, Seoul, Korea
- Biostatistics and Clinical Epidemiology Center, Samsung Medical Center, Seoul, Korea
| | - Heejin Yoo
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University of Medicine, Seoul, Korea
- Biostatistics and Clinical Epidemiology Center, Samsung Medical Center, Seoul, Korea
| | - Jun Pyo Kim
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University of Medicine, Seoul, Korea
- Biostatistics and Clinical Epidemiology Center, Samsung Medical Center, Seoul, Korea
- Samsung Alzheimers Convergence Research Center, Samsung Medical Center, Seoul, Korea
| | - Hyemin Jang
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University of Medicine, Seoul, Korea
- Biostatistics and Clinical Epidemiology Center, Samsung Medical Center, Seoul, Korea
- Samsung Alzheimers Convergence Research Center, Samsung Medical Center, Seoul, Korea
| | - Hyunjin Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea.
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea.
| | - Sang Won Seo
- Departments of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University of Medicine, Seoul, Korea.
- Biostatistics and Clinical Epidemiology Center, Samsung Medical Center, Seoul, Korea.
- Samsung Alzheimers Convergence Research Center, Samsung Medical Center, Seoul, Korea.
| |
Collapse
|
8
|
Li JQ, Song JH, Suckling J, Wang YJ, Zuo CT, Zhang C, Gao J, Song YQ, Xie AM, Tan L, Yu JT. Disease trajectories in older adults with non-AD pathologic change and comparison with Alzheimer's disease pathophysiology: A longitudinal study. Neurobiol Aging 2024; 134:106-114. [PMID: 38056216 DOI: 10.1016/j.neurobiolaging.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
Based on the 'AT(N)' system, individuals with normal amyloid biomarkers but abnormal tauopathy or neurodegeneration biomarkers are classified as non-Alzheimer's disease (AD) pathologic change. This study aimed to assess the long-term clinical and cognitive trajectories of individuals with non-AD pathologic change among older adults without dementia, comparing them to those with normal AD biomarkers and AD pathophysiology. Analyzing Alzheimer's Disease Neuroimaging Initiative data, we evaluated clinical outcomes and conversion risk longitudinally using mixed effects models and multivariate Cox proportional hazard models. We found that compared to individuals with A-T-N-, those with abnormal tauopathy or neurodegeneration biomarkers (A-T + N-, A-T-N + , and A-T + N + ) had a faster rate of cognitive decline and disease progression. Individuals with A-T + N + had a faster rate of decline than those with A-T + N-. Additionally, in individuals with the same baseline tauopathy and neurodegeneration biomarker status, the presence of baseline amyloid could accelerate cognitive decline and clinical progression. These findings provide a foundation for future studies on non-AD pathologic change and its comparison with AD pathophysiology.
Collapse
Affiliation(s)
- Jie-Qiong Li
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China.
| | - Jing-Hui Song
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - John Suckling
- Department of Psychiatry, University of Cambridge, Cambridge CB2 1TN, UK; Medical Research Council and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge CB2 1TN, UK; Cambridgeshire and Peterborough NHS Trust, UK
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Chuan-Tao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai 200433, China
| | - Can Zhang
- Genetics and Aging Research Unit, Mass GeneralInstitute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown 02138, MA 02129-2060, USA
| | - Jing Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Yu-Qiang Song
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - An-Mu Xie
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital,Qingdao University, Qingdao 266000, Shandong, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for NeurologicalDisorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200040, China.
| |
Collapse
|
9
|
Di Napoli J, Arighi A, Conte G, Carandini T, Sacchi L, Arcaro M, Fenoglio C, Sorrentino F, Mercurio M, Pietroboni AM, Giardinieri G, Triulzi F, Galimberti D, Scarpini E, Fumagalli GG. Predominant right temporal lobe atrophy: Clinical, neuropsychological and structural differences based on amyloid status. Eur J Neurol 2024; 31:e16124. [PMID: 37933893 PMCID: PMC11235810 DOI: 10.1111/ene.16124] [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: 05/31/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Predominant right temporal atrophy is a radiological sign usually associated with frontotemporal dementia but this sign can also be present in Alzheimer's disease. Given the overlap of clinical symptoms between the two conditions, it is important to know which characteristics allow them to be differentiated. OBJECTIVES To compare clinical, neuropsychological and structural magnetic resonance imaging (MRI) data of subjects with prominent right anterior temporal atrophy, depending on the status of amyloid biomarkers. METHODS Among patients followed in the dementia center of Ospedale Maggiore Policlinico, subjects with right anterior temporal atrophy, defined as grade 3 or 4 on the corresponding visual rating scale, were identified. Only subjects with both an MRI scan and amyloid status available were considered. For selected subjects, data were extracted from clinical and neuropsychological records at initial presentation and at last available follow-up. Two raters applied a protocol of eight visual rating scales to compare brain atrophy and white matter hyperintensities. RESULTS Of 497 subjects, 17 fulfilled the inclusion criteria: 7 amyloid-positive and 10 amyloid-negative. At initial presentation, executive dysfunction and topographical disorientation were more common in amyloid-positive patients. At follow-up, behavioral symptoms, such as social awkwardness and compulsive attitude, were more frequent in the amyloid-negative patients. Amyloid-positive patients presented an overall worse neuropsychological performance, especially in the language and visuospatial domain, and had higher scores on the right anterior cingulate visual rating scale. CONCLUSION Patients with predominant right temporal atrophy showed clinical, neuropsychological and radiological differences, depending on the status of amyloid biomarkers.
Collapse
Affiliation(s)
- Jacopo Di Napoli
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Andrea Arighi
- Neurodegenerative Diseases UnitFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Giorgio Conte
- Neuroradiology UnitFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Tiziana Carandini
- Neurodegenerative Diseases UnitFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Luca Sacchi
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Marina Arcaro
- Neurodegenerative Diseases UnitFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Chiara Fenoglio
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Federica Sorrentino
- Department of Biomedical Surgical and Dental SciencesUniversity of MilanMilanItaly
| | - Matteo Mercurio
- Neurodegenerative Diseases UnitFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Anna M. Pietroboni
- Neurodegenerative Diseases UnitFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Giulia Giardinieri
- Neurodegenerative Diseases UnitFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Fabio Triulzi
- Neuroradiology UnitFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | - Daniela Galimberti
- Neurodegenerative Diseases UnitFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
- Department of Biomedical Surgical and Dental SciencesUniversity of MilanMilanItaly
| | - Elio Scarpini
- Neurodegenerative Diseases UnitFondazione IRCCS Ca' GrandaOspedale Maggiore PoliclinicoMilanItaly
| | | |
Collapse
|
10
|
Caminiti SP, De Francesco S, Tondo G, Galli A, Redolfi A, Perani D. FDG-PET markers of heterogeneity and different risk of progression in amnestic MCI. Alzheimers Dement 2024; 20:159-172. [PMID: 37505996 PMCID: PMC10962797 DOI: 10.1002/alz.13385] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/19/2023] [Accepted: 06/12/2023] [Indexed: 07/30/2023]
Abstract
INTRODUCTION Amnestic mild cognitive impairment (aMCI) is emerging as a heterogeneous condition. METHODS We looked at a cohort of N = 207 aMCI subjects, with baseline fluorodeoxyglucose positron emission tomography (FDG-PET), T1 magnetic resonance imaging, cerebrospinal fluid (CSF), apolipoprotein E (APOE), and neuropsychological assessment. An algorithm based on FDG-PET hypometabolism classified each subject into subtypes, then compared biomarker measures and clinical progression. RESULTS Three subtypes emerged: hippocampal sparing-cortical hypometabolism, associated with younger age and the highest level of Alzheimer's disease (AD)-CSF pathology; hippocampal/cortical hypometabolism, associated with a high percentage of APOE ε3/ε4 or ε4/ε4 carriers; medial-temporal hypometabolism, characterized by older age, the lowest AD-CSF pathology, the most severe hippocampal atrophy, and a benign course. Within the whole cohort, the severity of temporo-parietal hypometabolism, correlated with AD-CSF pathology and marked the rate of progression of cognitive decline. DISCUSSION FDG-PET can distinguish clinically comparable aMCI at single-subject level with different risk of progression to AD dementia or stability. The obtained results can be useful for the optimization of pharmacological trials and automated-classification models. HIGHLIGHTS Algorithm based on FDG-PET hypometabolism demonstrates distinct subtypes across aMCI; Three different subtypes show heterogeneous biological profiles and risk of progression; The cortical hypometabolism is associated with AD pathology and cognitive decline; MTL hypometabolism is associated with the lowest conversion rate and CSF-AD pathology.
Collapse
Affiliation(s)
- Silvia Paola Caminiti
- Vita‐Salute San Raffaele UniversityMilanItaly
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Silvia De Francesco
- Laboratory of NeuroinformaticsIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Giacomo Tondo
- Vita‐Salute San Raffaele UniversityMilanItaly
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Alice Galli
- Vita‐Salute San Raffaele UniversityMilanItaly
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Alberto Redolfi
- Laboratory of NeuroinformaticsIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Daniela Perani
- Vita‐Salute San Raffaele UniversityMilanItaly
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
| | | |
Collapse
|
11
|
Mesulam MM, Gefen T, Flanagan M, Castellani R, Jamshidi P, Barbieri E, Sridhar J, Kawles A, Weintraub S, Geula C, Rogalski E. Frontotemporal Degeneration with Transactive Response DNA-Binding Protein Type C at the Anterior Temporal Lobe. Ann Neurol 2023; 94:1-12. [PMID: 37183762 PMCID: PMC10330481 DOI: 10.1002/ana.26677] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/28/2023] [Accepted: 05/06/2023] [Indexed: 05/16/2023]
Abstract
The anatomical distribution of most neurodegenerative diseases shows considerable interindividual variations. In contrast, frontotemporal lobar degeneration with transactive response DNA-binding protein type C (TDP-C) shows a consistent predilection for the anterior temporal lobe (ATL). The relatively selective atrophy of ATL in TDP-C patients has highlighted the importance of this region for complex cognitive and behavioral functions. This review includes observations on 28 TDP-C patients, 18 with semantic primary progressive aphasia and 10 with other syndromes. Longitudinal imaging allowed the delineation of progression trajectories. At post-mortem examination, the pathognomonic feature of TDP-C consisted of long, thick neurites found predominantly in superficial cortical layers. These neurites may represent dystrophic apical dendrites of layer III and V pyramidal neurons that are known to play pivotal roles in complex cortical computations. Other types of frontotemporal lobar degeneration TDP, such as TDP-A and TDP-B, are not associated with long dystrophic neurites in the cerebral cortex, and do not show similar predilection patterns for ATL. Research is beginning to identify molecular, structural, and immunological differences between pathological TDP-43 in TDP-C versus TDP-A and B. Parallel investigations based on proteomics, somatic mutations, and genome-wide association studies are detecting molecular features that could conceivably mediate the selective vulnerability of ATL to TDP-C. Future work will focus on characterizing the distinctive features of the abnormal TDP-C neurites, the mechanisms of neurotoxicity, initial cellular targets within the ATL, trajectory of spread, and the nature of ATL-specific markers that modulate vulnerability to TDP-C. ANN NEUROL 2023;94:1-12.
Collapse
Affiliation(s)
- Marek-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Margaret Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rudolph Castellani
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pouya Jamshidi
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elena Barbieri
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jaiashre Sridhar
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Allegra Kawles
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Psychiatry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
12
|
Dávila G, Torres-Prioris MJ, López-Barroso D, Berthier ML. Turning the Spotlight to Cholinergic Pharmacotherapy of the Human Language System. CNS Drugs 2023; 37:599-637. [PMID: 37341896 PMCID: PMC10374790 DOI: 10.1007/s40263-023-01017-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2023] [Indexed: 06/22/2023]
Abstract
Even though language is essential in human communication, research on pharmacological therapies for language deficits in highly prevalent neurodegenerative and vascular brain diseases has received little attention. Emerging scientific evidence suggests that disruption of the cholinergic system may play an essential role in language deficits associated with Alzheimer's disease and vascular cognitive impairment, including post-stroke aphasia. Therefore, current models of cognitive processing are beginning to appraise the implications of the brain modulator acetylcholine in human language functions. Future work should be directed further to analyze the interplay between the cholinergic system and language, focusing on identifying brain regions receiving cholinergic innervation susceptible to modulation with pharmacotherapy to improve affected language domains. The evaluation of language deficits in pharmacological cholinergic trials for Alzheimer's disease and vascular cognitive impairment has thus far been limited to coarse-grained methods. More precise, fine-grained language testing is needed to refine patient selection for pharmacotherapy to detect subtle deficits in the initial phases of cognitive decline. Additionally, noninvasive biomarkers can help identify cholinergic depletion. However, despite the investigation of cholinergic treatment for language deficits in Alzheimer's disease and vascular cognitive impairment, data on its effectiveness are insufficient and controversial. In the case of post-stroke aphasia, cholinergic agents are showing promise, particularly when combined with speech-language therapy to promote trained-dependent neural plasticity. Future research should explore the potential benefits of cholinergic pharmacotherapy in language deficits and investigate optimal strategies for combining these agents with other therapeutic approaches.
Collapse
Affiliation(s)
- Guadalupe Dávila
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain
- Department of Psychobiology and Methodology of Behavioral Sciences, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
| | - María José Torres-Prioris
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain
- Department of Psychobiology and Methodology of Behavioral Sciences, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
| | - Diana López-Barroso
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain
- Department of Psychobiology and Methodology of Behavioral Sciences, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
| | - Marcelo L Berthier
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain.
