1
|
Balint B, Neo S, Magrinelli F, Mulroy E, Latorre A, Stamelou M, Morris HR, Batla A, Bhatia KP. Ethnic Differences in Atypical Parkinsonism-is South Asian PSP Different? Mov Disord Clin Pract 2024; 11:1355-1364. [PMID: 39113437 PMCID: PMC11542300 DOI: 10.1002/mdc3.14182] [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: 03/21/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) is a progressive atypical parkinsonian condition that results in severe disability. There are few studies of PSP in patients of non-white European ancestry. OBJECTIVES We aim to perform deep phenotyping in a South Asian PSP cohort to uncover possible ethnic differences in disease characteristics. METHODS Consecutive PSP patients had their clinical records reviewed for clinical features operationalized in the Movement Disorder Society (MDS)-PSP diagnostic criteria and relevant investigations, including imaging and genetic tests. Clinical variables were summarized by descriptive statistics and Kaplan-Meier curves were generated for survival analysis. RESULTS Twenty-seven patients, comprising Indians (78%), Pakistanis (11%) and Sri Lankans (11%) were included. Mean age of symptom onset was 63.8 ± 7.0 years and 22% of patients had an early age of onset (<60 years). The most common presenting symptom was parkinsonism (56%), followed by cognitive dysfunction (37%), falls (33%) and dysarthria (26%). The predominance types at final review were distributed across PSP-RS (67%), PSP-PGF (15%), PSP-P (15%) and PSP-F (4%). Atypical clinical features like cerebellar signs (33%), REM-sleep behavior disorder (RBD) (55%), visual hallucinations (22%), and a family history of parkinsonism (20%) were evident in a proportion of patients. CONCLUSIONS We present a South Asian cohort of PSP patients with a higher than previously reported percentages of early-onset disease, family history and atypical clinical manifestations. These patients do not fit easily into the PSP phenotypes defined by the current MDS criteria. Dedicated clinicopathological and genetic tests are needed in this population to dissect the pathogenesis of clinically-defined PSP.
Collapse
Affiliation(s)
- Bettina Balint
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK
- Department of NeurologyUniversity Hospital Zurich and University of ZurichZurichSwitzerland
| | - Shermyn Neo
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK
- Department of NeurologyNational Neuroscience InstituteSingaporeSingapore
| | - Francesca Magrinelli
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK
| | - Eoin Mulroy
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK
| | - Anna Latorre
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK
| | - Maria Stamelou
- Parkinson and Movement Disorders DepartmentHYGEIA HospitalAthensGreece
- European University of CyprusNicosiaCyprus
| | - Huw R. Morris
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK
| | - Amit Batla
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK
| | - Kailash P. Bhatia
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUK
| |
Collapse
|
2
|
Lam TG, Ross SK, Ciener B, Xiao H, Flaherty D, Lee AJ, Dugger BN, Reddy H, Teich AF. Pathologic subtyping of Alzheimer's disease brain tissue reveals disease heterogeneity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.14.24315458. [PMID: 39484271 PMCID: PMC11527055 DOI: 10.1101/2024.10.14.24315458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
In recent years, multiple groups have shown that what is currently thought of as "Alzheimer's Disease" (AD) may be usefully viewed as several related disease subtypes. As these efforts have continued, a related issue is how common co-pathologies and ethnicity intersect with AD subtypes. The goal of this study was to use a dataset constituting 153 pathologic variables recorded on 666 AD brain autopsies to better define how co-pathologies and ethnicity relate to established AD subtypes. Pathologic clustering suggests 8 subtypes within this cohort, and further analysis reveals that the previously described continuum from hippocampal predominant to hippocampal sparing is well represented in our data. Small vessel disease is overall highest in a cluster with a low hippocampal/cortical tau ratio, and across all clusters small vessel disease segregates separately from Lewy body disease. Two AD clusters are identified with extensive Lewy bodies outside amygdala (one with a high hippocampal/cortical tau ratio and one with a low ratio), and we find an inverse relationship between cortical tau and Lewy body pathology across these two clusters. Finally, we find that brains from persons of Hispanic descent have significantly more AD pathology in multiple neuroanatomic areas. We find that Hispanic ethnicity is not uniformly distributed across clusters, and this is particularly pronounced in clusters with significant Lewy body pathology, where Hispanic donors are only found in a cluster with a low hippocampal/cortical tau ratio. In summary, our analysis of recorded pathologic data across two decades of banked brains reveals new relationships in the patterns of AD-related proteinopathy, co-pathology, and ethnicity, and highlights the utility of pathologic subtyping to classify AD pathology.
Collapse
Affiliation(s)
- Tiffany G. Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sophie K. Ross
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Benjamin Ciener
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Harrison Xiao
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Delaney Flaherty
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Annie J. Lee
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Brittany N. Dugger
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Hasini Reddy
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Andrew F. Teich
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
3
|
Barba L, Abu-Rumeileh S, Barthel H, Massa F, Foschi M, Bellomo G, Gaetani L, Thal DR, Parnetti L, Otto M. Clinical and diagnostic implications of Alzheimer's disease copathology in Lewy body disease. Brain 2024; 147:3325-3343. [PMID: 38991041 DOI: 10.1093/brain/awae203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/03/2024] [Accepted: 06/02/2024] [Indexed: 07/13/2024] Open
Abstract
Concomitant Alzheimer's disease (AD) pathology is a frequent event in the context of Lewy body disease (LBD), occurring in approximately half of all cases. Evidence shows that LBD patients with AD copathology show an accelerated disease course, a greater risk of cognitive decline and an overall poorer prognosis. However, LBD-AD cases may show heterogeneous motor and non-motor phenotypes with a higher risk of dementia and, consequently, be not rarely misdiagnosed. In this review, we summarize the current understanding of LBD-AD by discussing the synergistic effects of AD neuropathological changes and Lewy pathology and their clinical relevance. Furthermore, we provide an extensive overview of neuroimaging and fluid biomarkers under assessment for use in LBD-AD and their possible diagnostic and prognostic values. AD pathology can be predicted in vivo by means of CSF, MRI and PET markers, whereas the most promising technique to date for identifying Lewy pathology in different biological tissues is the α-synuclein seed amplification assay. Pathological imaging and CSF AD biomarkers are associated with a higher likelihood of cognitive decline in LBD but do not always mirror the neuropathological severity as in pure AD. Implementing the use of blood-based AD biomarkers might allow faster screening of LBD patients for AD copathology, thus improving the overall diagnostic sensitivity for LBD-AD. Finally, we discuss the literature on novel candidate biomarkers being exploited in LBD-AD to investigate other aspects of neurodegeneration, such as neuroaxonal injury, glial activation and synaptic dysfunction. The thorough characterization of AD copathology in LBD should be taken into account when considering differential diagnoses of dementia syndromes, to allow prognostic evaluation on an individual level, and to guide symptomatic and disease-modifying therapies.
Collapse
Affiliation(s)
- Lorenzo Barba
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig 04103, Germany
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Matteo Foschi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila 67100, Italy
- Department of Neuroscience, Neurology Unit, S. Maria delle Croci Hospital of Ravenna, AUSL Romagna, Ravenna 48121, Italy
| | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Dietmar R Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Leuven 3001, Belgium
- Department of Pathology, UZ Leuven, Leuven 3000, Belgium
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Markus Otto
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| |
Collapse
|
4
|
Kalia LV, Berg D, Kordower JH, Shannon KM, Taylor JP, Cardoso F, Goldman JG, Jeon B, Meissner WG, Tijssen MAJ, Burn DJ, Fung VSC. International Parkinson and Movement Disorder Society Viewpoint on Biological Frameworks of Parkinson's Disease: Current Status and Future Directions. Mov Disord 2024; 39:1710-1715. [PMID: 39250594 DOI: 10.1002/mds.30007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/11/2024] Open
Affiliation(s)
- Lorraine V Kalia
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Daniela Berg
- Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel and Christian Albrechts-University of Kiel, Kiel, Germany
| | - Jeffery H Kordower
- ASU-Banner Neurodegenerative Disease Research Center, Tempe, Arizona, USA
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - Kathleen M Shannon
- Department of Neurology, University of Wisconsin School of Public Health, Madison, Wisconsin, USA
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Francisco Cardoso
- Movement Disorders Unit, Neurology Service, Internal Medicine Department, The Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jennifer G Goldman
- Barrow Neurological Institute, Phoenix, Arizona, USA
- JPG Enterprises LLC, Chicago, Illinois, USA
| | - Beomseok Jeon
- BJ Center for Comprehensive Parkinson Care and Rare Movement Disorders, Chung-Ang University Health Care System, Hyundae Hospital, Namyangju-si, South Korea
| | - Wassilos G Meissner
- CHU Bordeaux, Service de Neurologie des Maladies Neurodégénératives, IMNc, Bordeaux, France
- Univ. Bordeaux, CNRS, IMN, UMR5293, Bordeaux, France
- Department of Medicine, University of Otago, Christchurch, and New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Marina A J Tijssen
- Department of Neurology, Expertise Centre Movement Disorders, University Medical Centre Groningen, Groningen, The Netherlands
| | - David J Burn
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Victor S C Fung
- Movement Disorders Unit, Department of Neurology, Westmead Hospital, Westmead, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
5
|
Keir G, Roytman M, Mashriqi F, Shahsavarani S, Franceschi AM. Atypical Parkinsonian Syndromes: Structural, Functional, and Molecular Imaging Features. AJNR Am J Neuroradiol 2024:ajnr.A8313. [PMID: 39209485 DOI: 10.3174/ajnr.a8313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/16/2024] [Indexed: 09/04/2024]
Abstract
Atypical parkinsonian syndromes, also known as Parkinson-plus syndromes, are a heterogeneous group of movement disorders, including dementia with Lewy bodies (DLB), progressive supranuclear palsy (PSP), multisystem atrophy (MSA), and corticobasal degeneration (CBD). This review highlights the characteristic structural, functional, and molecular imaging features of these complex disorders. DLB typically demonstrates parieto-occipital hypometabolism with involvement of the cuneus on FDG-PET, whereas dopaminergic imaging, such as [123I]-FP-CIT SPECT (DaTscan) or fluorodopa (FDOPA)-PET, can be utilized as an adjunct for diagnosis. PSP typically shows midbrain atrophy on structural imaging, whereas FDG-PET may be useful to depict frontal lobe hypometabolism and tau-PET confirms underlying tauopathy. MSA typically demonstrates putaminal or cerebellar atrophy, whereas FDG-PET highlights characteristic nigrostriatal or olivopontocerebellar hypometabolism, respectively. Finally, CBD typically shows asymmetric atrophy in the superior parietal lobules and corpus callosum, whereas FDG and tau-PET demonstrate asymmetric hemispheric and subcortical involvement contralateral to the side of clinical deficits. Additional advanced neuroimaging modalities and techniques described may assist in the diagnostic work-up or are promising areas of emerging research.
Collapse
Affiliation(s)
- Graham Keir
- From the Neuroradiology Division (G.K., M.R.), Department of Radiology, Weill Cornell Medical College, NY-Presbyterian Hospital, New York, New York
| | - Michelle Roytman
- From the Neuroradiology Division (G.K., M.R.), Department of Radiology, Weill Cornell Medical College, NY-Presbyterian Hospital, New York, New York
| | - Faizullah Mashriqi
- Neuroradiology Division (F.M., S.S., A.M.F.), Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, New York
| | - Shaya Shahsavarani
- Neuroradiology Division (F.M., S.S., A.M.F.), Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, New York
| | - Ana M Franceschi
- Neuroradiology Division (F.M., S.S., A.M.F.), Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Lenox Hill Hospital, New York, New York
| |
Collapse
|
6
|
Wise A, Li J, Yamakawa M, Loureiro J, Peterson B, Worringer K, Sivasankaran R, Palma JA, Mitic L, Heuer HW, Lario-Lago A, Staffaroni AM, Clark A, Taylor J, Ljubenkov PA, Vandevrede L, Grinberg LT, Spina S, Seeley WW, Miller BL, Boeve BF, Dickerson BC, Grossman M, Litvan I, Pantelyat A, Tartaglia MC, Zhang Z, Wills AMA, Rexach J, Rojas JC, Boxer AL. CSF Proteomics in Patients With Progressive Supranuclear Palsy. Neurology 2024; 103:e209585. [PMID: 38959435 PMCID: PMC11226322 DOI: 10.1212/wnl.0000000000209585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 05/15/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Identification of fluid biomarkers for progressive supranuclear palsy (PSP) is critical to enhance therapeutic development. We implemented unbiased DNA aptamer (SOMAmer) proteomics to identify novel CSF PSP biomarkers. METHODS This is a cross-sectional study in original (18 clinically diagnosed PSP-Richardson syndrome [PSP-RS], 28 cognitively healthy controls]), validation (23 PSP-RS, 26 healthy controls), and neuropathology-confirmed (21 PSP, 52 non-PSP frontotemporal lobar degeneration) cohorts. Participants were recruited through the University of California, San Francisco, and the 4-Repeat Neuroimaging Initiative. The original and neuropathology cohorts were analyzed with the SomaScan platform version 3.0 (5026-plex) and the validation cohort with version 4.1 (7595-plex). Clinical severity was measured with the PSP Rating Scale (PSPRS). CSF proteomic data were analyzed to identify differentially expressed targets, implicated biological pathways using enrichment and weighted consensus gene coexpression analyses, diagnostic value of top targets with receiver-operating characteristic curves, and associations with disease severity with linear regressions. RESULTS A total of 136 participants were included (median age 70.6 ± 8 years, 68 [50%] women). One hundred fifty-five of 5,026 (3.1%), 959 of 7,595 (12.6%), and 321 of 5,026 (6.3%) SOMAmers were differentially expressed in PSP compared with controls in original, validation, and neuropathology-confirmed cohorts, with most of the SOMAmers showing reduced signal (83.1%, 95.1%, and 73.2%, respectively). Three coexpression modules were associated with PSP across cohorts: (1) synaptic function/JAK-STAT (β = -0.044, corrected p = 0.002), (2) vesicle cytoskeletal trafficking (β = 0.039, p = 0.007), and (3) cytokine-cytokine receptor interaction (β = -0.032, p = 0.035) pathways. Axon guidance was the top dysregulated pathway in PSP in original (strength = 1.71, p < 0.001), validation (strength = 0.84, p < 0.001), and neuropathology-confirmed (strength = 0.78, p < 0.001) cohorts. A panel of axon guidance pathway proteins discriminated between PSP and controls in original (area under the curve [AUC] = 0.924), validation (AUC = 0.815), and neuropathology-confirmed (AUC = 0.932) cohorts. Two inflammatory proteins, galectin-10 and cytotoxic T lymphocyte-associated protein-4, correlated with PSPRS scores across cohorts. DISCUSSION Axon guidance pathway proteins and several other molecular pathways are downregulated in PSP, compared with controls. Proteins in these pathways may be useful targets for biomarker or therapeutic development.