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain.
| |
Collapse
|
13
|
Lu J, Zhang Z, Wu P, Liang X, Zhang H, Hong J, Clement C, Yen TC, Ding S, Wang M, Xiao Z, Rominger A, Shi K, Guan Y, Zuo C, Zhao Q. The heterogeneity of asymmetric tau distribution is associated with an early age at onset and poor prognosis in Alzheimer's disease. Neuroimage Clin 2023; 38:103416. [PMID: 37137254 PMCID: PMC10176076 DOI: 10.1016/j.nicl.2023.103416] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/13/2023] [Accepted: 04/22/2023] [Indexed: 05/05/2023]
Abstract
PURPOSE Left-right asymmetry, an important feature of brain development, has been implicated in neurodegenerative diseases, although it's less discussed in typical Alzheimer's disease (AD). We sought to investigate whether asymmetric tau deposition plays a potential role in AD heterogeneity. METHODS Two independent cohorts consisting of patients with mild cognitive impairment due to AD and AD dementia with tau PET imaging were enrolled [the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort with 18F-Flortaucipir, the Shanghai Memory Study (SMS) cohort with 18F-Florzolotau]. Based on the absolute global tau interhemispheric differences, each cohort was divided into two groups (asymmetric versus symmetric tau distribution). The two groups were cross-sectionally compared in terms of demographic, cognitive characteristics, and pathological burden. The cognitive decline trajectories were analyzed longitudinally. RESULTS Fourteen (23.3%) and 42 (48.3%) patients in the ADNI and SMS cohorts showed an asymmetric tau distribution, respectively. An asymmetric tau distribution was associated with an earlier age at disease onset (proportion of early-onset AD: ADNI/SMS/combined cohorts, p = 0.093/0.026/0.001) and more severe pathological burden (i.e., global tau burden: ADNI/SMS cohorts, p < 0.001/= 0.007). And patients with an asymmetric tau distribution were characterized by a steeper cognitive decline longitudinally (i.e., the annual decline of Mini-Mental Status Examination score: ADNI/SMS/combined cohorts, p = 0.053 / 0.035 / < 0.001). CONCLUSIONS Asymmetry in tau deposition, which may be associated with an earlier age at onset, more severe pathological burden, and a steeper cognitive decline, is potentially an important characteristic of AD heterogeneity.
Collapse
Affiliation(s)
- Jiaying Lu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China; Department of Nuclear Medicine, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Zhengwei Zhang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ping Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoniu Liang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Huiwei Zhang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jimin Hong
- Department of Nuclear Medicine, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Christoph Clement
- Department of Nuclear Medicine, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | | | - Saineng Ding
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Wang
- Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China; Department of Informatics, Technische Universität München, Munich, Germany
| | - Zhenxu Xiao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland; Department of Informatics, Technische Universität München, Munich, Germany
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China.
| | - Chuantao Zuo
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China.
| | - Qianhua Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China; MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Li JQ, Song JH, Suckling J, Wang YJ, Zuo CT, Zhang C, Gao J, Song YQ, Xie AM, Tan L, Yu JT. Disease trajectories in elders with suspected non-Alzheimer's pathophysiology and its comparison with Alzheimer's disease pathophysiology: a longitudinal study. RESEARCH SQUARE 2023:rs.3.rs-2744271. [PMID: 37034751 PMCID: PMC10081361 DOI: 10.21203/rs.3.rs-2744271/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Background According to the new 'AT(N)' system, those with a normal amyloid biomarker but with abnormal tauopathy or biomarkers of neurodegeneration or neuronal injury, have been labeled suspected non-Alzheimer's pathophysiology (SNAP). We aimed to estimate the long-term clinical and cognitive trajectories of SNAP individuals in non-demented elders and its comparison with individual in the Alzheimer's disease (AD) pathophysiology using 'AT(N)' system. Methods We included individuals with available baseline cerebrospinal fluid (CSF) Aβ (A), CSF phosphorylated tau examination (T) and 18F-uorodeoxyglucose PET or volumetric magnetic resonance imaging (N) from the Alzheimer's Disease Neuroimaging Initiative database. Longitudinal change in clinical outcomes are assessed using linear mixed effects models. Conversion risk from cognitively normal (CN) to cognitively impairment, and conversion from mild cognitive impairment (MCI) to dementia are assessed using multivariate Cox proportional hazard models. Results Totally, 366 SNAP individuals were included (114 A-T-N-, 154 A-T + N-, 54 A-T-N + and 44 A-T + N+) of whom 178 were CN and 188 were MCI. Compared with A-T-N-, CN elders with A-T + N-, A-T-N + and A-T + N + had a faster rate of ADNI-MEM score decline. Moreover, CN older individuals with A-T + N + also had a faster rate of decline in ADNI-MEM score than those with A-T + N- individuals. MCI patients with A-T + N + had a faster rate of ADNI-MEM and ADNI-EF decline and hippocampal volume loss compared with A-T-N- and A-T + N- profiles. CN older individuals with A-T + N + had an increased risk of conversion to cognitive impairment (CDR-GS ≥ 0.5) compared with A-T + N- and A-T-N-. In MCI patients, A-T + N + also had an increased risk of conversion to dementia compared with A-T + N- and A-T-N-. Compared with A-T + N-, CN elders and MCI patients with A + T + N- and A + T + N + had a faster rate of ADNI-MEM score, ADNI-EF score decline, and hippocampal volume loss. CN individuals with A + T + N + had a faster rate of ADNI-EF score decline compare with A-T + N + individuals. Moreover, MCI patients with A + T + N + also had a faster rate of decline in ADNI-MEM score, ADNI-EF score and hippocampal volume loss than those with A-T + N + individuals. Conclusions The findings from clinical, imaging and biomarker studies on SNAP, and its comparison with AD pathophysiology offered an important foundation for future studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Can Zhang
- Massachusetts General Hospital, Harvard Medical School
| | | | | | | | | | | |
Collapse
|
15
|
Kawles A, Minogue G, Zouridakis A, Keszycki R, Gill N, Nassif C, Coventry C, Zhang H, Rogalski E, Flanagan ME, Castellani R, Bigio EH, Mesulam MM, Geula C, Gefen T. Differential vulnerability of the dentate gyrus to tauopathies in dementias. Acta Neuropathol Commun 2023; 11:1. [PMID: 36597124 PMCID: PMC9811688 DOI: 10.1186/s40478-022-01485-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/05/2023] Open
Abstract
The dentate gyrus (DG), a key hippocampal subregion in memory processing, generally resists phosphorylated tau accumulation in the amnestic dementia of the Alzheimer's type due to Alzheimer's disease (DAT-AD), but less is known about the susceptibility of the DG to other tauopathies. Here, we report stereologic densities of total DG neurons and tau inclusions in thirty-two brains of human participants with autopsy-confirmed tauopathies with distinct isoform profiles-3R Pick's disease (PiD, N = 8), 4R corticobasal degeneration (CBD, N = 8), 4R progressive supranuclear palsy (PSP, N = 8), and 3/4R AD (N = 8). All participants were diagnosed during life with primary progressive aphasia (PPA), an aphasic clinical dementia syndrome characterized by progressive deterioration of language abilities with spared non-language cognitive abilities in early stages, except for five patients with DAT-AD as a comparison group. 51% of total participants were female. All specimens were stained immunohistochemically with AT8 to visualize tau pathology, and PPA cases were stained for Nissl substance to visualize neurons. Unbiased stereological analysis was performed in granule and hilar DG cells, and inclusion-to-neuron ratios were calculated. In the PPA group, PiD had highest mean total (granule + hilar) densities of DG tau pathology (p < 0.001), followed by CBD, AD, then PSP. PPA-AD cases showed more inclusions in hilar cells compared to granule cells, while the opposite was true in PiD and CBD. Inclusion-to-neuron ratios revealed, on average, 33% of all DG neurons in PiD cases contained a tau inclusion, compared to ~ 7% in CBD, 2% in AD, and 0.4% in PSP. There was no significant difference between DAT-AD and PPA-AD pathologic tau burden, suggesting that differences in DG burden are not specific to clinical phenotype. We conclude that the DG is differentially vulnerable to pathologic tau accumulation, raising intriguing questions about the structural integrity and functional significance of hippocampal circuits in neurodegenerative dementias.
Collapse
Affiliation(s)
- Allegra Kawles
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Grace Minogue
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
| | - Antonia Zouridakis
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
| | - Rachel Keszycki
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Nathan Gill
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Caren Nassif
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
| | - Hui Zhang
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Margaret E. Flanagan
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Rudolph Castellani
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Eileen H. Bigio
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - M. Marsel Mesulam
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology & Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, 300 E. Superior Street, Tarry Building, 8th Floor, Chicago, IL 60611 USA
- Department of Psychiatry & Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL USA
| |
Collapse
|
16
|
Characterization of the logopenic variant of Primary Progressive Aphasia: A systematic review and meta-analysis. Ageing Res Rev 2022; 82:101760. [PMID: 36244629 DOI: 10.1016/j.arr.2022.101760] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/11/2022] [Indexed: 01/31/2023]
Abstract
The linguistic and anatomical variability of the logopenic variant of Primary Progressive Aphasia (lv-PPA) as defined by current diagnostic criteria has been the topic of an intense debate. The present review and meta-analysis aims at characterizing the profile of lv-PPA, by a comprehensive analysis of the available literature on the neuropsychological, neuroimaging, electrophysiological, pathological, and genetic features of lv-PPA. We conducted a systematic bibliographic search, leading to the inclusion of 207 papers. Of them, 12 were used for the Anatomical Likelihood Estimation meta-analysis on grey matter revealed by magnetic resonance imaging data. The results suggest that the current guidelines outline a relatively consistent syndrome, characterized by a core set of linguistic and, to a lesser extent, non-linguistic deficits, mirroring the involvement of left temporal and parietal regions typically affected by Alzheimer Disease pathology. Variations of the lv-PPA profile are discussed in terms of heterogeneity of the neuropsychological instruments and the diagnostic criteria adopted.
Collapse
|
17
|
Dunlop SR, Ayala I, Spencer C, Flanagan ME, Mesulam MM, Gefen T, Geula C. Resistance of Basal Forebrain Cholinergic Neurons to TDP-43 Proteinopathy in Primary Progressive Aphasia. J Neuropathol Exp Neurol 2022; 81:910-919. [PMID: 36111818 PMCID: PMC9582786 DOI: 10.1093/jnen/nlac079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Basal forebrain cholinergic neurons (BFCN) display accumulation of neurofibrillary tangles and degeneration in Alzheimer disease and are targets of therapeutic intervention. This study determined vulnerability of BFCN to accumulation of TDP-43 in primary progressive aphasia with TDP-43 proteinopathy (PPA-TDP). Brains from 16 PPA participants with pathologically confirmed TDP-43 proteinopathy, with available paraffin-embedded sections (Group 1), or systematically sampled frozen sections (Group 2), were studied. Immunohistochemistry was performed with an antibody against phosphorylated TDP-43. BFCN were identified by their magnocellular appearance in Nissl preparations. Presence of TDP-43 inclusions and preinclusions in BFCN was determined and quantitative analysis was performed in Group 2. In Group 1, BFCN were completely free of inclusions except for occasional dystrophic neurites. Sparse TDP-43 preinclusions with smooth or granular staining in BFCN were detected. In Group 2, extremely rare TDP-43 intranuclear inclusions were detected in 0.1% of BFCN per section, along with occasional dystrophic neurites. Although sparse, significantly more preinclusions (1.4% of BFCN) were present when compared with inclusions. No hemispheric differences were noted. Small neurons near BFCN contained more preinclusions compared with BFCN. Thus, BFCN in PPA-TDP are resistant to TDP-43 proteinopathy and degeneration, suggesting that cholinergic therapy is unlikely to be effective in this disorder.
Collapse
Affiliation(s)
- Sara Rose Dunlop
- From the Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ivan Ayala
- From the Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Callen Spencer
- From the Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Margaret E Flanagan
- From the Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Marek-Marsel Mesulam
- From the Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tamar Gefen
- From the Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Changiz Geula
- From the Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
18
|
Focal amyloid and asymmetric tau in an imaging-to-autopsy case of clinical primary progressive aphasia with Alzheimer disease neuropathology. Acta Neuropathol Commun 2022; 10:111. [PMID: 35945628 PMCID: PMC9361632 DOI: 10.1186/s40478-022-01412-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/17/2022] [Indexed: 01/21/2023] Open
Abstract
Quantification of in vivo amyloid and tau PET imaging relationships with postmortem measurements are critical for validating the sensitivity and specificity imaging biomarkers across clinical phenotypes with Alzheimer disease neuropathologic change (ADNC). This study examined the quantitative relationship between regional binding of in vivo 18F-florbetapir amyloid PET and 18F-flortaucipir tau PET with postmortem stereological counts of amyloid plaques and neurofibrillary tangles (NFT) in a case of primary progressive aphasia (PPA) with ADNC, where neurodegeneration asymmetrically targets the left hemisphere. Beginning 2 years prior to death, a 63-year-old right-handed man presenting with agrammatic variant PPA underwent a florbetapir and flortaucpir PET scan, and neuropsychological assessments and magnetic resonance imaging sessions every 6 months. Florbetapir and flortaucpir PET standard uptake value ratios (SUVRs) were quantified from 8 left and right hemisphere brain regions with stereological quantification of amyloid plaques and NFTs from corresponding postmortem sections. Pearson's correlations and measures of asymmetry were used to examine relationships between imaging and autopsy measurements. The three visits prior to death revealed decline of language measures, with marked progression of atrophy. Florbetapir PET presented with an atypical focal pattern of uptake and showed a significant positive correlation with postmortem amyloid plaque density across the 8 regions (r = 0.92; p = 0.001). Flortaucipir PET had a left-lateralized distribution and showed a significant positive correlation with NFT density (r = 0.78; p = 0.023). Flortaucipir PET and NFT density indicated a medial temporal lobe sparing presentation of ADNC, demonstrating that AD does not always target the medial temporal lobe. This study adds additional evidence, in a non-amnestic phenotype of ADNC, that there is a strong correlation between AD PET biomarkers, florbetapir and flortaucipir, with quantitative neuropathology. The atypical and focal presentation of plaque density and florbetapir PET uptake suggests not all amyloid pathology presents as diffuse across neocortex.
Collapse
|
19
|
Wu Y, Wu X, Gao L, Yan Y, Geng Z, Zhou S, Zhu W, Tian Y, Yu Y, Wei L, Wang K. Abnormal Functional Connectivity of Thalamic Subdivisions in Alzheimer's Disease: A Functional Magnetic Resonance Imaging Study. Neuroscience 2022; 496:73-82. [PMID: 35690336 DOI: 10.1016/j.neuroscience.2022.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/23/2022] [Accepted: 06/02/2022] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD) is characterized by global cognitive impairment in multiple cognitive domains. Thalamic dysfunction during AD progression has been reported. However, there are limited studies regarding dysfunction in the functional connectivity (FC) of thalamic subdivisions and the relationship between such dysfunction and clinical assessments. This study examined dysfunction in the FC of thalamic subdivisions and determined the relationship between such dysfunction and clinical assessments. Forty-eight patients with AD and 47 matched healthy controls were recruited and assessed with scales for multiple cognitive domains. Group-wise comparisons of FC with thalamic subdivisions as seed points were conducted to identify abnormal cerebral regions. Moreover, correlation analysis was conducted to evaluate the relationship between abnormal FC and cognitive performance. Decreased FC of the intralaminar and medial nuclei with the left precuneus was observed in patients but not in heathy controls. The abnormal FC of the medial nuclei with the left precuneus was correlated with the Mini Mental State Examination score in the patient group. Using the FC values showing between-group differences, the linear support vector machine classifier achieved quite good in accuracy, sensitivity, specificity and area under the curve. Dysfunction in the FC of the intralaminar and medial thalamus with the precuneus may comprise a potential neural substrate for cognitive impairment during AD progression, which in turn may provide new treatment targets.
Collapse
Affiliation(s)
- Yue Wu
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui 230022, China
| | - Xingqi Wu
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui 230022, China
| | - Liying Gao
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui 230022, China
| | - Yibing Yan
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui 230022, China
| | - Zhi Geng
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui 230022, China; Department of Neurology, Second People's Hospital of Hefei City, The Hefei Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Shanshan Zhou
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui 230022, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui Province 230022, China
| | - Wanqiu Zhu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yanghua Tian
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui 230022, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui 230088, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui Province 230022, China
| | - Yongqiang Yu
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China.
| | - Ling Wei
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui 230022, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui Province 230022, China.
| | - Kai Wang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui 230022, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui 230088, China; The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, Anhui Province 230032, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui Province 230022, China.
| |
Collapse
|
20
|
Cho S, Cousins KAQ, Shellikeri S, Ash S, Irwin DJ, Liberman MY, Grossman M, Nevler N. Lexical and Acoustic Speech Features Relating to Alzheimer Disease Pathology. Neurology 2022; 99:e313-e322. [PMID: 35487701 PMCID: PMC9421771 DOI: 10.1212/wnl.0000000000200581] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/08/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES We compared digital speech and language features of patients with amnestic Alzheimer disease (aAD) or logopenic variant primary progressive aphasia (lvPPA) in a biologically confirmed cohort and related these features to neuropsychiatric test scores and CSF analytes. METHODS We included patients with aAD or lvPPA with CSF (phosphorylated tau ([p-tau]/β-amyloid [Aβ] ≥0.09, and total tau/Aβ ≥0.34) or autopsy confirmation of AD pathology and age-matched healthy controls (HC) recruited at the Frontotemporal Degeneration Center of the University of Pennsylvania for a cross-sectional study. We extracted speech and language variables with automated lexical and acoustic pipelines from participants' oral picture descriptions. We compared the groups and correlated distinct features with clinical ratings and CSF p-tau levels. RESULTS We examined patients with aAD (n = 44; age 62 ± 8 years; 24 women; Mini-Mental State Examination [MMSE] score 21.1 ± 4.8) or lvPPA (n = 21; age 64.1 ± 8.2 years; 11 women; MMSE score 23.0 ± 4.2) and HC (n = 28; age 65.9 ± 5.9 years, 15 women; MMSE score 29 ± 1). Patients with lvPPA produced fewer verbs (10.5 ± 2.3; p = 0.001) and adjectives (2.7 ± 1.3, p = 0.019) and more fillers (7.4 ± 3.9; p = 0.022) with lower lexical diversity (0.84 ± 0.1; p = 0.05) and higher pause rate (54.2 ± 19.2; p = 0.015) than individuals with aAD (verbs 12.5 ± 2; adjectives 3.8 ± 2; fillers 4.9 ± 4.5; lexical diversity 0.87 ± 0.1; pause rate 45.3 ± 12.8). Both groups showed some shared language impairments compared with HC. Word frequency (MMSE score: β = -1.6, p = 0.009; Boston Naming Test [BNT] score: β = -4.36, p < 0.001), adverbs (MMSE score: β = -1.9, p = 0.003; BNT score: β = -2.41, p = 0.041), pause rate (MMSE score: β = -1.21, p = 0.041; BNT score: β = -2.09, p = 0.041), and word length (MMSE score: β = 1.75, p = 0.001; BNT score: β = 2.94, p = 0.003) were significantly correlated with both MMSE and BNT scores, but other measures were not correlated with MMSE and/or BNT score. Prepositions (r = -0.36, p = 0.019), nouns (r = -0.31, p = 0.047), speech segment duration (r = -0.33, p = 0.032), word frequency (r = 0.33, p = 0.036), and pause rate (r = 0.34, p = 0.026) were correlated with patients' CSF p-tau levels. DISCUSSION Our measures captured language and speech differences between the 2 phenotypes that traditional language-based clinical assessments failed to identify. This work demonstrates the potential of natural speech in reflecting underlying variants with AD pathology.
Collapse
Affiliation(s)
- Sunghye Cho
- From the Linguistic Data Consortium (S.C., M.Y.L.) and Department of Neurology (K.A.Q.C., S.S., S.A., D.J.I., M.G., N.N.), University of Pennsylvania, Philadelphia.
| | - Katheryn Alexandra Quilico Cousins
- From the Linguistic Data Consortium (S.C., M.Y.L.) and Department of Neurology (K.A.Q.C., S.S., S.A., D.J.I., M.G., N.N.), University of Pennsylvania, Philadelphia
| | - Sanjana Shellikeri
- From the Linguistic Data Consortium (S.C., M.Y.L.) and Department of Neurology (K.A.Q.C., S.S., S.A., D.J.I., M.G., N.N.), University of Pennsylvania, Philadelphia
| | - Sharon Ash
- From the Linguistic Data Consortium (S.C., M.Y.L.) and Department of Neurology (K.A.Q.C., S.S., S.A., D.J.I., M.G., N.N.), University of Pennsylvania, Philadelphia
| | - David John Irwin
- From the Linguistic Data Consortium (S.C., M.Y.L.) and Department of Neurology (K.A.Q.C., S.S., S.A., D.J.I., M.G., N.N.), University of Pennsylvania, Philadelphia
| | - Mark Yoffe Liberman
- From the Linguistic Data Consortium (S.C., M.Y.L.) and Department of Neurology (K.A.Q.C., S.S., S.A., D.J.I., M.G., N.N.), University of Pennsylvania, Philadelphia
| | - Murray Grossman
- From the Linguistic Data Consortium (S.C., M.Y.L.) and Department of Neurology (K.A.Q.C., S.S., S.A., D.J.I., M.G., N.N.), University of Pennsylvania, Philadelphia
| | - Naomi Nevler
- From the Linguistic Data Consortium (S.C., M.Y.L.) and Department of Neurology (K.A.Q.C., S.S., S.A., D.J.I., M.G., N.N.), University of Pennsylvania, Philadelphia
| |
Collapse
|
21
|
Mesulam MM, Coventry CA, Bigio EH, Sridhar J, Gill N, Fought AJ, Zhang H, Thompson CK, Geula C, Gefen T, Flanagan M, Mao Q, Weintraub S, Rogalski EJ. Neuropathological fingerprints of survival, atrophy and language in primary progressive aphasia. Brain 2022; 145:2133-2148. [PMID: 35441216 PMCID: PMC9246707 DOI: 10.1093/brain/awab410] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/25/2021] [Accepted: 10/19/2021] [Indexed: 01/21/2023] Open
Abstract
Primary progressive aphasia is a neurodegenerative disease that selectively impairs language without equivalent impairment of speech, memory or comportment. In 118 consecutive autopsies on patients with primary progressive aphasia, primary diagnosis was Alzheimer's disease neuropathological changes (ADNC) in 42%, corticobasal degeneration or progressive supranuclear palsy neuropathology in 24%, Pick's disease neuropathology in 10%, transactive response DNA binding proteinopathy type A [TDP(A)] in 10%, TDP(C) in 11% and infrequent entities in 3%. Survival was longest in TDP(C) (13.2 ± 2.6 years) and shortest in TDP(A) (7.1 ± 2.4 years). A subset of 68 right-handed participants entered longitudinal investigations. They were classified as logopenic, agrammatic/non-fluent or semantic by quantitative algorithms. Each variant had a preferred but not invariant neuropathological correlate. Seventy-seven per cent of logopenics had ADNC, 56% of agrammatics had corticobasal degeneration/progressive supranuclear palsy or Pick's disease and 89% of semantics had TDP(C). Word comprehension impairments had strong predictive power for determining underlying neuropathology positively for TDP(C) and negatively for ADNC. Cortical atrophy was smallest in corticobasal degeneration/progressive supranuclear palsy and largest in TDP(A). Atrophy encompassed posterior frontal but not temporoparietal cortex in corticobasal degeneration/progressive supranuclear palsy, anterior temporal but not frontoparietal cortex in TDP(C), temporofrontal but not parietal cortex in Pick's disease and all three lobes with ADNC or TDP(A). There were individual deviations from these group patterns, accounting for less frequent clinicopathologic associations. The one common denominator was progressive asymmetric atrophy overwhelmingly favouring the left hemisphere language network. Comparisons of ADNC in typical amnestic versus atypical aphasic dementia and of TDP in type A versus type C revealed fundamental biological and clinical differences, suggesting that members of each pair may constitute distinct clinicopathologic entities despite identical downstream proteinopathies. Individual TDP(C) participants with unilateral left temporal atrophy displayed word comprehension impairments without additional object recognition deficits, helping to dissociate semantic primary progressive aphasia from semantic dementia. When common and uncommon associations were considered in the set of 68 participants, one neuropathology was found to cause multiple clinical subtypes, and one subtype of primary progressive aphasia to be caused by multiple neuropathologies, but with different probabilities. Occasionally, expected clinical manifestations of atrophy sites were absent, probably reflecting individual peculiarities of language organization. The hemispheric asymmetry of neurodegeneration and resultant language impairment in primary progressive aphasia reflect complex interactions among the cellular affinities of the degenerative disease, the constitutive biology of language cortex, familial or developmental vulnerabilities of this network and potential idiosyncrasies of functional anatomy in the affected individual.
Collapse
Affiliation(s)
- M Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
- Davee Department of Neurology, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Christina A Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Eileen H Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pathology, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jaiashre Sridhar
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Nathan Gill
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Preventive Medicine, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Angela J Fought
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hui Zhang
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Preventive Medicine, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Cynthia K Thompson
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
- School of Communication, Northwestern University, Evanston, IL 60208, USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Margaret Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pathology, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qinwen Mao
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Emily J Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
22
|
Kawles A, Nishihira Y, Feldman A, Gill N, Minogue G, Keszycki R, Coventry C, Spencer C, Lilek J, Ajroud K, Coppola G, Rademakers R, Rogalski E, Weintraub S, Zhang H, Flanagan ME, Bigio EH, Mesulam MM, Geula C, Mao Q, Gefen T. Cortical and subcortical pathological burden and neuronal loss in an autopsy series of FTLD-TDP-type C. Brain 2022; 145:1069-1078. [PMID: 34919645 PMCID: PMC9050539 DOI: 10.1093/brain/awab368] [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: 06/07/2021] [Revised: 08/11/2021] [Accepted: 08/29/2021] [Indexed: 10/31/2023] Open
Abstract
The TDP-43 type C pathological form of frontotemporal lobar degeneration is characterized by the presence of immunoreactive TDP-43 short and long dystrophic neurites, neuronal cytoplasmic inclusions, neuronal loss and gliosis and the absence of neuronal intranuclear inclusions. Frontotemporal lobar degeneration-TDP-type C cases are commonly associated with the semantic variant of primary progressive aphasia or behavioural variant frontotemporal dementia. Here, we provide detailed characterization of regional distributions of pathological TDP-43 and neuronal loss and gliosis in cortical and subcortical regions in 10 TDP-type C cases and investigate the relationship between inclusions and neuronal loss and gliosis. Specimens were obtained from the first 10 TDP-type C cases accessioned from the Northwestern Alzheimer's Disease Research Center (semantic variant of primary progressive aphasia, n = 7; behavioural variant frontotemporal dementia, n = 3). A total of 42 cortical (majority bilateral) and subcortical regions were immunostained with a phosphorylated TDP-43 antibody and/or stained with haematoxylin-eosin. Regions were evaluated for atrophy, and for long dystrophic neurites, short dystrophic neurites, neuronal cytoplasmic inclusions, and neuronal loss and gliosis using a semiquantitative 5-point scale. We calculated a 'neuron-to-inclusion' score (TDP-type C mean score - neuronal loss and gliosis mean score) for each region per case to assess the relationship between TDP-type C inclusions and neuronal loss and gliosis. Primary progressive aphasia cases demonstrated leftward asymmetry of cortical atrophy consistent with the aphasic phenotype. We also observed abundant inclusions and neurodegeneration in both cortical and subcortical regions, with certain subcortical regions emerging as particularly vulnerable to dystrophic neurites (e.g. amygdala, caudate and putamen). Interestingly, linear mixed models showed that regions with lowest TDP-type C pathology had high neuronal dropout, and conversely, regions with abundant pathology displayed relatively preserved neuronal densities (P < 0.05). This inverse relationship between the extent of TDP-positive inclusions and neuronal loss may reflect a process whereby inclusions disappear as their associated neurons are lost. Together, these findings offer insight into the putative substrates of neurodegeneration in unique dementia syndromes.