Collapse
Affiliation(s)
- Amy Wise
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jingyao Li
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Mai Yamakawa
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Joseph Loureiro
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Brant Peterson
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Kathleen Worringer
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Rajeev Sivasankaran
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jose-Alberto Palma
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Laura Mitic
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Hilary W Heuer
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Argentina Lario-Lago
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Adam M Staffaroni
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Annie Clark
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jack Taylor
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Peter A Ljubenkov
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Lawren Vandevrede
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Lea T Grinberg
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Salvatore Spina
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - William W Seeley
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Bruce L Miller
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Bradley F Boeve
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Bradford C Dickerson
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Murray Grossman
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Irene Litvan
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Alexander Pantelyat
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Maria Carmela Tartaglia
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Zihan Zhang
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Anne-Marie A Wills
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jessica Rexach
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Julio C Rojas
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Adam L Boxer
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| |
Collapse
|
7
|
Lo Buono V, Culicetto L, Berenati M, Stroscio G, Sorbera C, Brigandì A, Marino S, Di Lorenzo G, Quartarone A, De Cola MC. Psychological Factors Affecting Assertiveness in Subjects with Parkinson's Disease. J Clin Med 2024; 13:4625. [PMID: 39200767 PMCID: PMC11354411 DOI: 10.3390/jcm13164625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Background/Objectives: Assertiveness, defined as the positive affirmation of oneself, encompasses the ability to refuse requests, express anger, disagree or oppose others, show affection, and uphold personal beliefs without causing conflict. Deficits in assertive behavior are often linked to pathological changes in the basal ganglia and prefrontal dopaminergic systems, commonly observed in Parkinson's disease (PD), and are predictive of poor clinical outcomes. Psychological factors such as mood alterations and cognitive dysfunction may also impact assertiveness. This study investigated the psychological factors influencing assertiveness in individuals with PD. Methods: A cross-sectional study was conducted, involving 160 patients with PD attending a movement disorders outpatient clinic. The participants underwent assessment using a battery of standardized neuropsychological tests to evaluate cognitive function, assertiveness, mood, dysarthria, and quality of life (QoL). Results: All dimensions of assertiveness correlated with depression and anxiety. Individuals experiencing mood disturbances may struggle to express themselves assertively. Similarly, some dimensions of assertiveness correlated also with the QoL, indicating that, overall, well-being affects assertive behavior. Gender emerged as a significant influencer of assertiveness across all dimensions. Specifically, in subjects with PD, the male gender was associated with lower scores in assertiveness compared to women. No significant correlations were found between assertiveness and dysarthria. Conclusions: The findings highlight the importance of adopting a holistic approach to PD management, addressing not only motor symptoms but also psychological challenges which patients may encounter in their daily lives.
Collapse
Affiliation(s)
| | - Laura Culicetto
- IRCCS Centro Neurolesi “Bonino-Pulejo”, S.S. 113 Via Palermo C. da Casazza, 98124 Messina, Italy; (V.L.B.); (M.B.); (G.S.); (C.S.); (A.B.); (S.M.); (G.D.L.); (A.Q.); (M.C.D.C.)
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Tanner CM, Ostrem JL. Parkinson's Disease. N Engl J Med 2024; 391:442-452. [PMID: 39083773 DOI: 10.1056/nejmra2401857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Affiliation(s)
- Caroline M Tanner
- From the Movement Disorders and Neuromodulation Center, Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco, San Francisco
| | - Jill L Ostrem
- From the Movement Disorders and Neuromodulation Center, Department of Neurology, Weill Institute for Neuroscience, University of California, San Francisco, San Francisco
| |
Collapse
|
9
|
Wang HP, Scalco R, Saito N, Beckett L, Nguyen ML, Huie EZ, Honig LS, DeCarli C, Rissman RA, Teich AF, Mungas DM, Jin LW, Dugger BN. The neuropathological landscape of small vessel disease and Lewy pathology in a cohort of Hispanic and non-Hispanic White decedents with Alzheimer disease. Acta Neuropathol Commun 2024; 12:81. [PMID: 38790074 PMCID: PMC11127432 DOI: 10.1186/s40478-024-01773-4] [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: 02/22/2024] [Accepted: 04/03/2024] [Indexed: 05/26/2024] Open
Abstract
Cerebrovascular and α-synuclein pathologies are frequently observed alongside Alzheimer disease (AD). The heterogeneity of AD necessitates comprehensive approaches to postmortem studies, including the representation of historically underrepresented ethnic groups. In this cohort study, we evaluated small vessel disease pathologies and α-synuclein deposits among Hispanic decedents (HD, n = 92) and non-Hispanic White decedents (NHWD, n = 184) from three Alzheimer's Disease Research Centers: Columbia University, University of California San Diego, and University of California Davis. The study included cases with a pathological diagnosis of Intermediate/High AD based on the National Institute on Aging- Alzheimer's Association (NIA-AA) and/or NIA-Reagan criteria. A 2:1 random comparison sample of NHWD was frequency-balanced and matched with HD by age and sex. An expert blinded to demographics and center origin evaluated arteriolosclerosis, cerebral amyloid angiopathy (CAA), and Lewy bodies/Lewy neurites (LBs/LNs) with a semi-quantitative approach using established criteria. There were many similarities and a few differences among groups. HD showed more severe Vonsattel grading of CAA in the cerebellum (p = 0.04), higher CAA density in the posterior hippocampus and cerebellum (ps = 0.01), and increased LBs/LNs density in the frontal (p = 0.01) and temporal cortices (p = 0.03), as determined by Wilcoxon's test. Ordinal logistic regression adjusting for age, sex, and center confirmed these findings except for LBs/LNs in the temporal cortex. Results indicate HD with AD exhibit greater CAA and α-synuclein burdens in select neuroanatomic regions when compared to age- and sex-matched NHWD with AD. These findings aid in the generalizability of concurrent arteriolosclerosis, CAA, and LBs/LNs topography and severity within the setting of pathologically confirmed AD, particularly in persons of Hispanic descent, showing many similarities and a few differences to those of NHW descent and providing insights into precision medicine approaches.
Collapse
Affiliation(s)
- Hsin-Pei Wang
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Rebeca Scalco
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Naomi Saito
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Davis, CA, USA
| | - Laurel Beckett
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Davis, CA, USA
| | - My-Le Nguyen
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Emily Z Huie
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Lawrence S Honig
- Taub Institute for Research on Alzheimer's Disease and Aging Brain, Department of Neurology, Columbia University Medical Center, New York, USA
| | - Charles DeCarli
- Alzheimer's Disease Research Center, Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California San Diego, San Diego, La Jolla, CA, USA
| | - Andrew F Teich
- Taub Institute for Research on Alzheimer's Disease and Aging Brain, Department of Neurology, Department of Pathology and Cell Biology, Columbia University Medical Center, New York, USA
| | - Dan M Mungas
- Alzheimer's Disease Research Center, Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
- Alzheimer's Disease Research Center, Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA, USA.
- Alzheimer's Disease Research Center, Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
10
|
Nazeen S, Wang X, Zielinski D, Lam I, Hallacli E, Xu P, Ethier E, Strom R, Zanella CA, Nithianandam V, Ritter D, Henderson A, Saurat N, Afroz J, Nutter-Upham A, Benyamini H, Copty J, Ravishankar S, Morrow A, Mitchel J, Neavin D, Gupta R, Farbehi N, Grundman J, Myers RH, Scherzer CR, Trojanowski JQ, Van Deerlin VM, Cooper AA, Lee EB, Erlich Y, Lindquist S, Peng J, Geschwind DH, Powell J, Studer L, Feany MB, Sunyaev SR, Khurana V. Deep sequencing of proteotoxicity modifier genes uncovers a Presenilin-2/beta-amyloid-actin genetic risk module shared among alpha-synucleinopathies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583145. [PMID: 38496508 PMCID: PMC10942362 DOI: 10.1101/2024.03.03.583145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Whether neurodegenerative diseases linked to misfolding of the same protein share genetic risk drivers or whether different protein-aggregation pathologies in neurodegeneration are mechanistically related remains uncertain. Conventional genetic analyses are underpowered to address these questions. Through careful selection of patients based on protein aggregation phenotype (rather than clinical diagnosis) we can increase statistical power to detect associated variants in a targeted set of genes that modify proteotoxicities. Genetic modifiers of alpha-synuclein (ɑS) and beta-amyloid (Aβ) cytotoxicity in yeast are enriched in risk factors for Parkinson's disease (PD) and Alzheimer's disease (AD), respectively. Here, along with known AD/PD risk genes, we deeply sequenced exomes of 430 ɑS/Aβ modifier genes in patients across alpha-synucleinopathies (PD, Lewy body dementia and multiple system atrophy). Beyond known PD genes GBA1 and LRRK2, rare variants AD genes (CD33, CR1 and PSEN2) and Aβ toxicity modifiers involved in RhoA/actin cytoskeleton regulation (ARGHEF1, ARHGEF28, MICAL3, PASK, PKN2, PSEN2) were shared risk factors across synucleinopathies. Actin pathology occurred in iPSC synucleinopathy models and RhoA downregulation exacerbated ɑS pathology. Even in sporadic PD, the expression of these genes was altered across CNS cell types. Genome-wide CRISPR screens revealed the essentiality of PSEN2 in both human cortical and dopaminergic neurons, and PSEN2 mutation carriers exhibited diffuse brainstem and cortical synucleinopathy independent of AD pathology. PSEN2 contributes to a common-risk signal in PD GWAS and regulates ɑS expression in neurons. Our results identify convergent mechanisms across synucleinopathies, some shared with AD.
Collapse
Affiliation(s)
- Sumaiya Nazeen
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Xinyuan Wang
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dina Zielinski
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Whitehead Institute of Biomedical Research, Cambridge, MA, USA
| | - Isabel Lam
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Erinc Hallacli
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ping Xu
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth Ethier
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ronya Strom
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Camila A Zanella
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Vanitha Nithianandam
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dylan Ritter
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Alexander Henderson
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Nathalie Saurat
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Jalwa Afroz
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | | | - Hadar Benyamini
- Whitehead Institute of Biomedical Research, Cambridge, MA, USA
| | - Joseph Copty
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | | | - Autumn Morrow
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Jonathan Mitchel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Health Sciences & Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA
| | - Drew Neavin
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Renuka Gupta
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Nona Farbehi
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Jennifer Grundman
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Richard H Myers
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Clemens R Scherzer
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| | - Vivianna M Van Deerlin
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| | - Antony A Cooper
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Edward B Lee
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| | - Yaniv Erlich
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Susan Lindquist
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jian Peng
- Department of Computer Science, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Daniel H Geschwind
- Center for Autism Research and Treatment, Semel Institute, Program in Neurogenetics, Department of Neurology and Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Joseph Powell
- Garvan Institute of Medical Research, Sydney, NSW, Australia
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Shamil R Sunyaev
- Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vikram Khurana
- Division of Movement Disorders, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
11
|
Watanabe Y, Miyazaki Y, Hata M, Fukuma R, Aoki Y, Kazui H, Araki T, Taomoto D, Satake Y, Suehiro T, Sato S, Kanemoto H, Yoshiyama K, Ishii R, Harada T, Kishima H, Ikeda M, Yanagisawa T. A deep learning model for the detection of various dementia and MCI pathologies based on resting-state electroencephalography data: A retrospective multicentre study. Neural Netw 2024; 171:242-250. [PMID: 38101292 DOI: 10.1016/j.neunet.2023.12.009] [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: 01/21/2023] [Revised: 11/12/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
Dementia and mild cognitive impairment (MCI) represent significant health challenges in an aging population. As the search for noninvasive, precise and accessible diagnostic methods continues, the efficacy of electroencephalography (EEG) combined with deep convolutional neural networks (DCNNs) in varied clinical settings remains unverified, particularly for pathologies underlying MCI such as Alzheimer's disease (AD), dementia with Lewy bodies (DLB) and idiopathic normal-pressure hydrocephalus (iNPH). Addressing this gap, our study evaluates the generalizability of a DCNN trained on EEG data from a single hospital (Hospital #1). For data from Hospital #1, the DCNN achieved a balanced accuracy (bACC) of 0.927 in classifying individuals as healthy (n = 69) or as having AD, DLB, or iNPH (n = 188). The model demonstrated robustness across institutions, maintaining bACCs of 0.805 for data from Hospital #2 (n = 73) and 0.920 at Hospital #3 (n = 139). Additionally, the model could differentiate AD, DLB, and iNPH cases with bACCs of 0.572 for data from Hospital #1 (n = 188), 0.619 for Hospital #2 (n = 70), and 0.508 for Hospital #3 (n = 139). Notably, it also identified MCI pathologies with a bACC of 0.715 for Hospital #1 (n = 83), despite being trained on overt dementia cases instead of MCI cases. These outcomes confirm the DCNN's adaptability and scalability, representing a significant stride toward its clinical application. Additionally, our findings suggest a potential for identifying shared EEG signatures between MCI and dementia, contributing to the field's understanding of their common pathophysiological mechanisms.
Collapse
Affiliation(s)
- Yusuke Watanabe
- Institute for Advanced Co-creation Studies, Osaka University, Osaka, Japan
| | - Yuki Miyazaki
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masahiro Hata
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryohei Fukuma
- Institute for Advanced Co-creation Studies, Osaka University, Osaka, Japan; Department of Neurosurgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasunori Aoki
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Psychiatry, Nippon Life Hospital, Osaka, Japan
| | - Hiroaki Kazui
- Department of Neuropsychiatry, Kochi Medical School, Kochi University, Kochi, Japan
| | - Toshihiko Araki
- Department of Medical Technology, Osaka University Hospital, Osaka, Japan
| | - Daiki Taomoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuto Satake
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takashi Suehiro
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shunsuke Sato
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hideki Kanemoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenji Yoshiyama
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryouhei Ishii
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Occupational Therapy, Graduate School of Rehabilitation Science, Osaka Metropolitan University, Habikino, Japan
| | - Tatsuya Harada
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan; RIKEN, Tokyo, Japan
| | - Haruhiko Kishima
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takufumi Yanagisawa
- Institute for Advanced Co-creation Studies, Osaka University, Osaka, Japan; Department of Neurosurgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
12
|
Babiloni C, Noce G, Tucci F, Jakhar D, Ferri R, Panerai S, Catania V, Soricelli A, Salvatore M, Nobili F, Arnaldi D, Famà F, Buttinelli C, Giubilei F, Onofrj M, Stocchi F, Vacca L, Radicati F, Fuhr P, Gschwandtner U, Ransmayr G, Parnetti L, Marizzoni M, D'Antonio F, Bruno G, De Lena C, Güntekin B, Yıldırım E, Hanoğlu L, Yener G, Hünerli D, Taylor JP, Schumacher J, McKeith I, Frisoni GB, Antonini A, Ferreri F, Bonanni L, De Pandis MF, Del Percio C. Poor reactivity of posterior electroencephalographic alpha rhythms during the eyes open condition in patients with dementia due to Parkinson's disease. Neurobiol Aging 2024; 135:1-14. [PMID: 38142464 DOI: 10.1016/j.neurobiolaging.2023.11.010] [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: 07/11/2022] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 12/26/2023]
Abstract
Here, we hypothesized that the reactivity of posterior resting-state electroencephalographic (rsEEG) alpha rhythms during the transition from eyes-closed to -open condition might be lower in patients with Parkinson's disease dementia (PDD) than in patients with Alzheimer's disease dementia (ADD). A Eurasian database provided clinical-demographic-rsEEG datasets in 73 PDD patients, 35 ADD patients, and 25 matched cognitively unimpaired (Healthy) persons. The eLORETA freeware was used to estimate cortical rsEEG sources. Results showed substantial (greater than -10%) reduction (reactivity) in the posterior alpha source activities from the eyes-closed to the eyes-open condition in 88% of the Healthy seniors, 57% of the ADD patients, and only 35% of the PDD patients. In these alpha-reactive participants, there was lower reactivity in the parietal alpha source activities in the PDD group than in the healthy control seniors and the ADD patients. These results suggest that PDD patients show poor reactivity of mechanisms desynchronizing posterior rsEEG alpha rhythms in response to visual inputs. That neurophysiological biomarker may provide an endpoint for (non) pharmacological interventions for improving vigilance regulation in those patients.