Collapse
Affiliation(s)
- Allegra Kawles
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yasushi Nishihira
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Alex Feldman
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Nathan Gill
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Grace Minogue
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rachel Keszycki
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Callen Spencer
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jaclyn Lilek
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kaouther Ajroud
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Giovanni Coppola
- Department of Psychiatry and Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hui Zhang
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Eileen H Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - M -Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qinwen Mao
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
23
|
Verbal and Nonverbal Memory in Neurodegenerative and Stroke Aphasia: Evidence From the Turkish Version of the Three Words Three Shapes Test. Cogn Behav Neurol 2022; 35:49-65. [PMID: 35239599 DOI: 10.1097/wnn.0000000000000294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/05/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Although language impairment is the most salient feature of cognitive impairment in both primary progressive aphasia (PPA) and stroke aphasia (SA), memory can also be impaired in both patient populations. OBJECTIVE To identify distinctive features of verbal and nonverbal memory processing in individuals with PPA and those with SA. METHOD We gave individuals with PPA (n = 14), those with SA (n = 8), and healthy controls (HC; n = 13) a comprehensive neuropsychological test battery and the Turkish version of the Three Words Three Shapes Test (3W3S-Turkish). The 3W3S-Turkish Test includes five subtests: Copy, Incidental Recall, Acquisition, Delayed Recall, and Recognition. High-resolution brain scans were performed in a subset of individuals with PPA and those with SA. Lesion distribution was limited to the dorsal language areas in the SA group, whereas peak atrophy areas in the PPA group extended beyond the language network, including the medial temporal lobe, precuneus, and posterior/medial portions of the cingulate cortex. RESULTS Both the PPA and SA groups showed impairment in incidental recall, and the PPA group showed additional impairment in delayed recall. Greater impairment for verbal stimuli suggestive of material-specific memory impairment was evident in the PPA group's scores on the Incidental Recall and Delayed Recall subtests. Both aphasia groups retained the acquired information regardless of material type. CONCLUSION Although both aphasia groups shared similarities in the involvement of the dorsal prefrontal working memory/attention network, the PPA group showed greater impairment in delayed recall compared with the SA group.
Collapse
|
24
|
Sun CC, Yin ZP, Chen JG, Wang WJ, Zheng GD, Li JE, Chen LL, Zhang QF. Dihydromyricetin Improves Cognitive Impairments in d-Galactose-Induced Aging Mice through Regulating Oxidative Stress and Inhibition of Acetylcholinesterase. Mol Nutr Food Res 2022; 66:e2101002. [PMID: 34932880 DOI: 10.1002/mnfr.202101002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/02/2021] [Indexed: 02/05/2023]
Abstract
SCOPE Alzheimer's disease (AD) is a neurodegenerative disease with phenomena of cognitive impairments. Oxidative stress and cholinergic system dysfunction are two widely studied pathogenesis of AD. Dihydromyricetin (DMY) is a natural dihydroflavonol with many bioactivities. In this study, it is aimed to investigate the effects of DMY on cognitive impairment in d-galactose (d-gal) induced aging mice. METHODS AND RESULTS Mice are intraperitoneally injected with d-gal for 16 weeks, and DMY is supplemented in drinking water. The results show that DMY significantly improves d-gal-induced cognitive impairments in novel object recognition and Y-maze studies. H&E and TUNEL staining show that DMY could improve histopathological changes and cell apoptosis in mice brain. DMY effectively induces the activities of catalase, superoxide dismutase and glutathione peroxidase, and reduces malondialdehyde level in mice brain and liver. Furthermore, DMY reduces cholinergic injury by inhibiting the activity of Acetylcholinesterase (AChE) in mice brain. In vitro studies show that DMY is a non-competitive inhibitor of AChE with IC50 value of 161.2 µg mL-1 . CONCLUSION DMY alleviates the cognitive impairments in d-gal-induced aging mice partly through regulating oxidative stress and inhibition of acetylcholinesterase.
Collapse
Affiliation(s)
- Cui-Cui Sun
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Zhong-Ping Yin
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Ji-Guang Chen
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Wen-Jun Wang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Guo-Dong Zheng
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jing-En Li
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Ling-Li Chen
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Qing-Feng Zhang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China
| |
Collapse
|
25
|
Jellinger KA. Recent update on the heterogeneity of the Alzheimer’s disease spectrum. J Neural Transm (Vienna) 2021; 129:1-24. [DOI: 10.1007/s00702-021-02449-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/25/2021] [Indexed: 02/03/2023]
|
26
|
Arshad M, Ahmed K, Bashir M, Kosar N, Kanwal M, Ahmed M, Khan HU, Khan S, Rauf A, Waseem A, Mahmood T. Synthesis, structural properties and potent bioactivities supported by molecular docking and DFT studies of new hydrazones derived from 5-chloroisatin and 2-thiophenecarboxaldehyde. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.131204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
27
|
Martersteck A, Sridhar J, Coventry C, Weintraub S, Mesulam MM, Rogalski E. Relationships among tau burden, atrophy, age, and naming in the aphasic variant of Alzheimer's disease. Alzheimers Dement 2021; 17:1788-1797. [PMID: 34494711 DOI: 10.1002/alz.12445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Examination of pathologic, anatomic, and cognitive relationships has been limited in primary progressive aphasia (PPA) with underlying Alzheimer's disease (AD) neuropathology. METHODS Spatial relationships between tau positron emission tomography (PET), cortical thickness, age, and naming on the Boston Naming Test (BNT) in PPA with biomarker evidence of AD (PPA-AD) were examined. RESULTS Higher tau PET burden was associated with atrophy and younger age. There was a significant left-lateralized relationship between lower BNT and more atrophy, and between lower BNT and increased tau burden. Variance in naming was primarily shared between tau and atrophy (51%), but naming was uniquely explained more by atrophy (32%) than tau (16%). Higher left anterior temporal tau burden was associated with greater 1-year rate of decline in naming. DISCUSSION PPA-AD has a similar relationship between abnormal biomarkers as first described in amnestic AD, with differing spatial extent, reflecting the left-lateralized nature of the language network.
Collapse
Affiliation(s)
- Adam Martersteck
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University (NU) Feinberg School of Medicine, Chicago, Illinois, USA.,NU Feinberg School of Medicine, Department of Radiology, Chicago, Illinois, USA
| | - Jaiashre Sridhar
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University (NU) Feinberg School of Medicine, Chicago, Illinois, USA
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University (NU) Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University (NU) Feinberg School of Medicine, Chicago, Illinois, USA.,NU Feinberg School of Medicine, Department of Psychiatry and Behavioral Sciences, Chicago, Illinois, USA
| | - M-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University (NU) Feinberg School of Medicine, Chicago, Illinois, USA.,NU Feinberg School of Medicine, Department of Neurology, Chicago, Illinois, USA
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University (NU) Feinberg School of Medicine, Chicago, Illinois, USA.,NU Feinberg School of Medicine, Department of Psychiatry and Behavioral Sciences, Chicago, Illinois, USA
| |
Collapse
|
28
|
Velilla L, Hernández J, Giraldo-Chica M, Guzmán-Vélez E, Quiroz Y, Lopera F. A Spanish Neuropsychological Battery Discriminates Between the Behavioral Variant of Frontotemporal Dementia and Primary Progressive Aphasia in a Colombian Sample. Front Neurol 2021; 12:656478. [PMID: 34290661 PMCID: PMC8287023 DOI: 10.3389/fneur.2021.656478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/17/2021] [Indexed: 12/04/2022] Open
Abstract
The differential diagnosis among the behavioral variant of frontotemporal dementia FTD (bvFTD) and the linguist one primary progressive aphasia (PPA) is challenging. Presentations of dementia type or variants dominated by personality change or aphasia are frequently misinterpreted as psychiatric illness, stroke, or other conditions. Therefore, it is important to identify cognitive tests that can distinguish the distinct FTD variants to reduce misdiagnosis and best tailor interventions. We aim to examine the discriminative capacity of the most frequently used cognitive tests in their Spanish version for the context of dementia evaluation as well as the qualitative aspects of the neuropsychological performance such as the frequency and type of errors, perseverations, and false positives that can best discriminate between bvFTD and PPA. We also described mood and behavioral profiles of participants with mild to moderate probable bvFTD and PPA. A total of 55 subjects were included in this cross-sectional study: 20 with PPA and 35 with bvFTD. All participants underwent standard dementia screening that included a medical history and physical examination, brain MRI, a semistructured caregiver interview, and neuropsychological testing. We found that bvFTD patients had worse performance in executive function tests, and the PPA presented with the lower performance in language tests and the global score of Mini-Mental State Examination (MMSE). After running the linear discriminant model, we found three functions of cognitive test and subtests combination and three functions made by the Montreal Cognitive Assessment (MoCA) language subtest and performance errors that predicted group belonging. Those functions were more capable to classify bvFTD cases rather than PPA. In conclusion, our study supports that the combination of an individual test of executive function and language, MoCA's subtest, and performance errors as well have good accuracy to discriminate between bvFTD and PPA.
Collapse
Affiliation(s)
- Lina Velilla
- Neuroscience Group of Antioquia, The University of Antioquia, Medellín, Colombia
| | - Jonathan Hernández
- Psychology Department, University Institution of Envigado, Envigado, Colombia
| | | | - Edmarie Guzmán-Vélez
- Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Yakeel Quiroz
- Neuroscience Group of Antioquia, The University of Antioquia, Medellín, Colombia.,Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Francisco Lopera
- Neuroscience Group of Antioquia, The University of Antioquia, Medellín, Colombia
| |
Collapse
|
29
|
Molecular subtyping of Alzheimer's disease with consensus non-negative matrix factorization. PLoS One 2021; 16:e0250278. [PMID: 34014928 PMCID: PMC8136734 DOI: 10.1371/journal.pone.0250278] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 04/01/2021] [Indexed: 11/19/2022] Open
Abstract
Alzheimer's disease (AD) is a heterogeneous disease and exhibits diverse clinical presentations and disease progression. Some pathological and anatomical subtypes have been proposed. However, these subtypes provide a limited mechanistic understanding for AD. Leveraging gene expression data of 222 AD patients from The Religious Orders Study and Memory and Aging Project (ROSMAP) Study, we identified two AD molecular subtypes (synaptic type and inflammatory type) using consensus non-negative matrix factorization (NMF). Synaptic type is characterized by disrupted synaptic vesicle priming and recycling and synaptic plasticity. Inflammatory type is characterized by disrupted IL2, interferon alpha and gamma pathways. The two AD molecular subtypes were validated using independent data from Gene Expression Omnibus. We further demonstrated that the two molecular subtypes are associated with APOE genotypes, with synaptic type more prevalent in AD patients with E3E4 genotype and inflammatory type more prevalent in AD patients with E3E3 genotype (p = 0.031). In addition, two molecular subtypes are differentially represented in male and female AD, with synaptic type more prevalent in male and inflammatory type in female patients (p = 0.051). Identification of AD molecular subtypes has potential in facilitating disease mechanism understanding, clinical trial design, drug discovery, and precision medicine for AD.
Collapse
|
30
|
Buciuc M, Whitwell JL, Baker MC, Rademakers R, Dickson DW, Josephs KA. Old age genetically confirmed frontotemporal lobar degeneration with TDP-43 has limbic predominant TDP-43 deposition. Neuropathol Appl Neurobiol 2021; 47:1050-1059. [PMID: 33969528 DOI: 10.1111/nan.12727] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/20/2021] [Accepted: 05/01/2021] [Indexed: 12/13/2022]
Abstract
AIMS To assess the burden of transactive response DNA-binding protein of 43 kDa (TDP-43) inclusions in a unique cohort of old-age patients with genetic frontotemporal lobar degeneration (gFTLD-TDP) and compare these patients with sporadic old-age individuals with TDP-43, either in the presence of Alzheimer's disease (AD-TDP) or in isolation (pure-TDP). METHODS The brain bank at Mayo Clinic-Jacksonville was searched for cases ≥75 years old at death with TDP-43 extending into middle frontal cortex. Cases were split into the following groups: (1) gFTLD-TDP (n = 15) with progranulin (GRN)/C9ORF72 mutations; (2) AD-TDP (n = 10)-cases with median Braak neurofibrillary tangle (NFT) stage VI, Thal phase V; (3) pure-TDP (n = 10)-cases with median Braak NFT stage I, Thal phase I. Clinical data were abstracted; TDP-43 burden was calculated using digital pathology. RESULTS Amnestic Alzheimer's dementia was the clinical diagnosis in ≥50% patients in each group. The distribution of TDP-43 burden in gFTLD-TDP and AD-TDP, but not pure-TDP, was limbic-predominant targeting CA1 and subiculum. Patients with gFTLD-TDP had higher burden in entorhinal cortex compared to AD-TDP. TDP-43 burden in middle frontal cortex did not differ between the three groups. CONCLUSIONS In old age it is challenging to clinically and pathologically differentiate gFTLD-TDP from AD-TDP and pure-TDP-43 based on burden. Like AD-TDP, old age gFTLD-TDP have a limbic predominant TDP-43 distribution. The finding that amnestic Alzheimer's dementia was the most common clinical diagnosis regardless of group suggests that TDP-43 directly and indirectly targets limbic regions.
Collapse
Affiliation(s)
- Marina Buciuc
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Matthew C Baker
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | |
Collapse
|
31
|
Gallucci M, Mazzarolo AP, Focella L, Berlin E, Fiore V, Di Paola F, Bendini M, Zanusso G, Fenoglio C, Galimberti D, Bonanni L. More Atypical than Atypical Alzheimer's Disease Phenotypes: A Treviso Dementia (TREDEM) Registry Case Report. J Alzheimers Dis Rep 2021; 5:365-374. [PMID: 34189408 PMCID: PMC8203287 DOI: 10.3233/adr-210009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND A 57-year-old right-handed man was admitted to the Treviso Memory Clinic due to the presence of memory forgetfulness, repetition of the same questions, episodes of confusion, initial difficulties in performing complex tasks and easy distraction over the past two years, as well as recurrent and never-happened-before car accidents. OBJECTIVE We report a peculiar case of an early onset Alzheimer's disease (AD) with an unusual symptomatology, apparently not fitting in any of the categorized atypical forms of AD nor being representative of a typical amnestic AD. METHODS The patient underwent a neuropsychological, structural, and metabolic cerebral evaluation by MRI and 18F-FDG PET, together with the search for cerebral amyloid (amyloid PET), a genetic testing for dementia related genes and the dosage of CSF protein biomarkers of neurodegenerative conditions. RESULTS We observed a convergence of predominant frontal (dysexecutive, verbal disinhibition) and posterior (visuospatial) features of cognitive impairment. Structural MRI sequences showed subarachnoid spaces of the vault enlarged in the fronto-parietal region with anterior and posterior cortical atrophy. The hippocampus appeared preserved. The 18F-FDG PET scans showed hypometabolism in the prefrontal, lateral temporal, posterior parietal, and occipital regions bilaterally. The 18F-Flutemetamol scan showed a diffused uptake of the amyloid tracer at the cerebral cortex. CSF biomarkers were compatible with Alzheimer's disease (AD). CONCLUSION This case report presented with clinical phenotypic aspects atypical of AD, both frontal and posterior, never described as concomitant in the most accredited criteria for atypical AD, and appeared therefore more atypical than each of the atypical AD phenotypes already reported.
Collapse
Affiliation(s)
- Maurizio Gallucci
- Cognitive Impairment Center, Local Health Authority n.2 Marca Trevigiana, Treviso, Italy
- Associazione Alzheimer Treviso Onlus, Treviso, Italy
| | - Anna Paola Mazzarolo
- Cognitive Impairment Center, Local Health Authority n.2 Marca Trevigiana, Treviso, Italy
| | - Lucia Focella
- Cognitive Impairment Center, Local Health Authority n.2 Marca Trevigiana, Treviso, Italy
| | - Elisa Berlin
- Cognitive Impairment Center, Local Health Authority n.2 Marca Trevigiana, Treviso, Italy
| | - Vittorio Fiore
- Nuclear Medicine Unit, Local Health Authority n.2 Marca Trevigiana, Treviso, Italy
| | - Francesco Di Paola
- Neuroradiology Unit, Local Health Authority n.2 Marca Trevigiana, Treviso, Italy
| | - Matteo Bendini
- Neuroradiology Unit, Local Health Authority n.2 Marca Trevigiana, Treviso, Italy
| | - Gianluigi Zanusso
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | | | - Daniela Galimberti
- University of Milan, Dino Ferrari Center, Milan, Italy
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, Neurodegenerative Disease Unit, Milan, Italy
| | - Laura Bonanni
- Department of Neuroscience Imaging and Clinical Sciences and CESI, University G D’Annunzio of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
32
|
Gefen T, Mao Q, Kohler M, Moeller S, Kawles A, Coventry C, Spencer C, Lilek J, Ajroud K, Feldman A, Flanagan M, Rogalski E, Weintraub S, Rademaker A, Geula C, Mesulam MM, Bigio E. Primary Progressive Aphasia has a Unique Signature Distinct from Dementia of the Alzheimer's Type and Behavioral Variant Frontotemporal Dementia Regardless of Pathology. J Neuropathol Exp Neurol 2021; 79:1379-1381. [PMID: 33068393 DOI: 10.1093/jnen/nlaa080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Qinwen Mao
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Missia Kohler
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Stacey Moeller
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Allegra Kawles
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Callen Spencer
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jaclyn Lilek
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Kaouther Ajroud
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Alex Feldman
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Margaret Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Alfred Rademaker
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - M-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Eileen Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
33
|
Gefen T, Kawles A, Makowski-Woidan B, Engelmeyer J, Ayala I, Abbassian P, Zhang H, Weintraub S, Flanagan ME, Mao Q, Bigio EH, Rogalski E, Mesulam MM, Geula C. Paucity of Entorhinal Cortex Pathology of the Alzheimer's Type in SuperAgers with Superior Memory Performance. Cereb Cortex 2021; 31:3177-3183. [PMID: 33592091 DOI: 10.1093/cercor/bhaa409] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Advancing age is typically associated with declining memory capacity and increased risk of Alzheimer's disease (AD). Markers of AD such as amyloid plaques (AP) and neurofibrillary tangles (NFTs) are commonly found in the brains of cognitively average elderly but in more limited distribution than in those at the mild cognitive impairment and dementia stages of AD. Cognitive SuperAgers are individuals over age 80 who show superior memory capacity, at a level consistent with individuals 20-30 years their junior. Using a stereological approach, the current study quantitated the presence of AD markers in the memory-associated entorhinal cortex (ERC) of seven SuperAgers compared with six age-matched cognitively average normal control individuals. Amyloid plaques and NFTs were visualized using Thioflavin-S histofluorescence, 6E10, and PHF-1 immunohistochemistry. Unbiased stereological analysis revealed significantly more NFTs in ERC in cognitively average normal controls compared with SuperAgers (P < 0.05) by a difference of ~3-fold. There were no significant differences in plaque density. To highlight relative magnitude, cases with typical amnestic dementia of AD showed nearly 100 times more entorhinal NFTs than SuperAgers. The results suggest that resistance to age-related neurofibrillary degeneration in the ERC may be one factor contributing to preserved memory in SuperAgers.
Collapse
Affiliation(s)
- Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Allegra Kawles
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Beth Makowski-Woidan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Janessa Engelmeyer
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ivan Ayala
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Payam Abbassian
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hui Zhang
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qinwen Mao
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Eileen H Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - M Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
34
|
Mesulam MM, Coventry C, Kuang A, Bigio EH, Mao Q, Flanagan ME, Gefen T, Sridhar J, Geula C, Zhang H, Weintraub S, Rogalski EJ. Memory Resilience in Alzheimer Disease With Primary Progressive Aphasia. Neurology 2021; 96:e916-e925. [PMID: 33441454 PMCID: PMC8105903 DOI: 10.1212/wnl.0000000000011397] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/25/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether memory is preserved longitudinally in primary progressive aphasia (PPA) associated with Alzheimer disease (AD) and to identify potential factors that maintain memory despite underlying neurofibrillary degeneration of mediotemporal memory areas. METHODS Longitudinal memory assessment was done in 17 patients with PPA with autopsy or biomarker evidence of AD (PPA-AD) and 14 patients with amnestic dementia of the Alzheimer type with AD at autopsy (DAT-AD). RESULTS In PPA-AD, episodic memory, tested with nonverbal items, was preserved at the initial testing and showed no decline at retesting 2.35 ± 0.78 years later, at which time symptoms had been present for 6.26 ± 2.21 years. In contrast, language functions declined significantly during the same period. In DAT-AD, both verbal memory and language declined with equal severity. Although imaging showed asymmetric left-sided mediotemporal atrophy in PPA-AD, autopsy revealed bilateral hippocampo-entorhinal neurofibrillary degeneration at Braak stages V and VI. Compared to DAT-AD, however, the PPA-AD group had lower incidence of APOE ε4 and of mediotemporal TAR DNA-binding protein 43 (TDP-43) pathology. CONCLUSIONS Memory preservation in PPA is not just an incidental finding at onset but a core feature that persists for years despite the hippocampo-entorhinal AD neuropathology that is as severe as that of DAT-AD. Asymmetry of mediotemporal atrophy and a lesser impact of APOE ε4 and of TDP-43 on the integrity of memory circuitry may constitute some of the factors underlying this resilience. Our results also suggest that current controversies on memory in PPA-AD reflect inconsistencies in the diagnosis of logopenic PPA, the clinical variant most frequently associated with AD. CLINICALTRIALSGOV IDENTIFIER NCT00537004 and NCT03371706.
Collapse
Affiliation(s)
- M-Marsel Mesulam
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL.
| | - Christina Coventry
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| | - Alan Kuang
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| | - Eileen H Bigio
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| | - Qinwen Mao
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| | - Margaret E Flanagan
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| | - Tamar Gefen
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| | - Jaiashre Sridhar
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| | - Changiz Geula
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| | - Hui Zhang
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| | - Sandra Weintraub
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| | - Emily J Rogalski
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (M.-M.M., C.C., E.H.B., Q.M., M.E.F., T.G., J.S., C.G., H.Z., S.W., E.J.R.), Department of Preventive Medicine (A.K., H.Z.), Department of Pathology (E.H.B., Q.M., M.E.F.), and Department of Psychiatry and Behavioral Sciences (T.G., S.W., E.J.R.), Northwestern University Feinberg School of Medicine, Chicago. IL
| |
Collapse
|
35
|
Ohm DT, Fought AJ, Martersteck A, Coventry C, Sridhar J, Gefen T, Weintraub S, Bigio E, Mesulam M, Rogalski E, Geula C. Accumulation of neurofibrillary tangles and activated microglia is associated with lower neuron densities in the aphasic variant of Alzheimer's disease. Brain Pathol 2021; 31:189-204. [PMID: 33010092 PMCID: PMC7855834 DOI: 10.1111/bpa.12902] [Citation(s) in RCA: 42] [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: 06/17/2020] [Revised: 08/27/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
The neurofibrillary tangles (NFT) and amyloid-ß plaques (AP) that comprise Alzheimer's disease (AD) neuropathology are associated with neurodegeneration and microglial activation. Activated microglia exist on a dynamic spectrum of morphologic subtypes that include resting, surveillant microglia capable of converting to activated, hypertrophic microglia closely linked to neuroinflammatory processes and AD neuropathology in amnestic AD. However, quantitative analyses of microglial subtypes and neurons are lacking in non-amnestic clinical AD variants, including primary progressive aphasia (PPA-AD). PPA-AD is a language disorder characterized by cortical atrophy and NFT densities concentrated to the language-dominant hemisphere. Here, a stereologic investigation of five PPA-AD participants determined the densities and distributions of neurons and microglial subtypes to examine how cellular changes relate to AD neuropathology and may contribute to cortical atrophy. Adjacent series of sections were immunostained for neurons (NeuN) and microglia (HLA-DR) from bilateral language and non-language regions where in vivo cortical atrophy and Thioflavin-S-positive APs and NFTs were previously quantified. NeuN-positive neurons and morphologic subtypes of HLA-DR-positive microglia (i.e., resting [ramified] microglia and activated [hypertrophic] microglia) were quantified using unbiased stereology. Relationships between neurons, microglia, AD neuropathology, and cortical atrophy were determined using linear mixed models. NFT densities were positively associated with hypertrophic microglia densities (P < 0.01) and inversely related to neuron densities (P = 0.01). Hypertrophic microglia densities were inversely related to densities of neurons (P < 0.01) and ramified microglia (P < 0.01). Ramified microglia densities were positively associated with neuron densities (P = 0.02) and inversely related to cortical atrophy (P = 0.03). Our findings provide converging evidence of divergent roles for microglial subtypes in patterns of neurodegeneration, which includes hypertrophic microglia likely driving a neuroinflammatory response more sensitive to NFTs than APs in PPA-AD. Moreover, the accumulation of both NFTs and activated hypertrophic microglia in association with low neuron densities suggest they may collectively contribute to focal neurodegeneration characteristic of PPA-AD.
Collapse
Affiliation(s)
- Daniel T. Ohm
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Angela J. Fought
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIL
| | - Adam Martersteck
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Jaiashre Sridhar
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIL
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIL
| | - Eileen Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoIL
| | - M.‐Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIL
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIL
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| |
Collapse
|
36
|
Mesulam MM, Coventry C, Bigio EH, Geula C, Thompson C, Bonakdarpour B, Gefen T, Rogalski EJ, Weintraub S. Nosology of Primary Progressive Aphasia and the Neuropathology of Language. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:33-49. [PMID: 33433867 PMCID: PMC8103786 DOI: 10.1007/978-3-030-51140-1_3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Primary progressive aphasia (PPA) is a dementia syndrome associated with several neuropathologic entities, including Alzheimer's disease (AD) and all major forms of frontotemporal lobar degeneration (FTLD). It is classified into subtypes defined by the nature of the language domain that is most impaired. The asymmetric neurodegeneration of the hemisphere dominant for language (usually left) is one consistent feature of all PPA variants. This feature offers unique opportunities for exploring mechanisms of selective vulnerability in neurodegenerative diseases and the neuroanatomy of language. This chapter reviews some of the current trends in PPA research as well as the challenges that remain to be addressed on the nosology, clinicopathologic correlations, and therapy of this syndrome.
Collapse
Affiliation(s)
- M -Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease; Department of Neurology, Northwestern University, Chicago, IL, USA.
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Eileen H Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease; Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Cynthia Thompson
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease; Department of Communication Sciences and Disorders; Department of Neurology, Northwestern University, Evanston, IL, USA
| | - Borna Bonakdarpour
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease; Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease; Department of Psychiatry, Northwestern University, Chicago, IL, USA
| | - Emily J Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease; Department of Psychiatry, Northwestern University, Chicago, IL, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| |
Collapse
|
37
|
Wood JL, Weintraub S, Coventry C, Xu J, Zhang H, Rogalski E, Mesulam MM, Gefen T. Montreal Cognitive Assessment (MoCA) Performance and Domain-Specific Index Scores in Amnestic Versus Aphasic Dementia. J Int Neuropsychol Soc 2020; 26:927-931. [PMID: 32423505 PMCID: PMC7554137 DOI: 10.1017/s135561772000048x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE The Montreal Cognitive Assessment (MoCA) is a popular and simple-to-administer screening instrument to detect cognitive impairment. The MoCA generates a total score and six domain-specific index scores: (1) Memory, (2) Executive Functioning, (3) Attention, (4) Language, (5) Visuospatial, and (6) Orientation. It is unclear whether these MoCA scores can differentiate between distinct clinical dementia syndromes. This study compared MoCA Index scores between amnestic dementia of the Alzheimer's type (DAT) and primary progressive aphasia (PPA), a language-based dementia. METHOD Baseline MoCA data were analyzed from 33 DAT, 37 PPA, and 83 cognitively normal individuals enrolled in the Clinical Core of the Northwestern Alzheimer's Disease Center. A one-way analysis of covariance adjusted for age was used to compare MoCA scores among groups. A logistic regression model was implemented to observe individual likelihood of group affiliation based on MoCA Index scores. RESULTS The mean MoCA total score was significantly higher in controls compared to both patient groups (p < .001) but did not differ between DAT and PPA groups. However, in accordance with salient clinical features commonly observed in DAT versus PPA, Memory and Orientation Index scores were lowest in the DAT group (p < .001), whereas Language and Attention Index scores were lowest in the PPA group (p < .001). Multivariate logistic regression analysis showed that the individual effects of Memory (p = .001), Language (p = .002), and Orientation (p = .025) Indices were significant. CONCLUSIONS MoCA Index scores can help differentiate among distinct cognitive syndromes, suggesting it may be a useful brief screening tool to detect domain-specific cognitive impairment.