Collapse
Affiliation(s)
- Claudio Babiloni
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy; Hospital San Raffaele Cassino, Cassino, FR, Italy.
| | | | - Federico Tucci
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| | - Dharmendra Jakhar
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| | | | | | | | - Andrea Soricelli
- IRCCS Synlab SDN, Naples, Italy; Department of Motor Sciences and Healthiness, University of Naples Parthenope, Naples, Italy
| | | | - Flavio Nobili
- Dipartimento di Neuroscienze, Oftalmologia, Genetica, Riabilitazione e Scienze Materno-infantili (DiNOGMI), Università di Genova, Italy; Clinica neurologica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Dario Arnaldi
- Dipartimento di Neuroscienze, Oftalmologia, Genetica, Riabilitazione e Scienze Materno-infantili (DiNOGMI), Università di Genova, Italy; Neurofisiopatologia, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Famà
- Dipartimento di Neuroscienze, Oftalmologia, Genetica, Riabilitazione e Scienze Materno-infantili (DiNOGMI), Università di Genova, Italy; Neurofisiopatologia, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Carla Buttinelli
- Department of Neuroscience, Mental Health and Sensory Organs, Sapienza University of Rome, Rome, Italy
| | - Franco Giubilei
- Department of Neuroscience, Mental Health and Sensory Organs, Sapienza University of Rome, Rome, Italy
| | - Marco Onofrj
- Department of Neuroscience Imaging and Clinical Sciences, CESI, and Department of Medicine and Aging Sciences, University G d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Fabrizio Stocchi
- IRCCS San Raffaele, Rome, Italy; Telematic University San Raffaele, Rome, Italy
| | | | | | - Peter Fuhr
- Universitätsspital Basel, Abteilung Neurophysiologie, Petersgraben 4, 4031 Basel, Switzerland; Departments of Neurology and of Clinical Research, University Hospital Basel, Switzerland
| | - Ute Gschwandtner
- Universitätsspital Basel, Abteilung Neurophysiologie, Petersgraben 4, 4031 Basel, Switzerland; Departments of Neurology and of Clinical Research, University Hospital Basel, Switzerland
| | - Gerhard Ransmayr
- Department of Neurology 2, Med Campus III, Faculty of Medicine, Johannes Kepler University, Kepler University Hospital, Krankenhausstr. 9, A-4020 Linz., Austria
| | - Lucilla Parnetti
- Centre for Memory Disturbances, Lab of Clinical Neurochemistry, Section of Neurology, University of Perugia, Italy
| | - Moira Marizzoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Fabrizia D'Antonio
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Bruno
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | | | - Bahar Güntekin
- Department of Biophysics, School of Medicine, Istanbul Medipol University, Istanbul, Turkey; Research Institute for Health Sciences and Technologies (SABITA), Neuroscience Research Center, Istanbul Medipol University, Istanbul, Turkey
| | - Ebru Yıldırım
- Program of Electroneurophysiology, Vocational School, Istanbul Medipol University, Istanbul, Turkey
| | - Lutfu Hanoğlu
- Department of Neurology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Görsev Yener
- Izmir University of Economics, Faculty of Medicine, Izmir, Turkey
| | - Duygu Hünerli
- Health Sciences Institute, Department of Neurosciences, Dokuz Eylül University, Izmir, Turkey
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - Julia Schumacher
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - Ian McKeith
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - Giovanni B Frisoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Memory Clinic and LANVIE - Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Angelo Antonini
- Unit and Study Center for Neurodegenerative diseases (CESNE), Department of Neuroscience, University of Padua, Padua, Italy
| | - Florinda Ferreri
- Unit and Study Center for Neurodegenerative diseases (CESNE), Department of Neuroscience, University of Padua, Padua, Italy; Department of Clinical Neurophysiology, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Laura Bonanni
- Department of Medicine and Aging Sciences, University G d'Annunzio of Chieti-Pescara, Chieti, Italy
| | | | - Claudio Del Percio
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
13
|
Walker L, Simpson H, Thomas AJ, Attems J. Prevalence, distribution, and severity of cerebral amyloid angiopathy differ between Lewy body diseases and Alzheimer's disease. Acta Neuropathol Commun 2024; 12:28. [PMID: 38360761 PMCID: PMC10870546 DOI: 10.1186/s40478-023-01714-7] [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: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 02/17/2024] Open
Abstract
Dementia with Lewy bodies (DLB), Parkinson's disease dementia (PDD), and Parkinson's disease (PD) collectively known as Lewy body diseases (LBDs) are neuropathologically characterised by α-synuclein deposits (Lewy bodies and Lewy neurites). However, LBDs also exhibit pathology associated with Alzheimer's disease (AD) (i.e. hyperphosphorylated tau and amyloid β (Aβ). Aβ can be deposited in the walls of blood vessels in the brains of individuals with AD, termed cerebral amyloid angiopathy (CAA). The aim of this study was to investigate the type and distribution of CAA in DLB, PDD, and PD and determine if this differs from AD. CAA type, severity, and topographical distribution was assessed in 94 AD, 30 DLB, 17 PDD, and 11 PD cases, and APOE genotype evaluated in a subset of cases where available. 96.3% AD cases, 70% DLB cases and 82.4% PDD cases exhibited CAA (type 1 or type 2). However only 45.5% PD cases had CAA. Type 1 CAA accounted for 37.2% of AD cases, 10% of DLB cases, and 5.9% of PDD cases, and was not observed in PD cases. There was a hierarchical topographical distribution in regions affected by CAA where AD and DLB displayed the same distribution pattern that differed from PDD and PD. APOE ε4 was associated with severity of CAA in AD cases. Topographical patterns and severity of CAA in DLB more closely resembled AD rather than PDD, and as type 1 CAA is associated with clinical dementia in AD, further investigations are warranted into whether the increased presence of type 1 CAA in DLB compared to PDD are related to the onset of cognitive symptoms and is a distinguishing factor between LBDs. Possible alignment of the the topographical distribution of CAA and microbleeds in DLB warrants further investigation. CAA in DLB more closely resembles AD rather than PDD or PD, and should be taken into consideration when stratifying patients for clinical trials or designing disease modifying therapies.
Collapse
Affiliation(s)
- Lauren Walker
- Translational and Clinical Research Institute, Newcastle University, Edwardson building, Campus for Ageing and Vitality, Newcastle-upon-Tyne, NE4 5PL, UK.
| | - Harry Simpson
- Translational and Clinical Research Institute, Newcastle University, Edwardson building, Campus for Ageing and Vitality, Newcastle-upon-Tyne, NE4 5PL, UK
| | - Alan J Thomas
- Translational and Clinical Research Institute, Newcastle University, Edwardson building, Campus for Ageing and Vitality, Newcastle-upon-Tyne, NE4 5PL, UK
| | - Johannes Attems
- Translational and Clinical Research Institute, Newcastle University, Edwardson building, Campus for Ageing and Vitality, Newcastle-upon-Tyne, NE4 5PL, UK
| |
Collapse
|
14
|
Sekiya H, Koga S, Murakami A, DeTure M, Ross OA, Uitti RJ, Cheshire WP, Wszolek ZK, Dickson DW. Frequency of Comorbid Pathologies and Their Clinical Impact in Multiple System Atrophy. Mov Disord 2024; 39:380-390. [PMID: 37986699 PMCID: PMC10922743 DOI: 10.1002/mds.29670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/12/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Mixed pathology is common at autopsy for a number of age-associated neurodegenerative disorders; however, the frequency of comorbid pathologies in multiple system atrophy (MSA) and their clinical correlations are poorly understood. OBJECTIVE We determined the frequency of comorbid pathologic processes in autopsy-confirmed MSA and assessed their clinical correlates. METHODS This study included 160 neuropathologically established MSA from the Mayo Clinic brain bank. Clinical information, including age at onset or death, clinical subtype, initial symptoms, antemortem clinical diagnosis, and cognitive dysfunction was collected. We assessed comorbid pathologies including Alzheimer's disease neuropathologic change, Lewy-related pathology, argyrophilic grain disease, age-related τ astrogliopathy, transactive DNA-binding protein 43 pathology, cerebral amyloid angiopathy, and cerebrovascular small vessel disease and examined their clinical impact. RESULTS The majority of MSA patients (62%) had no significant comorbid pathologies. There was a positive correlation between age at onset or death with the number of comorbid pathologies; however, even in the highest quartile group (average age at death 78 ± 6 years), the average number of comorbid pathologies was <2. Logistic regression analysis revealed that none of the assessed variables, including sex, age at onset, and the presence or absence of each comorbid pathology, were significantly associated with cognitive dysfunction. CONCLUSIONS The majority of MSA patients do not have comorbid pathologies, even in advanced age, indicating that MSA is unique among neurodegenerative disorders in this regard. There was minimal clinical impact of comorbid pathologies in MSA. These findings warrant focusing on α-synuclein for the treatment strategy for MSA. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Hiroaki Sekiya
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Aya Murakami
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Ryan J Uitti
- Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | | | | | | |
Collapse
|
15
|
Walker L, Attems J. Prevalence of Concomitant Pathologies in Parkinson's Disease: Implications for Prognosis, Diagnosis, and Insights into Common Pathogenic Mechanisms. JOURNAL OF PARKINSON'S DISEASE 2024; 14:35-52. [PMID: 38143370 PMCID: PMC10836576 DOI: 10.3233/jpd-230154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/11/2023] [Indexed: 12/26/2023]
Abstract
Pathologies characteristic of Alzheimer's disease (i.e., hyperphosphorylated tau and amyloid-β (Aβ) plaques), cardiovascular disease, and limbic predominant TDP-43 encephalopathy (LATE) often co-exist in patients with Parkinson's disease (PD), in addition to Lewy body pathology (α-synuclein). Numerous studies point to a putative synergistic relationship between hyperphosphorylation tau, Aβ, cardiovascular lesions, and TDP-43 with α-synuclein, which may alter the stereotypical pattern of pathological progression and accelerate cognitive decline. Here we discuss the prevalence and relationships between common concomitant pathologies observed in PD. In addition, we highlight shared genetic risk factors and developing biomarkers that may provide better diagnostic accuracy for patients with PD that have co-existing pathologies. The tremendous heterogeneity observed across the PD spectrum is most likely caused by the complex interplay between pathogenic, genetic, and environmental factors, and increasing our understanding of how these relate to idiopathic PD will drive research into finding accurate diagnostic tools and disease modifying therapies.
Collapse
Affiliation(s)
- Lauren Walker
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Johannes Attems
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, UK
| |
Collapse
|
16
|
Jellinger KA. Mild cognitive impairment in dementia with Lewy bodies: an update and outlook. J Neural Transm (Vienna) 2023; 130:1491-1508. [PMID: 37418039 DOI: 10.1007/s00702-023-02670-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023]
Abstract
Dementia with Lewy bodies (DLB), the second most common degenerative neurocognitive disorder after Alzheimer disease (AD), is frequently preceded by a period of mild cognitive impairment (MCI), in which cognitive decline is associated with impairment of executive functions/attention, visuospatial deficits, or other cognitive domains and a variety of noncognitive and neuropsychiatric symptoms, many of which are similar but less severe than in prodromal AD. While 36-38% remain in the MCI state, at least the same will convert to dementia. Biomarkers are slowing of the EEG rhythms, atrophy of hippocampus and nucleus basalis of Meynert, temporoparietal hypoperfusion, signs of degeneration of the nigrostriatal dopaminergic, cholinergic and other neurotransmitter systems, and inflammation. Functional neuroimaging studies revealed disturbed connectivity of frontal and limbic networks associated with attention and cognitive controls, dopaminergic and cholinergic circuits manifested prior to overt brain atrophy. Sparse neuropathological data showed varying Lewy body and AD-related stages associated with atrophy of entorhinal, hippocampal, and mediotemporal cortices. Putative pathomechanisms of MCI are degeneration of limbic, dopaminergic, and cholinergic systems with Lewy pathology affecting specific neuroanatomical pathways associated with progressing AD-related lesions, but many pathobiological mechanisms involved in the development of MCI in LBD remain to be elucidated as a basis for early diagnosis and future adequate treatment modalities to prevent progression of this debilitating disorder.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
17
|
Harvey J, Pishva E, Chouliaras L, Lunnon K. Elucidating distinct molecular signatures of Lewy body dementias. Neurobiol Dis 2023; 188:106337. [PMID: 37918758 DOI: 10.1016/j.nbd.2023.106337] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/15/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023] Open
Abstract
Dementia with Lewy bodies and Parkinson's disease dementia are common neurodegenerative diseases that share similar neuropathological profiles and spectra of clinical symptoms but are primarily differentiated by the order in which symptoms manifest. The question of whether a distinct molecular pathological profile could distinguish these disorders is yet to be answered. However, in recent years, studies have begun to investigate genomic, epigenomic, transcriptomic and proteomic differences that may differentiate these disorders, providing novel insights in to disease etiology. In this review, we present an overview of the clinical and pathological hallmarks of Lewy body dementias before summarizing relevant research into genetic, epigenetic, transcriptional and protein signatures in these diseases, with a particular interest in those resolving "omic" level changes. We conclude by suggesting future research directions to address current gaps and questions present within the field.
Collapse
Affiliation(s)
- Joshua Harvey
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Ehsan Pishva
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Leonidas Chouliaras
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK; Specialist Dementia and Frailty Service, Essex Partnership University NHS Foundation Trust, Epping, UK
| | - Katie Lunnon
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
| |
Collapse
|
18
|
Tunold JA, Tan MMX, Koga S, Geut H, Rozemuller AJM, Valentino R, Sekiya H, Martin NB, Heckman MG, Bras J, Guerreiro R, Dickson DW, Toft M, van de Berg WDJ, Ross OA, Pihlstrøm L. Lysosomal polygenic risk is associated with the severity of neuropathology in Lewy body disease. Brain 2023; 146:4077-4087. [PMID: 37247383 PMCID: PMC10545498 DOI: 10.1093/brain/awad183] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
Intraneuronal accumulation of misfolded α-synuclein is the pathological hallmark of Parkinson's disease and dementia with Lewy bodies, often co-occurring with variable degrees of Alzheimer's disease related neuropathology. Genetic association studies have successfully identified common variants associated with disease risk and phenotypic traits in Lewy body disease, yet little is known about the genetic contribution to neuropathological heterogeneity. Using summary statistics from Parkinson's disease and Alzheimer's disease genome-wide association studies, we calculated polygenic risk scores and investigated the relationship with Lewy, amyloid-β and tau pathology. Associations were nominated in neuropathologically defined samples with Lewy body disease from the Netherlands Brain Bank (n = 217) and followed up in an independent sample series from the Mayo Clinic Brain Bank (n = 394). We also generated stratified polygenic risk scores based on single-nucleotide polymorphisms annotated to eight functional pathways or cell types previously implicated in Parkinson's disease and assessed for association with Lewy pathology in subgroups with and without significant Alzheimer's disease co-pathology. In an ordinal logistic regression model, the Alzheimer's disease polygenic risk score was associated with concomitant amyloid-β and tau pathology in both cohorts. Moreover, both cohorts showed a significant association between lysosomal pathway polygenic risk and Lewy pathology, which was more consistent than the association with a general Parkinson's disease risk score and specific to the subset of samples without significant concomitant Alzheimer's disease related neuropathology. Our findings provide proof of principle that the specific risk alleles a patient carries for Parkinson's and Alzheimer's disease also influence key aspects of the underlying neuropathology in Lewy body disease. The interrelations between genetic architecture and neuropathology are complex, as our results implicate lysosomal risk loci specifically in the subset of samples without Alzheimer's disease co-pathology. Our findings hold promise that genetic profiling may help predict the vulnerability to specific neuropathologies in Lewy body disease, with potential relevance for the further development of precision medicine in these disorders.
Collapse
Affiliation(s)
- Jon-Anders Tunold
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| | - Manuela M X Tan
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Hanneke Geut
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
- Program Neurodegeneration, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Rebecca Valentino
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Hiroaki Sekiya
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Nicholas B Martin
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Michael G Heckman
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jose Bras
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
- Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI 49503, USA
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
- Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI 49503, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
- Program Neurodegeneration, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
19
|
Jellinger KA. Depression in dementia with Lewy bodies: a critical update. J Neural Transm (Vienna) 2023; 130:1207-1218. [PMID: 37418037 DOI: 10.1007/s00702-023-02669-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
Depression with an estimated prevalence of 35% is a frequent manifestation of dementia with Lewy bodies (DLB), having negative effects on cognitive performance and life expectancy, yet the underlying neurobiology is poorly understood and most likely heterogeneous. Depressive symptoms in DLB can occur during the clinical course and, together with apathy, is a common prodromal neuropsychiatric symptom of this neurocognitive disorder in the group of Lewy body synucleinopathies. There are no essential differences in the frequency of depression in DLB and Parkinson disease-dementia (PDD), while its severity is up to twice as high as in Alzheimer disease (AD). Depression in DLB that is frequently underdiagnosed and undertreated, has been related to a variety of pathogenic mechanisms associated with the basic neurodegenerative process, in particular dysfunctions of neurotransmitter systems (decreased monoaminergic/serotonergic, noradrenergic and dopaminergic metabolism), α-synuclein pathology, synaptic zinc dysregulation, proteasome inhibition, gray matter volume loss in prefrontal and temporal areas as well as dysfunction of neuronal circuits with decreased functional connectivity of specific brain networks. Pharmacotherapy should avoid tricyclic antidepressants (anticholinergic adverse effects), second-generation antidepressants being a better choice, while modified electroconvulsive therapy, transcranial magnetic stimulation therapy and deep brain stimulation may be effective for pharmacotherapy-resistant cases. Since compared to depression in other dementias like Alzheimer disease and other parkinsonian syndromes, our knowledge of its molecular basis is limited, and further studies to elucidate the heterogeneous pathogenesis of depression in DLB are warranted.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
20
|
Jellinger KA. Morphological characteristics differentiate dementia with Lewy bodies from Parkinson disease with and without dementia. J Neural Transm (Vienna) 2023:10.1007/s00702-023-02660-3. [PMID: 37306790 DOI: 10.1007/s00702-023-02660-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/01/2023] [Indexed: 06/13/2023]
Abstract
Dementia with Lewy bodies (DLB) and Parkinson disease (PD) with and without dementia are entities of a spectrum of Lewy body diseases. About 26.3% of all PD patients develop dementia increasing up to 83%. Parkinson disease-dementia (PDD) and DLB share many clinical and morphological features that separate them from non-demented PD (PDND). Clinically distinguished by the temporal sequence of motor and cognitive symptoms, the pathology of PDD and DLB includes variable combinations of Lewy body (LB) and Alzheimer (AD) lesions, both being more severe in DLB, but much less frequent and less severe in PDND. The objective of this study was to investigate the morphological differences between these three groups. 290 patients with pathologically confirmed PD were reviewed. 190 of them had clinical dementia; 110 met the neuropathological criteria of PDD and 80 of DLB. The major demographic and clinical data were obtained from medical records. Neuropathology included semiquantitative assessment of LB and AD pathologies including cerebral amyloid angiopathy (CAA). PDD patients were significantly older than PDND and DLB ones (83.9 vs 77.9 years, p < 0.05); the age of DLB patients was between them (80.0 years), while the disease duration was shortest in DLB. Brain weight was lowest in DLB, which showed higher Braak LB scores (mean 5.2 vs 4.2) and highest Braak tau stages (mean 5.2 vs 4.4 and 2.3, respectively). Thal Aβ phases were also highest in DLB (mean 4.1 vs 3.0 and 1.8, respectively). Major findings were frequency and degree of CAA, being highest in DLB (95% vs 50% and 24%, with scores 2.9 vs 0.7 and 0.3, respectively), whereas other small vessel lesions showed no significant differences. Striatal Aβ deposits also differentiated DLB from the other groups. This and other studies of larger cohorts of PD patients indicate that the association of CAA and cortical tau-but less-LB pathologies are associated with more severe cognitive decline and worse prognosis that distinguish DLB from PDD and PDND. The particular impact of both CAA and tau pathology supports the concept of a pathogenic continuum ranging from PDND to DLB + AD within the spectrum of age-related synucleinopathies.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
21
|
New insights from a multi-ethnic Asian progressive supranuclear palsy cohort. Parkinsonism Relat Disord 2023; 108:105296. [PMID: 36682278 DOI: 10.1016/j.parkreldis.2023.105296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) is a rare, disabling, neurodegenerative disease, with few studies done in Asian populations. METHODS We prospectively characterized the clinical features and disease burden in a consecutively-recruited multi-ethnic Asian PSP cohort. Patients were extensively phenotyped using the Movement Disorder Society (MDS-PSP) clinical diagnostic criteria and the PSP-Clinical Deficits Scale (PSP-CDS). Caregiver burden was measured using the modified Zarit Burden Interview (ZBI). Investigations (neuroimaging and genetic tests) were reviewed. RESULTS There were 104 patients (64.4% male; 67.3% Chinese, 21.2% Indians, 9.6% Malays), consisting of 48.1% Richardson syndrome (PSP-RS), 37.5% parkinsonian phenotype (PSP-P), and 10.6% progressive gait freezing phenotype (PSP-PGF). Mean age at motor onset was 66.3 ± 7.7 years, with no significant differences between the PSP phenotypes. Interestingly, REM-sleep behaviour disorder (RBD) symptoms and visual hallucinations (considered rare in PSP) were reported in 23.5% and 22.8% of patients, respectively, and a family history of possible neurodegenerative or movement disorder in 20.4%. PSP-CDS scores were highest (worst) in PSP-RS; and correlated moderately with disease duration (rs = 0.45, P < 0.001) and weakly with caregiver burden (rs = 0.22, P = 0.029) in the overall cohort. Three of 48 (6.3%) patients who had whole-exome sequencing harboured pathogenic/likely pathogenic GBA variants. CONCLUSIONS Significant heterogeneity in clinical features and disease burden, and high rates of RBD symptoms, visual hallucinations, and familial involvement were observed in this relatively large cohort. Our findings highlight important considerations when assessing Asian patients, and provide further support for the notion of overlapping neurobiology between PSP and Lewy body disorders.
Collapse
|
22
|
Olszewska DA, Lang AE. The definition of precision medicine in neurodegenerative disorders and the one disease-many diseases tension. HANDBOOK OF CLINICAL NEUROLOGY 2023; 192:3-20. [PMID: 36796946 DOI: 10.1016/b978-0-323-85538-9.00005-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Precision medicine is a patient-centered approach that aims to translate new knowledge to optimize the type and timing of interventions for the greatest benefit to individual patients. There is considerable interest in applying this approach to treatments designed to slow or halt the progression of neurodegenerative diseases. Indeed, effective disease-modifying treatment (DMT) remains the greatest unmet therapeutic need in this field. In contrast to the enormous progress in oncology, precision medicine in the field of neurodegeneration faces multiple challenges. These are related to major limitations in our understanding of many aspects of the diseases. A critical barrier to advances in this field is the question of whether the common sporadic neurodegenerative diseases (of the elderly) are single uniform disorders (particularly related to their pathogenesis) or whether they represent a collection of related but still very distinct disease states. In this chapter, we briefly touch on lessons from other fields of medicine that might be applied to the development of precision medicine for DMT in neurodegenerative diseases. We discuss why DMT trials may have failed to date, and particularly the importance of appreciating the multifaceted nature of disease heterogeneity and how this has and will impact on these efforts. We conclude with comments on how we can move from this complex disease heterogeneity to the successful application of precision medicine principles in DMT for neurodegenerative diseases.
Collapse
Affiliation(s)
- Diana A Olszewska
- Department of Neurology, Division of Movement Disorders, Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada
| | - Anthony E Lang
- Department of Neurology, Division of Movement Disorders, Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada.
| |
Collapse
|
23
|
Donadio V, Sturchio A, Rizzo G, Abu Rumeileh S, Liguori R, Espay AJ. Pathology vs pathogenesis: Rationale and pitfalls in the clinicopathology model of neurodegeneration. HANDBOOK OF CLINICAL NEUROLOGY 2023; 192:35-55. [PMID: 36796947 DOI: 10.1016/b978-0-323-85538-9.00001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
In neurodegenerative disorders, the term pathology is often implicitly referred to as pathogenesis. Pathology has been conceived as a window into the pathogenesis of neurodegenerative disorders. This clinicopathologic framework posits that what can be identified and quantified in postmortem brain tissue can explain both premortem clinical manifestations and the cause of death, a forensic approach to understanding neurodegeneration. As the century-old clinicopathology framework has yielded little correlation between pathology and clinical features or neuronal loss, the relationship between proteins and degeneration is ripe for revisitation. There are indeed two synchronous consequences of protein aggregation in neurodegeneration: the loss of the soluble/normal proteins on one; the accrual of the insoluble/abnormal fraction of these proteins on the other. The omission of the first part in the protein aggregation process is an artifact of the early autopsy studies: soluble, normal proteins have disappeared, with only the remaining insoluble fraction amenable to quantification. We here review the collective evidence from human data suggesting that protein aggregates, known collectively as pathology, are the consequence of many biological, toxic, and infectious exposures, but may not explain alone the cause or pathogenesis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Vincenzo Donadio
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy.
| | - Andrea Sturchio
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, Stockholm, Sweden; James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| | - Giovanni Rizzo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Samir Abu Rumeileh
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
24
|
Abstract
Parkinson's disease (PD) is clinically, pathologically, and genetically heterogeneous, resisting distillation to a single, cohesive disorder. Instead, each affected individual develops a virtually unique form of Parkinson's syndrome. Clinical manifestations consist of variable motor and nonmotor features, and myriad overlaps are recognized with other neurodegenerative conditions. Although most commonly characterized by alpha-synuclein protein pathology throughout the central and peripheral nervous systems, the distribution varies and other pathologies commonly modify PD or trigger similar manifestations. Nearly all PD is genetically influenced. More than 100 genes or genetic loci have been identified, and most cases likely arise from interactions among many common and rare genetic variants. Despite its complex architecture, insights from experimental genetic dissection coalesce to reveal unifying biological themes, including synaptic, lysosomal, mitochondrial, andimmune-mediated mechanisms of pathogenesis. This emerging understanding of Parkinson's syndrome, coupled with advances in biomarkers and targeted therapies, presages successful precision medicine strategies.
Collapse
Affiliation(s)
- Hui Ye
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA; ,
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Laurie A Robak
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA;
| | - Meigen Yu
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA;
| | - Matthew Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, USA;
- Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA
| | - Joshua M Shulman
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA; ,
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA;
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA;
- Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
25
|
Coughlin DG, Hiniker A, Peterson C, Kim Y, Arezoumandan S, Giannini L, Pizzo D, Weintraub D, Siderowf A, Litvan I, Rissman RA, Galasko D, Hansen L, Trojanowski JQ, Lee E, Grossman M, Irwin D. Digital Histological Study of Neocortical Grey and White Matter Tau Burden Across Tauopathies. J Neuropathol Exp Neurol 2022; 81:953-964. [PMID: 36269086 PMCID: PMC9677241 DOI: 10.1093/jnen/nlac094] [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: 02/03/2023] Open
Abstract
3R/4R-tau species are found in Alzheimer disease (AD) and ∼50% of Lewy body dementias at autopsy (LBD+tau); 4R-tau accumulations are found in progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD). Digital image analysis techniques can elucidate patterns of tau pathology more precisely than traditional methods but repeatability across centers is unclear. We calculated regional percentage areas occupied by tau pathological inclusions from the middle frontal cortex (MFC), superior temporal cortex (STC), and angular gyrus (ANG) from cases from the University of Pennsylvania and the University of California San Diego with AD, LBD+tau, PSP, or CBD (n = 150) using QuPath. In both cohorts, AD and LBD+tau had the highest grey and white matter tau burden in the STC (p ≤ 0.04). White matter tau burden was relatively higher in 4R-tauopathies than 3R/4R-tauopathies (p < 0.003). Grey and white matter tau were correlated in all diseases (R2=0.43-0.79, p < 0.04) with the greatest increase of white matter per unit grey matter tau observed in PSP (p < 0.02 both cohorts). Grey matter tau negatively correlated with MMSE in AD and LBD+tau (r = -4.4 to -5.4, p ≤ 0.02). These data demonstrate the feasibility of cross-institutional digital histology studies that generate finely grained measurements of pathology which can be used to support biomarker development and models of disease progression.
Collapse
Affiliation(s)
- David G Coughlin
- From the Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Annie Hiniker
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Claire Peterson
- Digital Neuropathology Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yongya Kim
- From the Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Sanaz Arezoumandan
- Digital Neuropathology Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lucia Giannini
- Digital Neuropathology Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurology, Erasmus University Medical Center, Alzheimer Center, Rotterdam, The Netherlands
| | - Donald Pizzo
- Center for Advanced Laboratory Medicine, University of California San Diego, La Jolla, California, USA
| | - Daniel Weintraub
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrew Siderowf
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Irene Litvan
- From the Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Robert A Rissman
- From the Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Douglas Galasko
- From the Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Lawrence Hansen
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Irwin
- Digital Neuropathology Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
26
|
Esposito P, Ismail N. Linking Puberty and the Gut Microbiome to the Pathogenesis of Neurodegenerative Disorders. Microorganisms 2022; 10:2163. [PMID: 36363755 PMCID: PMC9697368 DOI: 10.3390/microorganisms10112163] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 08/31/2023] Open
Abstract
Puberty is a critical period of development marked by the maturation of the central nervous system, immune system, and hypothalamic-pituitary-adrenal axis. Due to the maturation of these fundamental systems, this is a period of development that is particularly sensitive to stressors, increasing susceptibility to neurodevelopmental and neurodegenerative disorders later in life. The gut microbiome plays a critical role in the regulation of stress and immune responses, and gut dysbiosis has been implicated in the development of neurodevelopmental and neurodegenerative disorders. The purpose of this review is to summarize the current knowledge about puberty, neurodegeneration, and the gut microbiome. We also examine the consequences of pubertal exposure to stress and gut dysbiosis on the development of neurodevelopmental and neurodegenerative disorders. Understanding how alterations to the gut microbiome, particularly during critical periods of development (i.e., puberty), influence the pathogenesis of these disorders may allow for the development of therapeutic strategies to prevent them.
Collapse
Affiliation(s)
- Pasquale Esposito
- NISE Laboratory, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Nafissa Ismail
- NISE Laboratory, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
27
|
Constant AB, Basavaraju R, France J, Honig LS, Marder KS, Provenzano FA. Longitudinal Patterns of Cortical Atrophy on MRI in Patients With Alzheimer Disease With and Without Lewy Body Pathology. Neurology 2022; 99:e1843-e1852. [PMID: 36123123 PMCID: PMC9620811 DOI: 10.1212/wnl.0000000000200947] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 05/19/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Although Alzheimer disease (AD) and dementia with Lewy bodies (DLBs) represent 2 different pathologies, they have clinical overlap, and there is a significant degree of co-occurrence of their neuropathologic findings. Many studies have examined imaging characteristics in clinically diagnosed patients; however, there is a relative lack of longitudinal studies that have studied patients with pathologic confirmation. We examined whether there were differences in longitudinal patterns of cortical atrophy between patients with both AD and DLB (AD/DLB) vs those with AD alone. METHODS We collected and analyzed clinical and neuroimaging data from the AD Neuroimaging Initiative (ADNI) database for patients who underwent autopsy. The rates of change in various neuropsychological assessments were not significantly different between patients with AD/DLB and AD, and each group had neuropsychological outcomes consistent with disease progression. For our neuroimaging analysis, we used a linear mixed-effects model to examine whether there were longitudinal differences in cortical rates of atrophy between patients with AD/DLB and AD. RESULTS Autopsies and serial neuroimaging were available on 48 patients (24 AD and 24 AD/DLB). Patients with AD alone had significantly higher atrophy rates in the left cuneus, lateral occipital, and parahippocampal regions over time when compared with patients with concomitant DLB, after covarying for interval from imaging to autopsy, sex, and total estimated intracranial volume. Site ID was included as a random effect to account for site differences. For these regions, the rate of decline over time in the AD/DLB group was less steep by a difference of 0.1887, 0.395, and 0.0989, respectively (p = 0.022, 0.006, and 0.006). The lattermost left cuneus volume measurement and Braak Lewy score had a Pearson product-moment correlation of 0.37, p = 0.009, while the lattermost left parahippocampal volume measurement and Braak neurofibrillary tangle score had a Pearson product-moment correlation of -0.327, p = 0.02. DISCUSSION Patients with AD had more significant atrophy in the left cuneus, lateral occipital, and parahippocampal regions when compared with patients with AD/DLB. These regions are known to distinguish DLB and AD pathology cross-sectionally but here are shown to distinguish longitudinal disease progression.