Collapse
Affiliation(s)
- Jessica L. Wood
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Jiahui Xu
- Division of Biostatistics, Department of Preventive Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
- Quantitative Data Sciences Core (QDSC), Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Hui Zhang
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
- Division of Biostatistics, Department of Preventive Medicine, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
- Quantitative Data Sciences Core (QDSC), Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Marek-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
- Department of Neurology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
38
|
Spina S, Brown JA, Deng J, Gardner RC, Nana AL, Hwang JHL, Gaus SE, Huang EJ, Kramer JH, Rosen HJ, Kornak J, Neuhaus J, Miller BL, Grinberg LT, Boxer AL, Seeley WW. Neuropathological correlates of structural and functional imaging biomarkers in 4-repeat tauopathies. Brain 2020; 142:2068-2081. [PMID: 31081015 DOI: 10.1093/brain/awz122] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 02/23/2019] [Accepted: 03/07/2019] [Indexed: 12/24/2022] Open
Abstract
Neurodegenerative dementia syndromes are characterized by spreading of pathological protein deposition along syndrome-specific neural networks. Structural and functional MRI measures can assess the integrity of these networks and have been proposed as biomarkers of disease progression for clinical trials. The relationship between in vivo imaging measures and pathological features, at the single subject level, remains largely unknown. Patient-specific maps of atrophy and seed-based intrinsic connectivity disruption, as compared to normal controls, were obtained for 27 patients subsequently diagnosed with progressive supranuclear palsy (n = 16, seven males, age at death 68.9 ± 6.0 years, imaging-to-pathology interval = 670.2 ± 425.1 days) or corticobasal degeneration (n = 11, two males, age at death 66.7 ± 5.4 years, imaging-to-pathology interval = 696.2 ± 482.2 days). A linear mixed effect model with crossed random effects was used to test regional and single-subject level associations between post-mortem regional measures of neurodegeneration and tau inclusion burden, on the one hand, and regional volume loss and seed-based intrinsic connectivity reduction, on the other. A significant association was found between tau inclusion burden and in vivo volume loss, at the regional level and independent of neurodegeneration severity, in both progressive supranuclear palsy [n = 340 regions; beta 0.036; 95% confidence interval (CI): 0.001, 0.072; P = 0.046] and corticobasal degeneration (n = 215 regions; beta 0.044; 95% CI: 0.009, 0.079; P = 0.013). We also found a significant association between post-mortem neurodegeneration and in vivo volume loss in both progressive supranuclear palsy (n = 340 regions; beta 0.155; 95% CI: 0.061, 0.248; P = 0.001) and corticobasal degeneration (n = 215 regions; beta 0.277; 95% CI: 0.104, 0.450; P = 0.002). We found a significant association between regional neurodegeneration and intrinsic connectivity dysfunction in corticobasal degeneration (n = 215 regions; beta 0.074; 95% CI: 0.005, 0.143; P = 0.035), but no other associations between post-mortem measures of tauopathy and intrinsic connectivity dysfunction reached statistical significance. Our data suggest that in vivo structural imaging measures reflect independent contributions from neurodegeneration and tau burden in progressive supranuclear palsy and corticobasal degeneration. Seed-based measures of intrinsic connectivity dysfunction showed less reliable predictive value when used as in vivo biomarkers of tauopathy. The findings provide important guidance for the use of imaging biomarkers as indirect in vivo assays of microscopic pathology.
Collapse
Affiliation(s)
- Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - Jesse A Brown
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - Jersey Deng
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - Raquel C Gardner
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - Alissa L Nana
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - Ji-Hye L Hwang
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - Stephanie E Gaus
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - Eric J Huang
- Department of Pathology, University of California San Francisco, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - Howie J Rosen
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - John Kornak
- Department of Epidemiology and Biostatistics, University of California San Francisco, USA
| | - John Neuhaus
- Department of Epidemiology and Biostatistics, University of California San Francisco, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA.,Department of Pathology, University of California San Francisco, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California San Francisco, USA.,Department of Pathology, University of California San Francisco, USA
| |
Collapse
|
39
|
Kim KW, Park S, Jo H, Cho SH, Kim SJ, Kim Y, Jang H, Na DL, Seo SW, Kim HJ. Identifying a subtype of Alzheimer's disease characterised by predominant right focal cortical atrophy. Sci Rep 2020; 10:7256. [PMID: 32350336 PMCID: PMC7190862 DOI: 10.1038/s41598-020-64180-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 04/09/2020] [Indexed: 11/23/2022] Open
Abstract
We aimed to identify an Alzheimer’s disease (AD) subtype with right predominant focal atrophy. We recruited 17 amyloid PET positive logopenic variant primary progressive aphasia (lvPPA) and 226 amyloid PET positive AD patients. To identify AD with right focal atrophy (Rt-AD), we selected cortical areas that showed more atrophy in lvPPA than in AD and calculated an asymmetry index (AI) for this area in each individual. Using a receiver operating characteristic curve, we found that the optimal AI cut-off to discriminate lvPPA from AD was −3.1 (mean AI – 1.00 standard deviation) (sensitivity 88.2, specificity 89.8). We identified 32 Rt-AD patients whose AI was above mean AI + 1.00 standard deviation, 38 Lt-AD patients whose AI was lower than mean AI − 1.00 standard deviation, and 173 Symmetric-AD patients whose AI was within mean AI ± 1.00 standard deviation. We characterized clinical and cognitive profiles of Rt-AD patients by comparing with those of Lt-AD and Symmetric-AD patients. Compared to Symmetric-AD patients, Rt-AD patients had asymmetric focal atrophy in the right temporoparietal area and showed poor performance on visuospatial function testing (p = 0.009). Our findings suggested that there is an AD variant characterized by right focal atrophy and visuospatial dysfunction.
Collapse
Affiliation(s)
- Ko Woon Kim
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurology, Jeonbuk National University Medical School & Hospital, Jeonju, Korea.,Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, Korea.,Biomedical Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Seongbeom Park
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Korea
| | - Hyunjin Jo
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Soo Hyun Cho
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurology, Chonnam National University Hospital, Gwangju, Korea
| | - Seung Joo Kim
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurology, Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon, Korea
| | - Yeshin Kim
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea.,Department of Neurology, Kangwon National University Hospital, Kangwon National University College of Medicine, Chuncheon, Korea
| | - Hyemin Jang
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Korea
| | - Duk L Na
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Sang Won Seo
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Hee Jin Kim
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea. .,Neuroscience Center, Samsung Medical Center, Seoul, Korea. .,Samsung Alzheimer Research Center, Samsung Medical Center, Seoul, Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea. .,Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Korea.
| |
Collapse
|
40
|
Weintraub S, Teylan M, Rader B, Chan KCG, Bollenbeck M, Kukull WA, Coventry C, Rogalski E, Bigio E, Mesulam MM. APOE is a correlate of phenotypic heterogeneity in Alzheimer disease in a national cohort. Neurology 2020; 94:e607-e612. [PMID: 31704790 PMCID: PMC7136069 DOI: 10.1212/wnl.0000000000008666] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 08/15/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To compare the proportion of APOE ε4 genotype carriers in aphasic vs amnestic variants of Alzheimer disease (AD). METHOD The proportion of APOE ε4 carriers was compared among the following 3 groups: (1) 42 patients with primary progressive aphasia (PPA) and AD pathology (PPA/AD) enrolled in the Northwestern Alzheimer Disease Center Clinical Core; (2) 1,418 patients with autopsy-confirmed AD and amnestic dementia of the Alzheimer type (DAT/AD); and (3) 2,608 cognitively normal controls (NC). The latter 2 groups were compiled from the National Alzheimer Coordinating Center database. Logistic regression models analyzed the relationship between groups and APOE ε4 carrier status, adjusting for age at onset and sex as needed. RESULTS Using NC as the reference and adjusting for sex and age, the DAT/AD group was 3.97 times more likely to be APOE ε4 carriers. Adjusting for sex and age at symptom onset, the DAT/AD group was 2.46 times as likely to be carriers compared to PPA/AD. There was no significant difference in the proportion of APOE ε4 carriers for PPA/AD compared to NC. PPA subtypes included 24 logopenic, 10 agrammatic nonfluent, and 8 either mixed (n = 5) or too severe (n = 3) to subtype. The proportion of carriers and noncarriers was similar for logopenic and agrammatic subtypes, both having fewer carriers. CONCLUSION The proportion of APOE ε4 carriers was elevated in amnestic but not aphasic manifestations of AD. These results suggest that APOE ε4 is an anatomically selective risk factor that preferentially increases the vulnerability to AD pathology of memory-related medial temporal areas rather than language-related neocortices.
Collapse
Affiliation(s)
- Sandra Weintraub
- From Mesulam Center for Cognitive Neurology and Alzheimer's Disease (S.W., B.R., C.C., E.R., E.B., M.-M.M.) and the Departments of Psychiatry and Behavioral Sciences (S.W., E.R.), Pathology (E.B.), and Neurology (M.-M.M.), Northwestern Feinberg School of Medicine, Chicago, IL; and Department of Epidemiology, National Alzheimer Coordinating Center (M.T., M.B., W.A.K.), and Department of Biostatistics (K.C.G.C.), University of Washington, Seattle.
| | - Merilee Teylan
- From Mesulam Center for Cognitive Neurology and Alzheimer's Disease (S.W., B.R., C.C., E.R., E.B., M.-M.M.) and the Departments of Psychiatry and Behavioral Sciences (S.W., E.R.), Pathology (E.B.), and Neurology (M.-M.M.), Northwestern Feinberg School of Medicine, Chicago, IL; and Department of Epidemiology, National Alzheimer Coordinating Center (M.T., M.B., W.A.K.), and Department of Biostatistics (K.C.G.C.), University of Washington, Seattle
| | - Benjamin Rader
- From Mesulam Center for Cognitive Neurology and Alzheimer's Disease (S.W., B.R., C.C., E.R., E.B., M.-M.M.) and the Departments of Psychiatry and Behavioral Sciences (S.W., E.R.), Pathology (E.B.), and Neurology (M.-M.M.), Northwestern Feinberg School of Medicine, Chicago, IL; and Department of Epidemiology, National Alzheimer Coordinating Center (M.T., M.B., W.A.K.), and Department of Biostatistics (K.C.G.C.), University of Washington, Seattle
| | - Kwun C G Chan
- From Mesulam Center for Cognitive Neurology and Alzheimer's Disease (S.W., B.R., C.C., E.R., E.B., M.-M.M.) and the Departments of Psychiatry and Behavioral Sciences (S.W., E.R.), Pathology (E.B.), and Neurology (M.-M.M.), Northwestern Feinberg School of Medicine, Chicago, IL; and Department of Epidemiology, National Alzheimer Coordinating Center (M.T., M.B., W.A.K.), and Department of Biostatistics (K.C.G.C.), University of Washington, Seattle
| | - Mark Bollenbeck
- From Mesulam Center for Cognitive Neurology and Alzheimer's Disease (S.W., B.R., C.C., E.R., E.B., M.-M.M.) and the Departments of Psychiatry and Behavioral Sciences (S.W., E.R.), Pathology (E.B.), and Neurology (M.-M.M.), Northwestern Feinberg School of Medicine, Chicago, IL; and Department of Epidemiology, National Alzheimer Coordinating Center (M.T., M.B., W.A.K.), and Department of Biostatistics (K.C.G.C.), University of Washington, Seattle
| | - Walter A Kukull
- From Mesulam Center for Cognitive Neurology and Alzheimer's Disease (S.W., B.R., C.C., E.R., E.B., M.-M.M.) and the Departments of Psychiatry and Behavioral Sciences (S.W., E.R.), Pathology (E.B.), and Neurology (M.-M.M.), Northwestern Feinberg School of Medicine, Chicago, IL; and Department of Epidemiology, National Alzheimer Coordinating Center (M.T., M.B., W.A.K.), and Department of Biostatistics (K.C.G.C.), University of Washington, Seattle
| | - Christina Coventry
- From Mesulam Center for Cognitive Neurology and Alzheimer's Disease (S.W., B.R., C.C., E.R., E.B., M.-M.M.) and the Departments of Psychiatry and Behavioral Sciences (S.W., E.R.), Pathology (E.B.), and Neurology (M.-M.M.), Northwestern Feinberg School of Medicine, Chicago, IL; and Department of Epidemiology, National Alzheimer Coordinating Center (M.T., M.B., W.A.K.), and Department of Biostatistics (K.C.G.C.), University of Washington, Seattle
| | - Emily Rogalski
- From Mesulam Center for Cognitive Neurology and Alzheimer's Disease (S.W., B.R., C.C., E.R., E.B., M.-M.M.) and the Departments of Psychiatry and Behavioral Sciences (S.W., E.R.), Pathology (E.B.), and Neurology (M.-M.M.), Northwestern Feinberg School of Medicine, Chicago, IL; and Department of Epidemiology, National Alzheimer Coordinating Center (M.T., M.B., W.A.K.), and Department of Biostatistics (K.C.G.C.), University of Washington, Seattle
| | - Eileen Bigio
- From Mesulam Center for Cognitive Neurology and Alzheimer's Disease (S.W., B.R., C.C., E.R., E.B., M.-M.M.) and the Departments of Psychiatry and Behavioral Sciences (S.W., E.R.), Pathology (E.B.), and Neurology (M.-M.M.), Northwestern Feinberg School of Medicine, Chicago, IL; and Department of Epidemiology, National Alzheimer Coordinating Center (M.T., M.B., W.A.K.), and Department of Biostatistics (K.C.G.C.), University of Washington, Seattle
| | - M-Marsel Mesulam
- From Mesulam Center for Cognitive Neurology and Alzheimer's Disease (S.W., B.R., C.C., E.R., E.B., M.-M.M.) and the Departments of Psychiatry and Behavioral Sciences (S.W., E.R.), Pathology (E.B.), and Neurology (M.-M.M.), Northwestern Feinberg School of Medicine, Chicago, IL; and Department of Epidemiology, National Alzheimer Coordinating Center (M.T., M.B., W.A.K.), and Department of Biostatistics (K.C.G.C.), University of Washington, Seattle
| |
Collapse
|
41
|
Resende EDPF, Nolan AL, Petersen C, Ehrenberg AJ, Spina S, Allen IE, Rosen HJ, Kramer J, Miller BL, Seeley WW, Gorno-Tempini ML, Miller Z, Grinberg LT. Language and spatial dysfunction in Alzheimer disease with white matter thorn-shaped astrocytes. Neurology 2020; 94:e1353-e1364. [PMID: 32001514 DOI: 10.1212/wnl.0000000000008937] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Alzheimer disease (AD) shows a broad array of clinical presentations, but the mechanisms underlying these phenotypic variants remain elusive. Aging-related astrogliopathy (ARTAG) is a relatively recent term encompassing a broad array of tau deposition in astroglia outside the range of traditional tauopathies. White matter thorn-shaped astrocyte (WM-TSA) clusters, a specific ARTAG subtype, has been associated with atypical language presentation of AD in a small study lacking replication. To interrogate the impact of WM-TSA in modifying clinical phenotype in AD, we investigated a clinicopathologic sample of 83 persons with pure cortical AD pathology and heterogeneous clinical presentations. METHODS We mapped WM-TSA presence and density throughout cortical areas and interrogated whether WM-TSA correlated with atypical AD presentation or worse performance in neuropsychological testing. RESULTS WM-TSA was present in nearly half of the cases and equally distributed in typical and atypical AD presentations. Worsening language and visuospatial functions were correlated with higher WM-TSA density in language-related and visuospatial-related regions, respectively. These findings were unrelated to regional neurofibrillary tangle burden. Next, unsupervised clustering divided the participants into 2 groups: a high-WM-TSA (n = 9) and low-WM-TSA (n = 74) pathology signature. The high-WM-TSA group scored significantly worse in language but not in other cognitive domains. CONCLUSIONS The negative impact of WM-TSA pathology to language and possibly visuospatial networks suggests that WM-TSA is not as benign as other ARTAG types and may be explored as a framework to understand the mechanisms and impact of astrocytic tau deposition in AD in humans.