Collapse
Affiliation(s)
- Allison Beers Constant
- From the Columbia University Vagelos College of Physicians & Surgeons (A.B.C.), New York, NY; Department of Neurology (R.B., J.F., L.S.H., K.S.M., F.A.P.), Columbia University Medical Center, New York, NY; Department of Neurology (L.S.H., K.S.M., F.A.P.), Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY; and Department of Neurology (L.S.H., K.S.M.), Getrude H. Sergievsky Center, Columbia University Medical Center, New York, NY
| | - Rakshathi Basavaraju
- From the Columbia University Vagelos College of Physicians & Surgeons (A.B.C.), New York, NY; Department of Neurology (R.B., J.F., L.S.H., K.S.M., F.A.P.), Columbia University Medical Center, New York, NY; Department of Neurology (L.S.H., K.S.M., F.A.P.), Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY; and Department of Neurology (L.S.H., K.S.M.), Getrude H. Sergievsky Center, Columbia University Medical Center, New York, NY
| | - Jeanelle France
- From the Columbia University Vagelos College of Physicians & Surgeons (A.B.C.), New York, NY; Department of Neurology (R.B., J.F., L.S.H., K.S.M., F.A.P.), Columbia University Medical Center, New York, NY; Department of Neurology (L.S.H., K.S.M., F.A.P.), Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY; and Department of Neurology (L.S.H., K.S.M.), Getrude H. Sergievsky Center, Columbia University Medical Center, New York, NY
| | - Lawrence S Honig
- From the Columbia University Vagelos College of Physicians & Surgeons (A.B.C.), New York, NY; Department of Neurology (R.B., J.F., L.S.H., K.S.M., F.A.P.), Columbia University Medical Center, New York, NY; Department of Neurology (L.S.H., K.S.M., F.A.P.), Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY; and Department of Neurology (L.S.H., K.S.M.), Getrude H. Sergievsky Center, Columbia University Medical Center, New York, NY
| | - Karen S Marder
- From the Columbia University Vagelos College of Physicians & Surgeons (A.B.C.), New York, NY; Department of Neurology (R.B., J.F., L.S.H., K.S.M., F.A.P.), Columbia University Medical Center, New York, NY; Department of Neurology (L.S.H., K.S.M., F.A.P.), Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY; and Department of Neurology (L.S.H., K.S.M.), Getrude H. Sergievsky Center, Columbia University Medical Center, New York, NY
| | - Frank Anthony Provenzano
- From the Columbia University Vagelos College of Physicians & Surgeons (A.B.C.), New York, NY; Department of Neurology (R.B., J.F., L.S.H., K.S.M., F.A.P.), Columbia University Medical Center, New York, NY; Department of Neurology (L.S.H., K.S.M., F.A.P.), Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY; and Department of Neurology (L.S.H., K.S.M.), Getrude H. Sergievsky Center, Columbia University Medical Center, New York, NY.
| |
Collapse
|
28
|
Blackman J, Morrison HD, Lloyd K, Gimson A, Banerjee LV, Green S, Cousins R, Rudd S, Harding S, Coulthard E. The past, present, and future of sleep measurement in mild cognitive impairment and early dementia—towards a core outcome set: a scoping review. Sleep 2022; 45:6563140. [PMID: 35373837 PMCID: PMC9272273 DOI: 10.1093/sleep/zsac077] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/28/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
Study Objectives
Sleep abnormalities emerge early in dementia and may accelerate cognitive decline. Their accurate characterization may facilitate earlier clinical identification of dementia and allow for assessment of sleep intervention efficacy. This scoping review determines how sleep is currently measured and reported in Mild Cognitive Impairment (MCI) and early dementia, as a basis for future core outcome alignment.
Methods
This review follows the PRISMA Guidelines for Scoping Reviews. CINAHL, Embase, Medline, Psychinfo, and British Nursing Index databases were searched from inception—March 12, 2021. Included studies had participants diagnosed with MCI and early dementia and reported on sleep as a key objective/ outcome measure.
Results
Nineteen thousand five hundred and ninety-six titles were returned following duplicate removal with 188 studies [N] included in final analysis. Sleep data was reported on 17 139 unique, diagnostically diverse participants (n). “Unspecified MCI” was the most common diagnosis amongst patients with MCI (n = 5003, 60.6%). Despite technological advances, sleep was measured most commonly by validated questionnaires (n = 12 586, N = 131). Fewer participants underwent polysomnography (PSG) (n = 3492, N = 88) and actigraphy (n = 3359, N = 38) with little adoption of non-PSG electroencephalograms (EEG) (n = 74, N = 3). Sleep outcome parameters were reported heterogeneously. 62/165 (37.6%) were described only once in the literature (33/60 (60%) in interventional studies). There was underrepresentation of circadian (n = 725, N = 25) and micro-architectural (n = 360, N = 12) sleep parameters.
Conclusions
Alongside under-researched areas, there is a need for more detailed diagnostic characterization. Due to outcome heterogeneity, we advocate for international consensus on core sleep outcome parameters to support causal inference and comparison of therapeutic sleep interventions.
Collapse
Affiliation(s)
- Jonathan Blackman
- Bristol Medical School, University of Bristol , Bristol , UK
- Bristol Brain Centre, North Bristol NHS Trust , Bristol , UK
| | - Hamish Duncan Morrison
- Bristol Medical School, University of Bristol , Bristol , UK
- Bristol Brain Centre, North Bristol NHS Trust , Bristol , UK
| | - Katherine Lloyd
- Bristol Medical School, University of Bristol , Bristol , UK
- Bristol Brain Centre, North Bristol NHS Trust , Bristol , UK
| | - Amy Gimson
- Bristol Brain Centre, North Bristol NHS Trust , Bristol , UK
| | | | - Sebastian Green
- Bristol Medical School, University of Bristol , Bristol , UK
- Bristol Brain Centre, North Bristol NHS Trust , Bristol , UK
| | - Rebecca Cousins
- Bristol Brain Centre, North Bristol NHS Trust , Bristol , UK
| | - Sarah Rudd
- Library and Knowledge Service, North Bristol NHS Trust , Bristol , UK
| | - Sam Harding
- Research and Innovation, North Bristol NHS Trust , Bristol , UK
| | - Elizabeth Coulthard
- Bristol Medical School, University of Bristol , Bristol , UK
- Bristol Brain Centre, North Bristol NHS Trust , Bristol , UK
| |
Collapse
|
29
|
Jellinger KA. Are there morphological differences between Parkinson's disease-dementia and dementia with Lewy bodies? Parkinsonism Relat Disord 2022; 100:24-32. [PMID: 35691178 DOI: 10.1016/j.parkreldis.2022.05.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/21/2022] [Accepted: 05/30/2022] [Indexed: 12/17/2022]
Abstract
Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are two major neurocognitive disorders in the spectrum of Lewy body diseases that overlap in many clinical and neuropathological features, although they show several differences. Clinically distinguished mainly based on the duration of parkinsonism prior to development of dementia, their morphology is characterized by a variable combination of Lewy body (LB) and Alzheimer's disease (AD) pathologies, the latter usually being more frequent and severe in DLB. OBJECTIVE The aims of the study were to investigate essential neuropathological differences between PDD and DLB in a larger cohort of autopsy cases. METHODS 110 PDD autopsy cases were compared with 78 DLB cases. The major demographic, clinical (duration of illness, final MMSE) and neuropathological data were assessed retrospectively. Neuropathological studies used standardized methods and immunohistochemistry for phospho-tau, β-amyloid (Aß) and α-synuclein, with semiquantitative assessment of the major histological lesions. RESULTS PDD patients were significantly older at death than DLB ones (mean 83.9 vs. 79.8 years), with a significantly longer disease duration (mean 9.2 vs. 6.7 years). Braak LB scores and particularly neuritic Braak stages were significantly higher in the DLB group (mean 5.1and 5.1 vs. 4.2 and 4.4, respectively), as were Thal Aβ phases (mean 4.1 vs. 3.0). Diffuse striatal Aβ plaques were considerable in 55% and moderate in 45% of DLB cases, but were extremely rare in PDD. The most significant differences concerned the frequency and degree of cerebral amyloid angiopathy (CAA), being significantly higher in DLB (98.7 vs. 50%, and mean degree of 2.9 vs. 0.72, respectively). Worse prognosis in DLB than in PDD was linked to both increased Braak neuritic stages and more severe CAA. INTERPRETATION These and other recent studies imply the association of CAA, more severe concomitant AD pathology, and striatal Aβ load with cognitive decline and more rapid disease process that distinguishes DLB from PDD, while the influence of other cerebrovascular diseases or co-pathologies in both disorders was not specifically examined. The importance of both CAA and tau pathology in DLB and much less in PDD supports the concept of a pathogenetic continuum from Parkinson's disease (PD) - > PDD - > DLB - > DLB + AD and subtypes of AD with LB pathology within the spectrum of age-related proteinopathies.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Vienna, Austria, Alberichgasse 5/13, A-1150, Vienna, Austria.
| |
Collapse
|
30
|
Giannoccaro MP, Avoni P, Rizzo G, Incensi A, Infante R, Donadio V, Liguori R. Presence of Skin α-Synuclein Deposits Discriminates Parkinson's Disease from Progressive Supranuclear Palsy and Corticobasal Syndrome. JOURNAL OF PARKINSON'S DISEASE 2022; 12:585-591. [PMID: 34864689 PMCID: PMC8925116 DOI: 10.3233/jpd-212904] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Previous studies reported skin phosphorylated α-synuclein (p-syn) deposits in Parkinson's disease (PD) patients but not in patients with parkinsonism due to tauopathies, although data on the latter are limited. OBJECTIVE We aimed to assess the presence of skin p-syn deposits in patients with clinical diagnosis of parkinsonism usually due to tauopathy and PD. METHODS We consecutively recruited 26 patients, 18 fulfilling clinical diagnostic criteria of progressive supranuclear palsy (PSP) and 8 of corticobasal syndrome (CBS), 26 patients with PD, and 26 healthy controls (HC). All subjects underwent skin biopsy to study p-syn deposits in skin nerves by immunofluorescence. RESULTS Skin p-syn deposits were present in only two of the PSP/CBS patients and none of the HC. Conversely, all PD patients showed p-syn deposition (p < 0.001, Chi-square). The two p-syn positive patients were diagnosed with PSP and CBS, respectively. Although clinical and MRI findings supported these diagnoses, both patients had some atypical features more typical of synucleinopathies. CONCLUSION The detection of skin p-syn deposits may help in the differential diagnosis of parkinsonism. Indeed, in this study, all PD patients and only two out of 26 with a clinical diagnosis of PSP/CBS had skin p-syn deposits. Furthermore, these two patients showed clinical features that could suggest an atypical synucleinopathy presentation or a mixed pathology.
Collapse
Affiliation(s)
- Maria Pia Giannoccaro
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Patologia Neuromuscolare e Neuroimmunologia, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Patrizia Avoni
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Giovanni Rizzo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Alex Incensi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Patologia Neuromuscolare e Neuroimmunologia, Bologna, Italy
| | - Rossella Infante
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Vincenzo Donadio
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Patologia Neuromuscolare e Neuroimmunologia, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Rocco Liguori
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Patologia Neuromuscolare e Neuroimmunologia, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| |
Collapse
|
31
|
Scott GD, Arnold MR, Beach TG, Gibbons CH, Kanthasamy AG, Lebovitz RM, Lemstra AW, Shaw LM, Teunissen CE, Zetterberg H, Taylor AS, Graham TC, Boeve BF, Gomperts SN, Graff-Radford NR, Moussa C, Poston KL, Rosenthal LS, Sabbagh MN, Walsh RR, Weber MT, Armstrong MJ, Bang JA, Bozoki AC, Domoto-Reilly K, Duda JE, Fleisher JE, Galasko DR, Galvin JE, Goldman JG, Holden SK, Honig LS, Huddleston DE, Leverenz JB, Litvan I, Manning CA, Marder KS, Pantelyat AY, Pelak VS, Scharre DW, Sha SJ, Shill HA, Mari Z, Quinn JF, Irwin DJ. Fluid and Tissue Biomarkers of Lewy Body Dementia: Report of an LBDA Symposium. Front Neurol 2022; 12:805135. [PMID: 35173668 PMCID: PMC8841880 DOI: 10.3389/fneur.2021.805135] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
The Lewy Body Dementia Association (LBDA) held a virtual event, the LBDA Biofluid/Tissue Biomarker Symposium, on January 25, 2021, to present advances in biomarkers for Lewy body dementia (LBD), which includes dementia with Lewy bodies (DLBs) and Parkinson's disease dementia (PDD). The meeting featured eight internationally known scientists from Europe and the United States and attracted over 200 scientists and physicians from academic centers, the National Institutes of Health, and the pharmaceutical industry. Methods for confirming and quantifying the presence of Lewy body and Alzheimer's pathology and novel biomarkers were discussed.
Collapse
Affiliation(s)
- Gregory D. Scott
- Department of Pathology, Oregon Health and Science University, Portland, OR, United States
- Department of Pathology and Laboratory Services, VA Portland Medical Center, Portland, OR, United States
| | - Moriah R. Arnold
- Graduate Program in Biomedical Sciences, School of Medicine M.D./Ph.D. Program, Oregon Health and Science University, Portland, OR, United States
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology and Brain and Body Donation Program, Banner Sun Health Research Institute, Sun City, AZ, United States
| | - Christopher H. Gibbons
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Anumantha G. Kanthasamy
- Department of Physiology and Pharmacology, Center for Brain Sciences and Neurodegenerative Diseases, University of Georgia, Athens, GA, United States
| | | | - Afina W. Lemstra
- Department of Neurology, Amsterdam University Medical Center (UMC), Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London (UCL) Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at University College London, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | | | - Todd C. Graham
- Lewy Body Dementia Association, Lilburn, GA, United States
| | - Bradley F. Boeve
- Department of Neurology and Center for Sleep Medicine, Mayo Clinic, Rochester, MN, United States
| | - Stephen N. Gomperts
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | | | - Charbel Moussa
- Department of Neurology, Georgetown University Medical Center, Washington DC, CA, United States
| | - Kathleen L. Poston
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Liana S. Rosenthal
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Marwan N. Sabbagh
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Ryan R. Walsh
- Barrow Neurological Institute and Muhammed Ali Parkinson Center, Phoenix, AZ, United States
| | - Miriam T. Weber
- Department of Neurology, University of Rochester, Rochester, NY, United States
| | - Melissa J. Armstrong
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jee A. Bang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Andrea C. Bozoki
- Department of Neurology, University of North Carolina, Chapel Hill, NC, United States
| | | | - John E. Duda
- Parkinson's Disease Research, Education and Clinical Center, Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jori E. Fleisher
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, United States
| | - Douglas R. Galasko
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - James E. Galvin
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jennifer G. Goldman
- Shirley Ryan Abilitylab and Department of Physical Medicine and Rehabilitation and Neurology, Parkinson's Disease and Movement Disorders, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Samantha K. Holden
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lawrence S. Honig
- Columbia University Irving Medical Center, New York, NY, United States
| | - Daniel E. Huddleston
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - James B. Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, United States
| | - Irene Litvan
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Carol A. Manning
- Department of Neurology, University of Virginia, Charlottesville, VA, United States
| | - Karen S. Marder
- Columbia University Irving Medical Center, New York, NY, United States
| | - Alexander Y. Pantelyat
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Victoria S. Pelak
- Departments of Neurology and Ophthalmology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Douglas W. Scharre
- Department of Neurology, Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sharon J. Sha
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Holly A. Shill
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Zoltan Mari
- Lou Ruvo Center for Brain Health, Cleveland Clinic Lerner College of Medicine, Las Vegas, NV, United States
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Department of Neurology, VA Portland Medical Center, Portland, OR, United States
| | - David J. Irwin
- Department of Neurology, University of Pennsylvania Health System, Philadelphia, PA, United States
- Digital Neuropathology Laboratory, Philadelphia, PA, United States
- Lewy Body Disease Research Center of Excellence, Philadelphia, PA, United States
- Frontotemporal Degeneration Center, Philadelphia, PA, United States
| |
Collapse
|
32
|
Shakir MN, Dugger BN. Advances in Deep Neuropathological Phenotyping of Alzheimer Disease: Past, Present, and Future. J Neuropathol Exp Neurol 2022; 81:2-15. [PMID: 34981115 PMCID: PMC8825756 DOI: 10.1093/jnen/nlab122] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is a neurodegenerative disorder characterized pathologically by the presence of neurofibrillary tangles and amyloid beta (Aβ) plaques in the brain. The disease was first described in 1906 by Alois Alzheimer, and since then, there have been many advancements in technologies that have aided in unlocking the secrets of this devastating disease. Such advancements include improving microscopy and staining techniques, refining diagnostic criteria for the disease, and increased appreciation for disease heterogeneity both in neuroanatomic location of abnormalities as well as overlap with other brain diseases; for example, Lewy body disease and vascular dementia. Despite numerous advancements, there is still much to achieve as there is not a cure for AD and postmortem histological analyses is still the gold standard for appreciating AD neuropathologic changes. Recent technological advances such as in-vivo biomarkers and machine learning algorithms permit great strides in disease understanding, and pave the way for potential new therapies and precision medicine approaches. Here, we review the history of human AD neuropathology research to include the notable advancements in understanding common co-pathologies in the setting of AD, and microscopy and staining methods. We also discuss future approaches with a specific focus on deep phenotyping using machine learning.
Collapse
Affiliation(s)
- Mustafa N Shakir
- From the Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA (MNS, BND)
| | - Brittany N Dugger
- From the Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California, USA (MNS, BND)
| |
Collapse
|
33
|
Apátiga-Pérez R, Soto-Rojas LO, Campa-Córdoba BB, Luna-Viramontes NI, Cuevas E, Villanueva-Fierro I, Ontiveros-Torres MA, Bravo-Muñoz M, Flores-Rodríguez P, Garcés-Ramirez L, de la Cruz F, Montiel-Sosa JF, Pacheco-Herrero M, Luna-Muñoz J. Neurovascular dysfunction and vascular amyloid accumulation as early events in Alzheimer's disease. Metab Brain Dis 2022; 37:39-50. [PMID: 34406560 DOI: 10.1007/s11011-021-00814-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/23/2021] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is clinically characterized by a progressive loss of cognitive functions and short-term memory. AD patients present two distinctive neuropathological lesions: neuritic plaques and neurofibrillary tangles (NFTs), constituted of beta-amyloid peptide (Aβ) and phosphorylated and truncated tau proteins. Aβ deposits around cerebral blood vessels (cerebral amyloid angiopathy, CAA) is a major contributor to vascular dysfunction in AD. Vascular amyloid deposits could be early events in AD due to dysfunction in the neurovascular unit (NVU) and the blood-brain barrier (BBB), deterioration of the gliovascular unit, and/or decrease of cerebral blood flow (CBF). These pathological events can lead to decreased Aβ clearance, facilitate a neuroinflammatory environment as well as synaptic dysfunction and, finally, lead to neurodegeneration. Here, we review the histopathological AD hallmarks and discuss the two-hit vascular hypothesis of AD, emphasizing the role of neurovascular dysfunction as an early factor that favors vascular Aβ aggregation and neurodegeneration. Addtionally, we emphasize that pericyte degeneration is a key and early element in AD that can trigger amyloid vascular accumulation and NVU/BBB dysfunction. Further research is required to better understand the early pathophysiological mechanisms associated with NVU alteration and CAA to generate early biomarkers and timely treatments for AD.
Collapse
Affiliation(s)
- Ricardo Apátiga-Pérez
- National Dementia BioBank. Ciencias Biológicas. Facultad de Estudios Superiores Cuautitlán, Universidad Nacional 13 Autónoma de México, Estado de México, México
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, México
| | - Luis O Soto-Rojas
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Estado de México, Mexico
| | - B Berenice Campa-Córdoba
- National Dementia BioBank. Ciencias Biológicas. Facultad de Estudios Superiores Cuautitlán, Universidad Nacional 13 Autónoma de México, Estado de México, México
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, México
| | - Nabil Itzi Luna-Viramontes
- National Dementia BioBank. Ciencias Biológicas. Facultad de Estudios Superiores Cuautitlán, Universidad Nacional 13 Autónoma de México, Estado de México, México
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, México
| | - Elvis Cuevas
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food and Drug Administration, Jefferson, AR, USA
| | | | | | | | | | - Linda Garcés-Ramirez
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, México
| | - Fidel de la Cruz
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, México
| | - José Francisco Montiel-Sosa
- National Dementia BioBank. Ciencias Biológicas. Facultad de Estudios Superiores Cuautitlán, Universidad Nacional 13 Autónoma de México, Estado de México, México
| | - Mar Pacheco-Herrero
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Santiago de los Caballeros, Dominican Republic.
| | - José Luna-Muñoz
- National Dementia BioBank. Ciencias Biológicas. Facultad de Estudios Superiores Cuautitlán, Universidad Nacional 13 Autónoma de México, Estado de México, México.
- Banco Nacional de Cerebros-UNPHU, Universidad Nacional Pedro Henríquez Ureña, Santo Domingo, República Dominicana.
| |
Collapse
|
34
|
Hijazi Z, Yassi N, O'Brien JT, Watson R. The influence of cerebrovascular disease in dementia with Lewy bodies and Parkinson's disease dementia. Eur J Neurol 2021; 29:1254-1265. [PMID: 34923713 DOI: 10.1111/ene.15211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/08/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Lewy body dementia (LBD), including dementia with Lewy bodies (DLB) and Parkinson's disease dementia (PDD), is a common form of neurodegenerative dementia. The frequency and influence of comorbid cerebrovascular disease is not understood but has potentially important clinical management implications. METHODS A systematic literature search was conducted (Medline and Embase) for studies including participants with DLB and/or PDD assessing cerebrovascular lesions (imaging and pathological studies). They included white matter changes, cerebral amyloid angiopathy (CAA), cerebral microbleeds (CMB), macroscopic infarcts, micro-infarcts and intracerebral haemorrhage. RESULTS Of 4411 articles, 63 studies were included. Cerebrovascular lesions commonly studied included white matter changes (41 studies) and CMB (18 studies). There was an increased severity of white matter changes on magnetic resonance imaging (visualized as white matter hyperintensities, WMH), but not neuropathology, in LBD compared to PD without dementia and age-matched controls. CMB prevalence in DLB was highly variable but broadly similar to Alzheimer's disease (AD) (0-48%), with a lobar predominance. No relationship was found between large cortical or small subcortical infarcts or intracerebral haemorrhage and presence of LBD. CONCLUSION The underlying mechanisms of WMH in LBD require further exploration, as their increased severity in LBD was not supported by neuropathological examination of white matter. CMB in LBD had a similar prevalence as AD. There is a need for larger studies assessing the influence of cerebrovascular lesions on clinical symptoms, disease progression and outcomes.
Collapse
Affiliation(s)
- Zina Hijazi
- Monash University School of Rural Health, Bendigo Hospital, Bendigo, VIC, Australia.,Department of Medicine, Bendigo Hospital, Bendigo, VIC, Australia
| | - Nawaf Yassi
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.,Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville, Australia
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Level E4, Box 189, Cambridge, CB2 0QC, UK
| | - Rosie Watson
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.,Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| |
Collapse
|
35
|
Koga S, Sekiya H, Kondru N, Ross OA, Dickson DW. Neuropathology and molecular diagnosis of Synucleinopathies. Mol Neurodegener 2021; 16:83. [PMID: 34922583 PMCID: PMC8684287 DOI: 10.1186/s13024-021-00501-z] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/10/2021] [Indexed: 12/11/2022] Open
Abstract
Synucleinopathies are clinically and pathologically heterogeneous disorders characterized by pathologic aggregates of α-synuclein in neurons and glia, in the form of Lewy bodies, Lewy neurites, neuronal cytoplasmic inclusions, and glial cytoplasmic inclusions. Synucleinopathies can be divided into two major disease entities: Lewy body disease and multiple system atrophy (MSA). Common clinical presentations of Lewy body disease are Parkinson's disease (PD), PD with dementia, and dementia with Lewy bodies (DLB), while MSA has two major clinical subtypes, MSA with predominant cerebellar ataxia and MSA with predominant parkinsonism. There are currently no disease-modifying therapies for the synucleinopathies, but information obtained from molecular genetics and models that explore mechanisms of α-synuclein conversion to pathologic oligomers and insoluble fibrils offer hope for eventual therapies. It remains unclear how α-synuclein can be associated with distinct cellular pathologies (e.g., Lewy bodies and glial cytoplasmic inclusions) and what factors determine neuroanatomical and cell type vulnerability. Accumulating evidence from in vitro and in vivo experiments suggests that α-synuclein species derived from Lewy body disease and MSA are distinct "strains" having different seeding properties. Recent advancements in in vitro seeding assays, such as real-time quaking-induced conversion (RT-QuIC) and protein misfolding cyclic amplification (PMCA), not only demonstrate distinct seeding activity in the synucleinopathies, but also offer exciting opportunities for molecular diagnosis using readily accessible peripheral tissue samples. Cryogenic electron microscopy (cryo-EM) structural studies of α-synuclein derived from recombinant or brain-derived filaments provide new insight into mechanisms of seeding in synucleinopathies. In this review, we describe clinical, genetic and neuropathologic features of synucleinopathies, including a discussion of the evolution of classification and staging of Lewy body disease. We also provide a brief discussion on proposed mechanisms of Lewy body formation, as well as evidence supporting the existence of distinct α-synuclein strains in Lewy body disease and MSA.
Collapse
Affiliation(s)
- Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, FL 32224 Jacksonville, USA
| | - Hiroaki Sekiya
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, FL 32224 Jacksonville, USA
| | - Naveen Kondru
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, FL 32224 Jacksonville, USA
| | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, FL 32224 Jacksonville, USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, FL 32224 Jacksonville, USA
| |
Collapse
|
36
|
Schumacher J, Gunter JL, Przybelski SA, Jones DT, Graff-Radford J, Savica R, Schwarz CG, Senjem ML, Jack CR, Lowe VJ, Knopman DS, Fields JA, Kremers WK, Petersen RC, Graff-Radford NR, Ferman TJ, Boeve BF, Thomas AJ, Taylor JP, Kantarci K. Dementia with Lewy bodies: association of Alzheimer pathology with functional connectivity networks. Brain 2021; 144:3212-3225. [PMID: 34114602 PMCID: PMC8634124 DOI: 10.1093/brain/awab218] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/19/2021] [Accepted: 04/22/2021] [Indexed: 11/22/2022] Open
Abstract
Dementia with Lewy bodies (DLB) is neuropathologically defined by the presence of α-synuclein aggregates, but many DLB cases show concurrent Alzheimer's disease pathology in the form of amyloid-β plaques and tau neurofibrillary tangles. The first objective of this study was to investigate the effect of Alzheimer's disease co-pathology on functional network changes within the default mode network (DMN) in DLB. Second, we studied how the distribution of tau pathology measured with PET relates to functional connectivity in DLB. Twenty-seven DLB, 26 Alzheimer's disease and 99 cognitively unimpaired participants (balanced on age and sex to the DLB group) underwent tau-PET with AV-1451 (flortaucipir), amyloid-β-PET with Pittsburgh compound-B (PiB) and resting-state functional MRI scans. The resing-state functional MRI data were used to assess functional connectivity within the posterior DMN. This was then correlated with overall cortical flortaucipir PET and PiB PET standardized uptake value ratio (SUVr). The strength of interregional functional connectivity was assessed using the Schaefer atlas. Tau-PET covariance was measured as the correlation in flortaucipir SUVr between any two regions across participants. The association between region-to-region functional connectivity and tau-PET covariance was assessed using linear regression. Additionally, we identified the region with highest and the region with lowest tau SUVrs (tau hot- and cold spots) and tested whether tau SUVr in all other brain regions was associated with the strength of functional connectivity to these tau hot and cold spots. A reduction in posterior DMN connectivity correlated with overall higher cortical tau- (r = -0.39, P = 0.04) and amyloid-PET uptake (r = -0.41, P = 0.03) in the DLB group, i.e. patients with DLB who have more concurrent Alzheimer's disease pathology showed a more severe loss of DMN connectivity. Higher functional connectivity between regions was associated with higher tau covariance in cognitively unimpaired, Alzheimer's disease and DLB. Furthermore, higher functional connectivity of a target region to the tau hotspot (i.e. inferior/medial temporal cortex) was related to higher flortaucipir SUVrs in the target region, whereas higher functional connectivity to the tau cold spot (i.e. sensory-motor cortex) was related to lower flortaucipir SUVr in the target region. Our findings suggest that a higher burden of Alzheimer's disease co-pathology in patients with DLB is associated with more Alzheimer's disease-like changes in functional connectivity. Furthermore, we found an association between the brain's functional network architecture and the distribution of tau pathology that has recently been described in Alzheimer's disease. We show that this relationship also exists in patients with DLB, indicating that similar mechanisms of connectivity-dependent occurrence of tau pathology might be at work in both diseases.
Collapse
Affiliation(s)
- Julia Schumacher
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey L Gunter
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Rodolfo Savica
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Julie A Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Walter K Kremers
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Tanis J Ferman
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Alan J Thomas
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
37
|
Tsamakis K, Mueller C. Challenges in Predicting Cognitive Decline in Dementia with Lewy Bodies. Dement Geriatr Cogn Disord 2021; 50:1-8. [PMID: 33780925 DOI: 10.1159/000515008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/01/2021] [Indexed: 11/19/2022] Open
Abstract
Despite being the second most common form of neurodegenerative dementia, dementia with Lewy bodies (DLB) is under-recognized and carries a worse prognosis than other subtypes of the condition. Cognitive impairment is a cardinal feature of all types of dementia and DLB presents with a distinct profile with deficits in attention, executive function, and visuoperceptual abilities. This difference from Alzheimer's disease and the common presence of neuropsychiatric symptoms may lead to challenges in predicting cognitive decline in this patient population. Firstly, the diagnosis of DLB is often delayed in clinical practice leading to variability from which time point in the disease course cognitive decline is measured. Secondly, the most frequently used measurement tools for cognitive difficulties focus on memory and naming rather than the domains affected by DLB. While there is now largely a consensus which tools are useful in diagnosing DLB, their validity in assessing deteriorating cognition is less clear. Thirdly, the presence of fluctuating cognition, the propensity to develop delirium episodes, as well as difficulties in distinguishing the two entities in clinical practice make it difficult to predict the disease course. Sleep disturbances are likely to influence cognitive decline but require further study in patients within established DLB. Fourthly, as in most cases of dementia, neuropathological comorbidities are frequently present in DLB. While the influence of Alzheimer's pathology on cognitive decline in DLB is comparatively well understood, the impact of other pathologies remains unclear. The recent definition of research criteria for mild cognitive impairment in DLB could facilitate earlier diagnosis and more structured follow-up. Assessment tools measuring cognitive domains predominantly affected in DLB need to be more consistently used in longitudinal studies and clinical practice, as well as concurrent measures of fluctuations in cognition. Greater availability of biomarkers and digital healthcare solutions can play an important role in enabling more accurate monitoring and prediction of cognitive decline in DLB.
Collapse
Affiliation(s)
- Konstantinos Tsamakis
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Second Department of Psychiatry, School of Medicine, University General Hospital 'ATTIKON', Athens, Greece
| | - Christoph Mueller
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,South London and Maudsley NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
38
|
Tandon A, Singh SJ, Chaturvedi RK. Nanomedicine against Alzheimer's and Parkinson's Disease. Curr Pharm Des 2021; 27:1507-1545. [PMID: 33087025 DOI: 10.2174/1381612826666201021140904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/06/2020] [Accepted: 08/18/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's and Parkinson's are the two most rampant neurodegenerative disorders worldwide. Existing treatments have a limited effect on the pathophysiology but are unable to fully arrest the progression of the disease. This is due to the inability of these therapeutic molecules to efficiently cross the blood-brain barrier. We discuss how nanotechnology has enabled researchers to develop novel and efficient nano-therapeutics against these diseases. The development of nanotized drug delivery systems has permitted an efficient, site-targeted, and controlled release of drugs in the brain, thereby presenting a revolutionary therapeutic approach. Nanoparticles are also being thoroughly studied and exploited for their role in the efficient and precise diagnosis of neurodegenerative conditions. We summarize the role of different nano-carriers and RNAi-conjugated nanoparticle-based therapeutics for their efficacy in pre-clinical studies. We also discuss the challenges underlying the use of nanomedicine with a focus on their route of administration, concentration, metabolism, and any toxic effects for successful therapeutics in these diseases.