Collapse
Affiliation(s)
- Elisa de Paula França Resende
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Amber L Nolan
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Cathrine Petersen
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Alexander J Ehrenberg
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Salvatore Spina
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Isabel E Allen
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Howard J Rosen
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Joel Kramer
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Bruce L Miller
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - William W Seeley
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Maria Luiza Gorno-Tempini
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Zachary Miller
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Lea T Grinberg
- From the Memory and Aging Center (E.d.P.F.R., A.L.N., C.P., A.J.E., S.S., I.E.A., H.J.R., J.K., B.L.M., W.W.S., M.L.G.-T., Z.M., L.T.G.), Weill Institute for Neurosciences, and Department of Biostatistics and Epidemiology (L.T.G.), University of California, San Francisco; Global Brain Health Institute based at University of California (E.d.P.F.R., L.T.G.), San Francisco; Trinity College (E.d.P.F.R., L.T.G.), Dublin, Ireland; Department of Neurology (E.d.P.F.R.), Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Integrative Biology (A.J.E.), University of California, Berkeley; and Department of Pathology (L.T.G.), Lim-22, Lim-66, University of Sao Paulo Medical School, Sao Paulo, Brazil.
| |
Collapse
|
42
|
Martersteck A, Sridhar J, Rader B, Coventry C, Parrish T, Mesulam MM, Rogalski E. Differential neurocognitive network perturbation in amnestic and aphasic Alzheimer disease. Neurology 2020; 94:e699-e704. [PMID: 31969462 DOI: 10.1212/wnl.0000000000008960] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 08/20/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine if Alzheimer disease (AD) is associated with aphasic rather than amnestic dementias in certain circumstances related in part to perturbations in different networks. METHODS Three groups were investigated: 14 participants suspected of having the neuropathology of AD based on clinically diagnosed amnestic dementia of the Alzheimer type (DAT), 26 individuals with primary progressive aphasia (PPA) with either a positive 18F-florbetapir amyloid PET scan or confirmed AD at autopsy, and 26 neurologically intact controls. The groups were compared using rs-fMRI. Seeds included the left hemisphere inferior frontal gyrus (IFG) for the language network, the left hippocampus for the episodic memory network, and the left posterior cingulate for the default mode network (DMN). RESULTS Greater connectivity perturbations were found from the hippocampus for the DAT group and from the IFG for the PPA group. Furthermore, connectivity alterations in the PPA group were more asymmetric and favored the language-dominant left hemisphere. Loss of connectivity from the DMN seed was of a similar magnitude in the PPA and DAT groups. CONCLUSIONS Despite the presumptive common underlying neuropathology of amyloid plaques and neurofibrillary tangles, the 2 groups displayed 2 different patterns of network perturbation, each concordant with the clinical presentation and the anatomy of neurodegeneration.
Collapse
Affiliation(s)
- Adam Martersteck
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (A.M., J.S., B.R., C.C., M.-M.M., E.R.) and Departments of Radiology (A.M., T.P.), Neurology (M.-M.M.), and Psychiatry and Behavioral Sciences (E.R.), Northwestern University Feinberg School of Medicine, Chicago, IL.
| | - Jaiashre Sridhar
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (A.M., J.S., B.R., C.C., M.-M.M., E.R.) and Departments of Radiology (A.M., T.P.), Neurology (M.-M.M.), and Psychiatry and Behavioral Sciences (E.R.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Benjamin Rader
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (A.M., J.S., B.R., C.C., M.-M.M., E.R.) and Departments of Radiology (A.M., T.P.), Neurology (M.-M.M.), and Psychiatry and Behavioral Sciences (E.R.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Christina Coventry
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (A.M., J.S., B.R., C.C., M.-M.M., E.R.) and Departments of Radiology (A.M., T.P.), Neurology (M.-M.M.), and Psychiatry and Behavioral Sciences (E.R.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Todd Parrish
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (A.M., J.S., B.R., C.C., M.-M.M., E.R.) and Departments of Radiology (A.M., T.P.), Neurology (M.-M.M.), and Psychiatry and Behavioral Sciences (E.R.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - M-Marsel Mesulam
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (A.M., J.S., B.R., C.C., M.-M.M., E.R.) and Departments of Radiology (A.M., T.P.), Neurology (M.-M.M.), and Psychiatry and Behavioral Sciences (E.R.), Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Emily Rogalski
- From the Mesulam Center for Cognitive Neurology and Alzheimer's Disease (A.M., J.S., B.R., C.C., M.-M.M., E.R.) and Departments of Radiology (A.M., T.P.), Neurology (M.-M.M.), and Psychiatry and Behavioral Sciences (E.R.), Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
43
|
Llibre-Guerra JJ, Li Y, Schindler SE, Gordon BA, Fagan AM, Morris JC, Benzinger TLS, Hassenstab J, Wang G, Allegri R, Berman SB, Chhatwal J, Farlow MR, Holtzman DM, Jucker M, Levin J, Noble JM, Salloway S, Schofield P, Karch C, Fox NC, Xiong C, Bateman RJ, McDade E. Association of Longitudinal Changes in Cerebrospinal Fluid Total Tau and Phosphorylated Tau 181 and Brain Atrophy With Disease Progression in Patients With Alzheimer Disease. JAMA Netw Open 2019; 2:e1917126. [PMID: 31825500 PMCID: PMC6991202 DOI: 10.1001/jamanetworkopen.2019.17126] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IMPORTANCE The amyloid/tau/neurodegeneration (A/T/N) framework uses cerebrospinal fluid (CSF) levels of total tau (tTau) as a marker of neurodegeneration and CSF levels of phosphorylated tau 181 (pTau181) as a marker of tau tangles. However, it is unclear whether CSF levels of tTau and pTau181 have similar or different trajectories over the course of Alzheimer disease. OBJECTIVES To examine the rates of change in CSF levels of tTau and pTau181 across the Alzheimer disease course and how the rates of change are associated with brain atrophy as measured by magnetic resonance imaging. DESIGN, SETTING, AND PARTICIPANTS This cohort study was set in tertiary research clinics. Each participant was a member of a pedigree with a known mutation for dominantly inherited Alzheimer disease. Participants were divided into 3 groups on the basis of the presence of a mutation and their Clinical Dementia Rating score. Data analysis was performed in June 2019. MAIN OUTCOMES AND MEASURES Rates of change of CSF tTau and pTau181 levels and their association with the rate of change of brain volume. RESULTS Data from 465 participants (283 mutation carriers and 182 noncarriers) were analyzed. The mean (SD) age of the cohort was 37.8 (11.3) years, and 262 (56.3%) were women. The mean (SD) follow-up duration was 2.7 (1.5) years. Two or more longitudinal CSF and magnetic resonance imaging assessments were available for 160 and 247 participants, respectively. Sixty-five percent of mutation carriers (183) did not have symptoms at baseline (Clinical Dementia Rating score, 0). For mutation carriers, the annual rates of change for CSF tTau and pTau181 became significantly different from 0 approximately 10 years before the estimated year of onset (mean [SE] rates of change, 5.5 [2.8] for tTau [P = .05] and 0.7 [0.3] for pTau 181 [P = .04]) and 15 years before onset (mean [SE] rates of change, 5.4 [3.9] for tTau [P = .17] and 1.1 [0.5] for pTau181 [P = .03]), respectively. The rate of change of pTau181 was positive and increased at the early stages of the disease, showing a positive rate of change starting at 15 estimated years before onset until 5 estimated years before onset (mean [SE], 0.4 [0.3]), followed by a positive but decreasing rate of change at year 0 (mean [SE], 0.1 [0.3]) and then negative rates of change at 5 years (mean [SE], -0.3 [0.4]) and 10 years (mean [SE], -0.6 [0.6]) after symptom onset. In individuals without symptoms (Clinical Dementia Rating score, 0), the rates of change of CSF tTau and pTau181 were negatively associated with brain atrophy (high rates of change in CSF measures were associated with low rates of change in brain volume in asymptomatic stages). After symptom onset (Clinical Dementia Rating score, >0), an increased rate of brain atrophy was not associated with rates of change of levels of both CSF tTau and pTau181. CONCLUSIONS AND RELEVANCE These findings suggest that CSF tTau and pTau181 may have different associations with brain atrophy across the disease time course. These results have implications for understanding the dynamics of disease pathobiology and interpreting neuronal injury biomarker concentrations in response to Alzheimer disease progression and disease-modifying therapies.
Collapse
Affiliation(s)
| | - Yan Li
- Department of Biostatistics, Washington University in St Louis, St Louis, Missouri
| | | | - Brian A. Gordon
- Department of Radiology, Washington University in St Louis, St Louis, Missouri
| | - Anne M. Fagan
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
- Department of Pathology and Immunology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - John C. Morris
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
- Hope Center for Neurological Disorders, St Louis, Missouri
- Knight Alzheimer’s Disease Research Center, St Louis, Missouri
| | | | - Jason Hassenstab
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| | - Guoqiao Wang
- Hertie Institute for Clinical Brain Research, Department of Cellular Neurology, University of Tübingen, Tübingen, Germany
| | - Ricardo Allegri
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires, Argentina
| | - Sarah B. Berman
- Department of Radiology, Washington University in St Louis, St Louis, Missouri
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | - David M. Holtzman
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
- Hope Center for Neurological Disorders, St Louis, Missouri
- Knight Alzheimer’s Disease Research Center, St Louis, Missouri
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, Department of Cellular Neurology, University of Tübingen, Tübingen, Germany
- DZNE-German Center for Neurodegenerative Diseases, Tübingen, Tübingen, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
- DZNE-German Center for Neurodegenerative Diseases, Munich, Munich, Germany
- SyNergy, Munich Cluster for Systems Neurology, Munich, Germany
| | - James M. Noble
- Taub Institute for Research on Alzheimer’s Disease, Aging Brain G.H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, New York
| | - Stephen Salloway
- Memory & Aging Program, Butler Hospital, Brown University, Providence, Rhode Island
| | - Peter Schofield
- Neuroscience Research Australia, Randwick, Sydney, New South Wales, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Celeste Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Nick C. Fox
- Dementia Research Centre, University College London, London, United Kingdom
| | - Chengjie Xiong
- Department of Biostatistics, Washington University in St Louis, St Louis, Missouri
| | - Randall J. Bateman
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, Missouri
| |
Collapse
|
44
|
Alzheimer's disease clinical variants show distinct regional patterns of neurofibrillary tangle accumulation. Acta Neuropathol 2019; 138:597-612. [PMID: 31250152 DOI: 10.1007/s00401-019-02036-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/16/2019] [Accepted: 06/16/2019] [Indexed: 10/26/2022]
Abstract
The clinical spectrum of Alzheimer's disease (AD) extends well beyond the classic amnestic-predominant syndrome. The previous studies have suggested differential neurofibrillary tangle (NFT) burden between amnestic and logopenic primary progressive aphasia presentations of AD. In this study, we explored the regional distribution of NFT pathology and its relationship to AD presentation across five different clinical syndromes. We assessed NFT density throughout six selected neocortical and hippocampal regions using thioflavin-S fluorescent microscopy in a well-characterized clinicopathological cohort of pure AD cases enriched for atypical clinical presentations. Subjects underwent apolipoprotein E genotyping and neuropsychological testing. Main cognitive domains (executive, visuospatial, language, and memory function) were assessed using an established composite z score. Our results showed that NFT regional burden aligns with the clinical presentation and region-specific cognitive scores. Cortical, but not hippocampal, NFT burden was higher among atypical clinical variants relative to the amnestic syndrome. In analyses of specific clinical variants, logopenic primary progressive aphasia showed higher NFT density in the superior temporal gyrus (p = 0.0091), and corticobasal syndrome showed higher NFT density in the primary motor cortex (p = 0.0205) relative to the amnestic syndrome. Higher NFT burden in the angular gyrus and CA1 sector of the hippocampus were independently associated with worsening visuospatial dysfunction. In addition, unbiased hierarchical clustering based on regional NFT densities identified three groups characterized by a low overall NFT burden, high overall burden, and cortical-predominant burden, respectively, which were found to differ in sex ratio, age, disease duration, and clinical presentation. In comparison, the typical, hippocampal sparing, and limbic-predominant subtypes derived from a previously proposed algorithm did not reproduce the same degree of clinical relevance in this sample. Overall, our results suggest domain-specific functional consequences of regional NFT accumulation. Mapping these consequences presents an opportunity to increase understanding of the neuropathological framework underlying atypical clinical manifestations.