Collapse
Affiliation(s)
- Ankit Tandon
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Sangh J Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Rajnish K Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| |
Collapse
|
39
|
Karanth S, Nelson PT, Katsumata Y, Kryscio RJ, Schmitt FA, Fardo DW, Cykowski MD, Jicha GA, Van Eldik LJ, Abner EL. Prevalence and Clinical Phenotype of Quadruple Misfolded Proteins in Older Adults. JAMA Neurol 2021; 77:1299-1307. [PMID: 32568358 DOI: 10.1001/jamaneurol.2020.1741] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Importance Quadruple misfolded proteins (tau neurofibrillary tangles, amyloid-β [Aβ], α-synuclein, and transactive response DNA-binding protein 43 [TDP-43]) in the same brain are relatively common in aging. However, the clinical presentation, associated factors, frequency in community-based cohorts, genetic characteristics, and cognitive trajectories associated with the quadruple misfolded proteins phenotype are not well understood. Objective To describe the quadruple misfolded proteins phenotype, including the trajectories of global cognition, in an autopsy cohort. Design, Setting, and Participants This retrospective cohort study used brain autopsy data from the University of Kentucky Alzheimer Disease Center (UK-ADC) Brain Bank. Participants were deceased individuals who were enrolled in a longitudinal community-based cohort study of aging and dementia in central Kentucky conducted by the UK-ADC. Included participants were enrolled in the UK-ADC cohort between January 1, 1989, and December 31, 2017; aged 55 years or older at baseline; and followed up for a mean duration of 10.4 years. The participants had Alzheimer disease pathology (tau and Aβ), α-synuclein, and TDP-43 data, along with Braak neurofibrillary tangle stage I to VI. Data analysis was conducted between February 1, 2019, and September 30, 2019. Main Outcomes and Measures Frequency of quadruple misfolded proteins was estimated, and proteinopathy group characteristics and associations with global cognition were evaluated. Multinomial logistic regression was used to estimate the association of proteinopathy group with participant characteristics, including age at death, sex, and apolipoprotein ε4 (APOE ε4) allele. Generalized estimating equations were used to estimate the probability of obtaining Mini-Mental State Examination (MMSE) scores within the normal cognition (27-30 points) and severe impairment (≤13 points) ranges during the 12 years before death. Results The final sample included 375 individuals (mean [SD] age at death, 86.9 [8.0] years); 232 women [61.9%]). Quadruple misfolded proteins were detected in 41 of 214 individuals with dementia (19.2%). Overall, 46 individuals (12.3%) had quadruple misfolded proteins, whereas 143 individuals (38.1%) had 3 proteinopathies. Dementia frequency was highest among those with quadruple misfolded proteins (41 [89.1%]), and participants with quadruple misfolded proteins had the lowest final mean (SD) MMSE scores of 13.4 (9.8) points. Adjusting for age at death and sex, the APOE ε4 allele was associated with higher odds of quadruple misfolded proteins (adjusted odds ratio, 2.55; 95% CI, 1.16- 5.62; P = .02). The quadruple misfolded proteins group had both the lowest probability of obtaining MMSE scores in the normal cognition range, even 12 years before death, and the highest probability of having MMSE scores in the severe impairment range. Conclusions and Relevance Quadruple misfolded proteins appear to be a common substrate for cognitive impairment and to be associated with an aggressive course of disease that typically ends with severe dementia. The prevalence of comorbid α-synuclein and TDP-43 with Alzheimer disease pathology (tau and Aβ) may complicate efforts to identify therapies to treat and prevent Alzheimer disease.
Collapse
Affiliation(s)
- Shama Karanth
- Department of Epidemiology, University of Kentucky, Lexington.,Sanders-Brown Center on Aging, University of Kentucky, Lexington
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington.,Department of Pathology, University of Kentucky, Lexington
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington.,Department of Biostatistics, University of Kentucky, Lexington
| | - Richard J Kryscio
- Sanders-Brown Center on Aging, University of Kentucky, Lexington.,Department of Biostatistics, University of Kentucky, Lexington.,Department of Statistics, University of Kentucky, Lexington
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, University of Kentucky, Lexington.,Department of Neurology, University of Kentucky, Lexington
| | - David W Fardo
- Sanders-Brown Center on Aging, University of Kentucky, Lexington.,Department of Biostatistics, University of Kentucky, Lexington
| | | | - Gregory A Jicha
- Sanders-Brown Center on Aging, University of Kentucky, Lexington.,Department of Neurology, University of Kentucky, Lexington
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, Lexington.,Department of Neuroscience, University of Kentucky, Lexington
| | - Erin L Abner
- Department of Epidemiology, University of Kentucky, Lexington.,Sanders-Brown Center on Aging, University of Kentucky, Lexington.,Department of Biostatistics, University of Kentucky, Lexington
| |
Collapse
|
40
|
Prodromal Parkinson disease subtypes - key to understanding heterogeneity. Nat Rev Neurol 2021; 17:349-361. [PMID: 33879872 DOI: 10.1038/s41582-021-00486-9] [Citation(s) in RCA: 180] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2021] [Indexed: 02/04/2023]
Abstract
In Parkinson disease (PD), pathological processes and neurodegeneration begin long before the cardinal motor symptoms develop and enable clinical diagnosis. In this prodromal phase, risk and prodromal markers can be used to identify individuals who are likely to develop PD, as in the recently updated International Parkinson and Movement Disorders Society research criteria for prodromal PD. However, increasing evidence suggests that clinical and prodromal PD are heterogeneous, and can be classified into subtypes with different clinical manifestations, pathomechanisms and patterns of spatial and temporal progression in the CNS and PNS. Genetic, pathological and imaging markers, as well as motor and non-motor symptoms, might define prodromal subtypes of PD. Moreover, concomitant pathology or other factors, including amyloid-β and tau pathology, age and environmental factors, can cause variability in prodromal PD. Patients with REM sleep behaviour disorder (RBD) exhibit distinct patterns of α-synuclein pathology propagation and might indicate a body-first subtype rather than a brain-first subtype. Identification of prodromal PD subtypes and a full understanding of variability at this stage of the disease is crucial for early and accurate diagnosis and for targeting of neuroprotective interventions to ensure efficacy.
Collapse
|
41
|
Chatzikonstantinou S, McKenna J, Karantali E, Petridis F, Kazis D, Mavroudis I. Electroencephalogram in dementia with Lewy bodies: a systematic review. Aging Clin Exp Res 2021; 33:1197-1208. [PMID: 32383032 DOI: 10.1007/s40520-020-01576-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/21/2020] [Indexed: 01/26/2023]
Abstract
Dementia with Lewy bodies (DLB) belongs to the spectrum of Lewy body dementia (LBD) that also encompasses Parkinson's disease dementia (PDD). It is a common neurodegenerative disorder characterized by memory decline, cognitive fluctuations, visual hallucinations, autonomic nervous system disturbance, REM sleep behavior disorder, and parkinsonism. Definite diagnosis can be established only through neuropathological confirmation of Lewy bodies' presence in brain tissue. Probable or possible diagnosis relies upon clinical features, imaging, polysomnography, and electroencephalogram (EEG) findings. Potential neurophysiological biomarkers for the diagnosis, management, and evaluation of treatment-response in DLB should be affordable and widely available outside academic centers. Increasing evidence supports the use of quantitative EEG (qEEG) as a potential DLB biomarker, with promising results in discriminating DLB from other dementias and in identifying subjects who are on the trajectory to develop DLB. Several studies evaluated the diagnostic value of EEG in DLB. Visual analysis and qEEG techniques have been implemented, showing a superiority of the last in terms of sensitivity and objectivity. In this systematic review, we attempt to provide a general synthesis of the current knowledge on EEG application in DLB. We review the findings from original studies and address the issues remaining to be further clarified.
Collapse
Affiliation(s)
- Simela Chatzikonstantinou
- Third Department of Neurology, Aristotle University of Thessaloniki, 3 Arsaki Street, Pefka, 57010, Thessaloníki, Greece.
| | | | - Eleni Karantali
- Third Department of Neurology, Aristotle University of Thessaloniki, 3 Arsaki Street, Pefka, 57010, Thessaloníki, Greece
| | - Fivos Petridis
- Third Department of Neurology, Aristotle University of Thessaloniki, 3 Arsaki Street, Pefka, 57010, Thessaloníki, Greece
| | - Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, 3 Arsaki Street, Pefka, 57010, Thessaloníki, Greece
| | - Ioannis Mavroudis
- Leeds Teaching Hospitals, Leeds, UK
- Medical School, Cyprus University, Nicosia, Cyprus
| |
Collapse
|
42
|
Jellinger KA. Significance of cerebral amyloid angiopathy and other co-morbidities in Lewy body diseases. J Neural Transm (Vienna) 2021; 128:687-699. [PMID: 33928445 DOI: 10.1007/s00702-021-02345-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/22/2021] [Indexed: 01/12/2023]
Abstract
Lewy body dementia (LBD) and Parkinson's disease-dementia (PDD) are two major neurocognitive disorders with Lewy bodies (LB) of unknown etiology. There is considerable clinical and pathological overlap between these two conditions that are clinically distinguished based on the duration of Parkinsonism prior to development of dementia. Their morphology is characterized by a variable combination of LB and Alzheimer's disease (AD) pathologies. Cerebral amyloid angiopathy (CAA), very common in aged persons and particularly in AD, is increasingly recognized for its association with both pathologies and dementia. To investigate neuropathological differences between LB diseases with and without dementia, 110 PDD and 60 LBD cases were compared with 60 Parkinson's disease (PD) cases without dementia (PDND). The major demographic and neuropathological data were assessed retrospectively. PDD patients were significantly older than PDND ones (83.9 vs 77.8 years; p < 0.05); the age of LB patients was in between both groups (mean 80.2 years), while the duration of disease was LBD < PDD < PDND (mean 6.7 vs 12.5 and 14.3 years). LBD patients had higher neuritic Braak stages (mean 5.1 vs 4.5 and 4.0, respectively), LB scores (mean 5.3 vs 4.2 and 4.0, respectively), and Thal amyloid phases (mean 4.1 vs 3.0 and 2.3, respectively) than the two other groups. CAA was more common in LBD than in the PDD and PDND groups (93 vs 50 and 21.7%, respectively). Its severity was significantly greater in LBD than in PDD and PDND (p < 0.01), involving mainly the occipital lobes. Moreover, striatal Aβ deposition highly differentiated LBD brains from PDD. Braak neurofibrillary tangle (NFT) stages, CAA, and less Thal Aβ phases were positively correlated with LB pathology (p < 0.05), which was significantly higher in LBD than in PDD < PDND. Survival analysis showed worse prognosis in LBD than in PDD (and PDND), which was linked to both increased Braak tau stages and more severe CAA. These and other recent studies imply the association of CAA-and both tau and LB pathologies-with cognitive decline and more rapid disease progression that distinguishes LBD from PDD (and PDND).
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
43
|
Gupta HV, Beach TG, Mehta SH, Shill HA, Driver-Dunckley E, Sabbagh MN, Belden CM, Liebsack C, Dugger BN, Serrano GE, Sue LI, Siderowf A, Pontecorvo MJ, Mintun MA, Joshi AD, Adler CH. Clinicopathological Correlation: Dopamine and Amyloid PET Imaging with Neuropathology in Three Subjects Clinically Diagnosed with Alzheimer's Disease or Dementia with Lewy Bodies. J Alzheimers Dis 2021; 80:1603-1612. [PMID: 33720879 PMCID: PMC10109539 DOI: 10.3233/jad-200323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Imaging biomarkers have the potential to distinguish between different brain pathologies based on the type of ligand used with PET. AV-45 PET (florbetapir, Amyvid™) is selective for the neuritic plaque amyloid of Alzheimer's disease (AD), while AV-133 PET (florbenazine) is selective for VMAT2, which is a dopaminergic marker. OBJECTIVE To report the clinical, AV-133 PET, AV-45 PET, and neuropathological findings of three clinically diagnosed dementia patients who were part of the Avid Radiopharmaceuticals AV133-B03 study as well as the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND). METHODS Three subjects who had PET imaging with both AV-133 and AV-45 as well as a standardized neuropathological assessment were included. The final clinical, PET scan, and neuropathological diagnoses were compared. RESULTS The clinical and neuropathological diagnoses were made blinded to PET scan results. The first subject had a clinical diagnosis of dementia with Lewy bodies (DLB); AV-133 PET showed bilateral striatal dopaminergic degeneration, and AV-45 PET was positive for amyloid. The final clinicopathological diagnosis was DLB and AD. The second subject was diagnosed clinically with probable AD; AV-45 PET was positive for amyloid, while striatal AV-133 PET was normal. The final clinicopathological diagnosis was DLB and AD. The third subject had a clinical diagnosis of DLB. Her AV-45 PET was positive for amyloid and striatal AV-133 showed dopaminergic degeneration. The final clinicopathological diagnosis was multiple system atrophy and AD. CONCLUSION PET imaging using AV-133 for the assessment of striatal VMAT2 density may help distinguish between AD and DLB. However, some cases of DLB with less-pronounced nigrostriatal dopaminergic neuronal loss may be missed.
Collapse
Affiliation(s)
- Harsh V Gupta
- Department of Neurology, The University of Kansas Health System, Kansas City, KS, USA
| | | | - Shyamal H Mehta
- Department of Neurology, Mayo Clinic College of Medicine, Scottsdale, AZ, USA
| | | | | | | | | | | | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, University of California-Davis School of Medicine, Sacramento, CA, USA
| | | | - Lucia I Sue
- Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Andrew Siderowf
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Charles H Adler
- Department of Neurology, Mayo Clinic College of Medicine, Scottsdale, AZ, USA
| |
Collapse
|
44
|
Combi R, Salsone M, Villa C, Ferini-Strambi L. Genetic Architecture and Molecular, Imaging and Prodromic Markers in Dementia with Lewy Bodies: State of the Art, Opportunities and Challenges. Int J Mol Sci 2021; 22:3960. [PMID: 33921279 PMCID: PMC8069386 DOI: 10.3390/ijms22083960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/03/2021] [Accepted: 04/09/2021] [Indexed: 11/16/2022] Open
Abstract
Dementia with Lewy bodies (DLB) is one of the most common causes of dementia and belongs to the group of α-synucleinopathies. Due to its clinical overlap with other neurodegenerative disorders and its high clinical heterogeneity, the clinical differential diagnosis of DLB from other similar disorders is often difficult and it is frequently underdiagnosed. Moreover, its genetic etiology has been studied only recently due to the unavailability of large cohorts with a certain diagnosis and shows genetic heterogeneity with a rare contribution of pathogenic mutations and relatively common risk factors. The rapid increase in the reported cases of DLB highlights the need for an easy, efficient and accurate diagnosis of the disease in its initial stages in order to halt or delay the progression. The currently used diagnostic methods proposed by the International DLB consortium rely on a list of criteria that comprises both clinical observations and the use of biomarkers. Herein, we summarize the up-to-now reported knowledge on the genetic architecture of DLB and discuss the use of prodromal biomarkers as well as recent promising candidates from alternative body fluids and new imaging techniques.
Collapse
Affiliation(s)
- Romina Combi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Maria Salsone
- Institute of Molecular Bioimaging and Physiology, National Research Council, 20054 Segrate (MI), Italy;
- Department of Clinical Neurosciences, Neurology-Sleep Disorder Center, IRCCS San Raffaele Scientific Institute, 20127 Milan, Italy
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Luigi Ferini-Strambi
- Department of Clinical Neurosciences, Neurology-Sleep Disorder Center, IRCCS San Raffaele Scientific Institute, 20127 Milan, Italy
- Department of Clinical Neurosciences, “Vita-Salute” San Raffaele University, 20127 Milan, Italy
| |
Collapse
|
45
|
Characterization of symptoms and determinants of disease burden in dementia with Lewy bodies: DEvELOP design and baseline results. ALZHEIMERS RESEARCH & THERAPY 2021; 13:53. [PMID: 33637117 PMCID: PMC7908769 DOI: 10.1186/s13195-021-00792-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/16/2021] [Indexed: 01/28/2023]
Abstract
BACKGROUND The DEmEntia with LEwy bOdies Project (DEvELOP) aims to phenotype patients with dementia with Lewy bodies (DLB) and study the symptoms and biomarkers over time. Here, we describe the design and baseline results of DEvELOP. We investigated the associations between core and suggestive DLB symptoms and different aspects of disease burden, i.e., instrumental activities of daily living (IADL) functioning, quality of life (QoL), and caregiver burden. METHODS We included 100 DLB patients (69 ± 6 years, 10%F, MMSE 25 ± 3) in the prospective DEvELOP cohort. Patients underwent extensive assessment including MRI, EEG/MEG, 123FP-CIT SPECT, and CSF and blood collection, with annual follow-up. Core (hallucinations, parkinsonism, fluctuations, RBD) and suggestive (autonomous dysfunction, neuropsychiatric symptoms) symptoms were assessed using standardized questionnaires. We used multivariate regression analyses, adjusted for age, sex, and MMSE, to evaluate how symptoms related to the Functional Activities Questionnaire, QoL-AD questionnaire, and Zarit Caregiver Burden Interview. RESULTS In our cohort, RBD was the most frequently reported core feature (75%), while visual hallucinations were least frequently reported (39%) and caused minimal distress. Suggestive clinical features were commonly present, of which orthostatic hypotension was most frequently reported (64%). Ninety-five percent of patients showed EEG/MEG abnormalities, 88% of 123FP-CIT SPECT scans were abnormal, and 53% had a CSF Alzheimer's disease profile. Presence of fluctuations, lower MMSE, parkinsonism, and apathy were associated with higher IADL dependency. Depression, constipation, and lower IADL were associated with lower QoL-AD. Apathy and higher IADL dependency predisposed for higher caregiver burden. CONCLUSION Baseline data of our prospective DLB cohort show clinically relevant associations between symptomatology and disease burden. Cognitive and motor symptoms are related to IADL functioning, while negative neuropsychiatric symptoms and functional dependency are important determinants of QoL and caregiver burden. Follow-up is currently ongoing to address specific gaps in DLB research.
Collapse
|
46
|
Nichols JB, Malek-Ahmadi M, Tariot PN, Serrano GE, Sue LI, Beach TG. Vascular Lesions, APOE ε4, and Tau Pathology in Alzheimer Disease. J Neuropathol Exp Neurol 2021; 80:240-246. [PMID: 33617650 PMCID: PMC7899190 DOI: 10.1093/jnen/nlaa160] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We sought to determine the associations among cerebral amyloid angiopathy (CAA), white matter rarefaction (WMR), circle of Willis atherosclerosis (CWA), and total microinfarct number with Braak neurofibrillary stage in postmortem individuals with and without Alzheimer disease (AD). Data from 355 cases of autopsied individuals with Braak stage I-VI who had antemortem consensus diagnoses of cognitively unimpaired (n = 183), amnestic mild cognitive impairment (n = 31), and AD dementia (n = 141) were used. The association between Braak stage and vascular lesions were individually assessed using multivariable linear regression that adjusted for age at death, APOE ε4 carrier status, sex, education, and neuritic plaque density. CAA (p = 0.007) and WMR (p < 0.001) were associated with Braak stage, independent of amyloid load; microinfarct number and CWA showed no association. Analyses of the interactions between APOE ε4 carrier status and vascular lesions found that greater WMR and positive ε4 carrier status were associated with higher Braak stages. These results suggest that CAA and WMR are statistically linked to the severity of AD-related NFT pathology. The statistical link between WMR and NFT load may be strengthened by the presence of APOE ε4 carrier status. An additional finding was that Lewy body pathology was most prevalent in higher Braak stages.
Collapse
Affiliation(s)
- Jodie B Nichols
- From the Arizona College of Osteopathic Medicine, Midwestern University, Glendale, Arizona, USA
| | | | | | - Geidy E Serrano
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Lucia I Sue
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Thomas G Beach
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| |
Collapse
|
47
|
Wolters EE, van de Beek M, Ossenkoppele R, Golla SSV, Verfaillie SCJ, Coomans EM, Timmers T, Visser D, Tuncel H, Barkhof F, Boellaard R, Windhorst AD, van der Flier WM, Scheltens P, Lemstra AW, van Berckel BNM. Tau PET and relative cerebral blood flow in dementia with Lewy bodies: A PET study. Neuroimage Clin 2020; 28:102504. [PMID: 33395993 PMCID: PMC7714680 DOI: 10.1016/j.nicl.2020.102504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/08/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Alpha-synuclein often co-occurs with Alzheimer's disease (AD) pathology in Dementia with Lewy Bodies (DLB). From a dynamic [18F]flortaucipir PET scan we derived measures of both tau binding and relative cerebral blood flow (rCBF). We tested whether regional tau binding or rCBF differed between DLB patients and AD patients and controls and examined their association with clinical characteristics of DLB. METHODS Eighteen patients with probable DLB, 65 AD patients and 50 controls underwent a dynamic 130-minute [18F]flortaucipir PET scan. DLB patients with positive biomarkers for AD based on cerebrospinal fluid or amyloid PET were considered as DLB with AD pathology (DLB-AD+). Receptor parametric mapping (cerebellar gray matter reference region) was used to extract regional binding potential (BPND) and R1, reflecting (AD-specific) tau pathology and rCBF, respectively. First, we performed regional comparisons of [18F]flortaucipir BPND and R1 between diagnostic groups. In DLB patients only, we performed regression analyses between regional [18F]flortaucipir BPND, R1 and performance on ten neuropsychological tests. RESULTS Regional [18F]flortaucipir BPND in DLB was comparable with tau binding in controls (p > 0.05). Subtle higher tau binding was observed in DLB-AD+ compared to DLB-AD- in the medial temporal and parietal lobe (both p < 0.05). Occipital and lateral parietal R1 was lower in DLB compared to AD and controls (all p < 0.01). Lower frontal R1 was associated with impaired performance on digit span forward (standardized beta, stβ = 0.72) and category fluency (stβ = 0.69) tests. Lower parietal R1 was related to lower delayed (stβ = 0.50) and immediate (stβ = 0.48) recall, VOSP number location (stβ = 0.70) and fragmented letters (stβ = 0.59) scores. Lower occipital R1 was associated to worse performance on VOSP fragmented letters (stβ = 0.61), all p < 0.05. CONCLUSION The amount of tau binding in DLB was minimal and did not differ from controls. However, there were DLB-specific occipital and lateral parietal relative cerebral blood flow reductions compared to both controls and AD patients. Regional rCBF, but not tau binding, was related to cognitive impairment. This indicates that assessment of rCBF may give more insight into disease mechanisms in DLB than tau PET.
Collapse
Affiliation(s)
- E E Wolters
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - M van de Beek
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - R Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - S S V Golla
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - S C J Verfaillie
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - E M Coomans
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - T Timmers
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - D Visser
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - H Tuncel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - F Barkhof
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Institutes of Neurology & Healthcare Engineering, UCL, London, United Kingdom
| | - R Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - A D Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - W M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ph Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - A W Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - B N M van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Ghirelli A, Tosakulwong N, Weigand SD, Clark HM, Ali F, Botha H, Duffy JR, Utianski RL, Buciuc M, Murray ME, Labuzan SA, Spychalla AJ, Pham NTT, Schwarz CG, Senjem ML, Machulda MM, Baker M, Rademakers R, Filippi M, Jack CR, Lowe VJ, Parisi JE, Dickson DW, Josephs KA, Whitwell JL. Sensitivity-Specificity of Tau and Amyloid β Positron Emission Tomography in Frontotemporal Lobar Degeneration. Ann Neurol 2020; 88:1009-1022. [PMID: 32869362 PMCID: PMC7861121 DOI: 10.1002/ana.25893] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To examine associations between tau and amyloid β (Aβ) molecular positron emission tomography (PET) and both Alzheimer-related pathology and 4-repeat tau pathology in autopsy-confirmed frontotemporal lobar degeneration (FTLD). METHODS Twenty-four patients had [18 F]-flortaucipir-PET and died with FTLD (progressive supranuclear palsy [PSP], n = 10; corticobasal degeneration [CBD], n = 10; FTLD-TDP, n = 3; and Pick disease, n = 1). All but 1 had Pittsburgh compound B (PiB)-PET. Braak staging, Aβ plaque and neurofibrillary tangle counts, and semiquantitative tau lesion scores were performed. Flortaucipir standard uptake value ratios (SUVRs) were calculated in a temporal meta region of interest (meta-ROI), entorhinal cortex and cortical/subcortical regions selected to match the tau lesion analysis. Global PiB SUVR was calculated. Autoradiography was performed in 1 PSP patient, with digital pathology used to quantify tau burden. RESULTS Nine cases (37.5%) had Aβ plaques. Global PiB SUVR correlated with Aβ plaque count, with 100% specificity and 50% sensitivity for diffuse plaques. Twenty-one (87.5%) had Braak stages I to IV. Flortaucipir correlated with neurofibrillary tangle counts in entorhinal cortex, but entorhinal and meta-ROI SUVRs were not elevated in Braak IV or primary age-related tauopathy. Flortaucipir uptake patterns differed across FTLD pathologies and could separate PSP and CBD. Flortaucipir correlated with tau lesion score in red nucleus and midbrain tegmentum across patients, but not in cortical or basal ganglia regions. Autoradiography demonstrated minimal uptake of flortaucipir, although flortaucipir correlated with quantitative tau burden across regions. INTERPRETATION Molecular PET shows expected correlations with Alzheimer-related pathology but lacks sensitivity to detect mild Alzheimer pathology in FTLD. Regional flortaucipir uptake was able to separate CBD and PSP. ANN NEUROL 2020;88:1009-1022.
Collapse
Affiliation(s)
- Alma Ghirelli
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
- Università Vita-Salute San Raffaele, Milan,
Italy
| | | | | | | | - Farwa Ali
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN,
USA
| | | | | | - Marina Buciuc
- 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
| | - Mary M. Machulda
- Department of Psychiatry and Psychology, Mayo Clinic,
Rochester, MN, USA
| | - Matthew Baker
- Department of Health Sciences Research, Mayo Clinic,
Rochester, MN, USA
| | - Rosa Rademakers
- Department of Health Sciences Research, Mayo Clinic,
Rochester, MN, USA
| | - Massimo Filippi
- Università Vita-Salute San Raffaele, Milan,
Italy
- Neurology and Neurophysiology Units, and Neuroimaging
Research Unit, INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific
Institute, Milan, Italy
| | | | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN,
USA
| | - Joseph E Parisi
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, MN, USA
| | | | | | | |
Collapse
|
49
|
Williams T, Sorrentino Z, Weinrich M, Giasson BI, Chakrabarty P. Differential cross-seeding properties of tau and α-synuclein in mouse models of tauopathy and synucleinopathy. Brain Commun 2020; 2:fcaa090. [PMID: 33094280 PMCID: PMC7567170 DOI: 10.1093/braincomms/fcaa090] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/01/2020] [Accepted: 06/08/2020] [Indexed: 11/17/2022] Open
Abstract
Co-occurrence of tau and α-synuclein pathologies in a subset of Alzheimer’s disease patients has led to the idea that mixed pathologies may play a unique characteristic role in the Alzheimer’s disease neurodegenerative cascade. To understand the aetiology of such mixed pathologies, we investigated cross-seeding by human recombinant tau and human recombinant α-synuclein fibrillar species in a mouse model of tauopathy (Line PS19) or synucleinopathy (Line M20). Unilateral hippocampal injection of tau fibrils or α-synuclein fibrils, and to a lesser extent tau + α-synuclein copolymer fibrils prepared from co-incubating individual recombinant monomers, induced robust phosphorylated tau pathology in PS19 mice relative to control mice. Though the tau + α-synuclein copolymer fibrils did not modulate induction of pathologies at the site of injection, examination of the whole brain showed that these copolymers exacerbated neuroanatomic transmission of seeded tau pathology compared to tau fibril-injected mice. Only α-synuclein fibrils, but not tau alone or tau + α-synuclein copolymers, triggered modest levels of endogenous phosphorylated α-synuclein pathology. Overall, data from the PS19 mice suggest that human α-synuclein fibrils can efficiently cross-seed human tau and have a modest priming effect on mouse α-synuclein, and the presence of tau fibrils does not exacerbate the priming process. In M20 mice, unilateral hippocampal injection of α-synuclein fibrils or tau fibrils induced robust bilateral phosphorylated α-synuclein pathology, while tau + α-synuclein copolymer injection resulted in restricted phosphorylated α-synuclein pathology predominantly in the ipsilateral cortex. This suggests that human tau fibrils can also induce human α-synuclein pathogenesis, and the presence of combinatorial seeds is not synergistic. None of these aggregates induced phosphorylated tau pathology in M20 mice, showing that mouse tau cannot be primed efficiently by human tau fibrils or human α-synuclein fibrils. Neuropathological analysis of the whole brain of M20 mice showed that tau + α-synuclein copolymer-injected mice had lower abundance of bilaterally transmitted α-synuclein pathologies relative to α-synuclein fibril-injected mice. Thus, the tau + α-synuclein copolymer fibrils show robust transmission properties preferentially in rodent model of tauopathies but not in synucleinopathy, probably signifying an enhanced cooperative relationship between tau and α-synuclein in the tau seeding process. Together, our data highlight the unique cross-seeding properties of tau and αSyn in neurodegenerative proteinopathies.
Collapse
Affiliation(s)
- Tosha Williams
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
| | - Zachary Sorrentino
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
| | - Mary Weinrich
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA.,Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA.,Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA.,Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
50
|
The combined effect of amyloid-β and tau biomarkers on brain atrophy in dementia with Lewy bodies. NEUROIMAGE-CLINICAL 2020; 27:102333. [PMID: 32674011 PMCID: PMC7363702 DOI: 10.1016/j.nicl.2020.102333] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 06/05/2020] [Accepted: 06/26/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Alzheimer's disease (AD)-related pathology is frequently found in patients with dementia with Lewy bodies (DLB). However, it is unknown how amyloid-β and tau-related pathologies influence neurodegeneration in DLB. Understanding the mechanisms underlying brain atrophy in DLB can improve our knowledge about disease progression, differential diagnosis, drug development and testing of anti-amyloid and anti-tau therapies in DLB. OBJECTIVES We aimed at investigating the combined effect of CSF amyloid-β42, phosphorylated tau and total tau on regional brain atrophy in DLB in the European DLB (E-DLB) cohort. METHODS 86 probable DLB patients from the E-DLB cohort with CSF and MRI data were included. Random forest was used to analyze the association of CSF biomarkers (predictors) with visual rating scales for medial temporal lobe atrophy (MTA), posterior atrophy (PA) and global cortical atrophy scale-frontal subscale (GCA-F) (outcomes), including age, sex, education and disease duration as extra predictors. RESULTS DLB patients with abnormal MTA scores had abnormal CSF Aβ42, shorter disease duration and older age. DLB patients with abnormal PA scores had abnormal levels of CSF Aβ42 and p-tau, older age, lower education and shorter disease duration. Abnormal GCA-F scores were associated with lower education, male sex, and older age, but not with any AD-related CSF biomarker. CONCLUSIONS This study shows preliminary data on the potential combined effect of amyloid-β and tau-related pathologies on the integrity of posterior brain cortices in DLB patients, whereas only amyloid-β seems to be related to MTA. Future availability of α-synuclein biomarkers will help us to understand the effect of α-synuclein and AD-related pathologies on brain integrity in DLB.
Collapse
|