Collapse
|
45
|
Ohm DT, Fought AJ, Rademaker A, Kim G, Sridhar J, Coventry C, Gefen T, Weintraub S, Bigio E, Mesulam MM, Rogalski E, Geula C. Neuropathologic basis of in vivo cortical atrophy in the aphasic variant of Alzheimer's disease. Brain Pathol 2019; 30:332-344. [PMID: 31446630 DOI: 10.1111/bpa.12783] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/16/2019] [Indexed: 12/22/2022] Open
Abstract
The neuropathologic basis of in vivo cortical atrophy in clinical dementia syndromes remains poorly understood. This includes primary progressive aphasia (PPA), a language-based dementia syndrome characterized by asymmetric cortical atrophy. The neurofibrillary tangles (NFTs) and amyloid-ß plaques (APs) of Alzheimer's disease (AD) can cause PPA, but a quantitative investigation of the relationships between NFTs, APs and in vivo cortical atrophy in PPA-AD is lacking. The present study measured cortical atrophy from corresponding bilateral regions in five PPA-AD participants with in vivo magnetic resonance imaging scans 7-30 months before death and acquired stereologic estimates of NFTs and dense-core APs visualized with the Thioflavin-S stain. Linear mixed models accounting for repeated measures and stratified by hemisphere and region (language vs. non-language) were used to determine the relationships between cortical atrophy and AD neuropathology and their regional selectivity. Consistent with the aphasic profile of PPA, left language regions displayed more cortical atrophy (P = 0.01) and NFT densities (P = 0.02) compared to right language homologues. Left language regions also showed more cortical atrophy (P < 0.01) and NFT densities (P = 0.02) than left non-language regions. A subset of data was analyzed to determine the predilection of AD neuropathology for neocortical regions compared to entorhinal cortex in the left hemisphere, which showed that the three most atrophied language regions had greater NFT (P = 0.04) and AP densities (P < 0.01) than the entorhinal cortex. These results provide quantitative evidence that NFT accumulation in PPA selectively targets the language network and may not follow the Braak staging of neurofibrillary degeneration characteristic of amnestic AD. Only NFT densities, not AP densities, were positively associated with cortical atrophy within left language regions (P < 0.01) and right language homologues (P < 0.01). Given previous findings from amnestic AD, the current study of PPA-AD provides converging evidence that NFTs are the principal determinants of atrophy and clinical phenotypes associated with AD.
Collapse
Affiliation(s)
- Daniel T Ohm
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Angela J Fought
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611.,Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Alfred Rademaker
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611.,Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Garam Kim
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Jaiashre Sridhar
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Eileen Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Marek Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611.,Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611
| |
Collapse
|
46
|
Ryals AJ, O’Neil JT, Mesulam MM, Weintraub S, Voss JL. Memory awareness disruptions in amnestic mild cognitive impairment: comparison of multiple awareness types for verbal and visuospatial material. NEUROPSYCHOLOGY, DEVELOPMENT, AND COGNITION. SECTION B, AGING, NEUROPSYCHOLOGY AND COGNITION 2019; 26:577-598. [PMID: 30080435 PMCID: PMC6453739 DOI: 10.1080/13825585.2018.1503994] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 07/09/2018] [Indexed: 12/12/2022]
Abstract
Successful memory is normally accompanied by explicit awareness of retrieval and confidence in the accuracy of the retrieval product. Prior findings suggest that these features of metamemory can be dissociated from retrieval accuracy in Amnestic Mild Cognitive Impairment (aMCI). However, the literature on this question contains variable and conflicting results, likely because of differences in experimental conditions. We sought to systematically evaluate memory awareness disruptions in aMCI using multiple measures and stimulus formats within the same individuals. Memory awareness was tested with global predictions and postdictions, judgments of learning, confidence level ratings, and modified feeling-of-knowing ratings in tasks of visuospatial and verbal memory. These tests were administered to 14 individuals with aMCI and 15 healthy, age-matched controls. Memory awareness accuracy was calculated as the correspondence between subjective judgments and memory performance.Individuals with aMCI demonstrated impaired global and trial-level retrospective task awareness for visuospatial and verbal stimuli. Additionally, modified feeling-of-knowing awareness was impaired selectively for verbal stimuli. Statistical effect sizes for global awareness impairments were comparable to impairments in several objective neuropsychological memory assessments.Memory awareness (metamemory) disruptions in aMCI were most evident for a subset of subjective judgment types and task input modalities. These findings advance understanding of the nature of memory impairments in aMCI and support the utility of incorporating memory awareness testing to better characterize memory integrity in older adults.
Collapse
Affiliation(s)
| | - Jonathan T. O’Neil
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago IL
| | - M.-Marsel Mesulam
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago IL
- Cognitive Neurology and Alzheimer’s Disease Center, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Sandra Weintraub
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago IL
- Cognitive Neurology and Alzheimer’s Disease Center, Northwestern University Feinberg School of Medicine, Chicago IL
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Joel L. Voss
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago IL
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago IL
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago IL
| |
Collapse
|
47
|
Mendez MF, Moheb N, Desarzant RE, Teng EH. The Progressive Acalculia Presentation of Parietal Variant Alzheimer's Disease. J Alzheimers Dis 2019; 63:941-948. [PMID: 29710718 DOI: 10.3233/jad-180024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Many patients with early-onset Alzheimer's disease (EOAD; age of onset <65 years) have non-amnestic presentations involving language (logopenic primary progressive aphasia, lvPPA), visuospatial abilities (posterior cortical atrophy, PCA), and even asymmetric symptoms consistent with corticobasal syndrome (CBS). An inferior parietal lobule variant of EOAD commonly presents with progressive difficulty with calculations. METHODS We reviewed 276 EOAD patients for presentations with predominant acalculia. These patients were diagnosed with clinically probable Alzheimer's disease (AD) verified by positron emission tomography (PET) or cerebrospinal fluid amyloid-β or tau biomarkers. RESULTS We identified 18 (9M/9F) (6.5%) EOAD patients with progressive acalculia that did not meet most criteria for lvPPA, visual PCA, or CBS. Their ages of onset and presentation were 56.6 (5.0) and 59.4 (6.5), respectively. Their acalculia was consistent with a primary acalculia ("anarithmetia") not explained by language or visuospatial impairments. Many also had anomia (14/18), ideomotor apraxia (13/18), and the complete Gerstmann's syndrome (7/18). Visual analysis of their diverse magnetic resonance imaging disclosed biparietal atrophy, disproportionately worse on the left. CONCLUSIONS Primary acalculia may be the most common manifestation of an inferior parietal presentation of EOAD affecting the left intraparietal sulcus. This parietal variant also commonly involves progressive anomia, ideomotor apraxia, and other elements of Gerstmann's syndrome. The early recognition of patients with this variant, which is distinguishable from lvPPA, visual PCA, or CBS, would be facilitated by its recognition as a unique subtype of EOAD.
Collapse
Affiliation(s)
- Mario F Mendez
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.,Neurobehavior Unit, V.A. Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Negar Moheb
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Randy E Desarzant
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.,Neurobehavior Unit, V.A. Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Edmond H Teng
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.,Neurobehavior Unit, V.A. Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
48
|
Tetzloff KA, Graff-Radford J, Martin PR, Tosakulwong N, Machulda MM, Duffy JR, Clark HM, Senjem ML, Schwarz CG, Spychalla AJ, Drubach DA, Jack CR, Lowe VJ, Josephs KA, Whitwell JL. Regional Distribution, Asymmetry, and Clinical Correlates of Tau Uptake on [18F]AV-1451 PET in Atypical Alzheimer's Disease. J Alzheimers Dis 2019; 62:1713-1724. [PMID: 29614676 DOI: 10.3233/jad-170740] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Despite common pathology, Alzheimer's disease (AD) can have multiple clinical presentations which pathological studies suggest result from differences in the regional distribution of tau pathology. Positron emission tomography (PET) ligands are now available that can detect tau proteins in vivo and hence can be used to investigate the biological mechanisms underlying atypical AD. OBJECTIVE To assess regional patterns of tau uptake on PET imaging in two atypical AD variants, posterior cortical atrophy (PCA) and logopenic progressive aphasia (lvPPA). METHODS Eighteen PCA and 19 lvPPA subjects that showed amyloid-β deposition on PET underwent tau-PET imaging with [18F]AV-1451. Group comparisons of tau uptake in PCA and lvPPA were performed using voxel-level and regional-level analyses. We also assessed the degree of lobar tau asymmetry and correlated regional tau uptake to age and performance on clinical evaluations. RESULTS Both syndromes showed diffuse tau uptake throughout all cortical regions, although PCA showed greater uptake in occipital regions compared to lvPPA, and lvPPA showed greater uptake in left frontal and temporal regions compared to PCA. While lvPPA showed predominant left-asymmetric tau deposition, PCA was more bilateral. Younger subjects showed greater tau uptake bilaterally in frontal and parietal lobes than older subjects, and sentence repetition, Boston naming test, simultanagnosia, and visuoperceptual function showed specific regional tau correlates. CONCLUSION Tau deposition is closely related to clinical presentation in atypical AD with age playing a role in determining the degree of cortical tau deposition.
Collapse
Affiliation(s)
| | | | - Peter R Martin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Joseph R Duffy
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.,Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
49
|
Lagarde J, Hahn V, Sarazin M. Afasia primaria progressiva. Neurologia 2019. [DOI: 10.1016/s1634-7072(19)42020-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
50
|
Verclytte S, Lopes R, Viard R, Rollin A, Vanhoutte M, Pasquier F, Pruvo JP, Leclerc X. Differences in cortical perfusion detected by arterial spin labeling in nonamnestic and amnestic subtypes of early-onset Alzheimer's disease. J Neuroradiol 2019; 47:284-291. [PMID: 30981825 DOI: 10.1016/j.neurad.2019.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/26/2019] [Accepted: 03/30/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Early-onset Alzheimer's disease (EOAD) begins before the age of 65 and is characterized by a faster clinical course and the frequency of nonamnestic symptoms compared to late onset Alzheimer disease (LOAD). However, the pathophysiological process of EOAD remains unclear. We expected that ASL may show widespread cortical hypoperfusion in EOAD compared to LOAD and in nonamnestic EOAD compared to amnestic EOAD. METHODS In this study, 26 EOAD patients (16 amnestic and 10 nonamnestic patients), 29 LOAD patients and 12 healthy controls underwent pseudo-continuous ASL and 3D FFE T1 sequences. Statistical comparisons between EOAD, LOAD and control groups were made after surface-based analysis of CBF maps in regressing out the cortical thickness. RESULTS ASL showed a more severe hypoperfusion in nonamnestic EOAD patients compared to amnestic EOAD ones, with mean CBF values (± std) of 26.9 (± 3.8) and 46.6 (± 24.1) mL/100 g/min respectively (P = 0.014), located in the bilateral temporo-parietal neocortex, the precuneus, the posterior cingulate cortices (PCC) and frontal lobes. Comparison between EOAD and LOAD patients showed a trend to hypoperfusion in the left parietal lobe, PCC and precuneus in EOAD (P < 0.001 uncorrected). CONCLUSIONS Different patterns of hypoperfusion between nonamnestic and amnestic EOAD subtypes were identified, with a more severe and extensive hypoperfusion in nonamnestic patients. A trend towards more severe hypoperfusion was detected in EOAD compared to LOAD. Further studies are needed to validate ASL as a potential tool for the distinction of EOAD subtypes and the prediction of the time course of the disease.
Collapse
Affiliation(s)
- Sebastien Verclytte
- Imaging department, Lille catholic hospitals, Lille catholic university, 59000 Lille, France.
| | - Renaud Lopes
- U1171, In-vivo imaging platform, predictive medicine and therapeutic research institute, université Lille Nord de France, 59000 Lille, France
| | - Romain Viard
- U1171, In-vivo imaging platform, predictive medicine and therapeutic research institute, université Lille Nord de France, 59000 Lille, France
| | - Adeline Rollin
- Memory resources and research center, CHU de Lille, 59000 Lille, France
| | - Matthieu Vanhoutte
- U1171, In-vivo imaging platform, predictive medicine and therapeutic research institute, université Lille Nord de France, 59000 Lille, France
| | - Florence Pasquier
- Memory resources and research center, CHU de Lille, 59000 Lille, France
| | - Jean-Pierre Pruvo
- U1171, In-vivo imaging platform, predictive medicine and therapeutic research institute, université Lille Nord de France, 59000 Lille, France; Department of neuroradiology, CHU de Lille, 59000 Lille, France
| | - Xavier Leclerc
- U1171, In-vivo imaging platform, predictive medicine and therapeutic research institute, université Lille Nord de France, 59000 Lille, France; Department of neuroradiology, CHU de Lille, 59000 Lille, France
| |
Collapse
|