151
|
Ferreira D, Mohanty R, Murray ME, Nordberg A, Kantarci K, Westman E. The hippocampal sparing subtype of Alzheimer's disease assessed in neuropathology and in vivo tau positron emission tomography: a systematic review. Acta Neuropathol Commun 2022; 10:166. [PMID: 36376963 PMCID: PMC9664780 DOI: 10.1186/s40478-022-01471-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/30/2022] [Indexed: 11/16/2022] Open
Abstract
Neuropathology and neuroimaging studies have identified several subtypes of Alzheimer's disease (AD): hippocampal sparing AD, typical AD, and limbic predominant AD. An unresolved question is whether hippocampal sparing AD cases can present with neurofibrillary tangles (NFT) in association cortices while completely sparing the hippocampus. To address that question, we conducted a systematic review and performed original analyses on tau positron emission tomography (PET) data. We searched EMBASE, PubMed, and Web of Science databases until October 2022. We also implemented several methods for AD subtyping on tau PET to identify hippocampal sparing AD cases. Our findings show that seven out of the eight reviewed neuropathologic studies included cases at Braak stages IV or higher and therefore, could not identify hippocampal sparing cases with NFT completely sparing the hippocampus. In contrast, tau PET did identify AD participants with tracer retention in the association cortex while completely sparing the hippocampus. We conclude that tau PET can identify hippocampal sparing AD cases with NFT completely sparing the hippocampus. Based on the accumulating data, we suggest two possible pathways of tau spread: (1) a canonical pathway with early involvement of transentorhinal cortex and subsequent involvement of limbic regions and association cortices, and (2) a less common pathway that affects association cortices with limbic involvement observed at end stages of the disease or not at all.
Collapse
Affiliation(s)
- Daniel Ferreira
- Division of Clinical Geriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 (NEO building, floor 7th), 14152, Huddinge, Stockholm, Sweden.
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| | - Rosaleena Mohanty
- Division of Clinical Geriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 (NEO building, floor 7th), 14152, Huddinge, Stockholm, Sweden
| | | | - Agneta Nordberg
- Division of Clinical Geriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 (NEO building, floor 7th), 14152, Huddinge, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Eric Westman
- Division of Clinical Geriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 (NEO building, floor 7th), 14152, Huddinge, Stockholm, Sweden.
- Department of Neuroimaging, Center for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
152
|
Gietl AF, Frisoni GB. Early termination of pivotal trials in Alzheimer's disease-Preserving optimal value for participants and science. Alzheimers Dement 2022; 18:1980-1987. [PMID: 35220681 PMCID: PMC9790521 DOI: 10.1002/alz.12605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 12/14/2021] [Accepted: 01/03/2022] [Indexed: 01/28/2023]
Abstract
Participants in Alzheimer's disease late-phase clinical trials are frequently confronted with a situation of early termination. We discuss measures to protect the perceived value of study participation and to maximize the scientific value under such circumstances. A communication strategy should ensure that trial participants maintain a positive relationship with the research team and have their informational needs optimally met. Measures to maximize the scientific value may include data/sample sharing, strategies for personalized medicine, as well as scientific follow-up. Critical for the success of such a concept are networks of excellence, extending models of existing initiatives like Global Alzheimer's Platform Foundation Network (GAP-Net). These networks could fundamentally strengthen the role of clinical investigators if they decide on their involvement in trials based upon their estimation of the scientific value and benefit for the participants, actively contribute to scientific analyses, and mediate optimal communication among the relevant trial stakeholders.
Collapse
Affiliation(s)
- Anton F. Gietl
- Institute for Regenerative Medicine, Center for Prevention and Dementia TherapyUniversity of ZurichSchlierenSwitzerland,University Hospital for Geriatric PsychiatrySwitzerland
| | | |
Collapse
|
153
|
Niotis K, Akiyoshi K, Carlton C, Isaacson R. Dementia Prevention in Clinical Practice. Semin Neurol 2022; 42:525-548. [PMID: 36442814 DOI: 10.1055/s-0042-1759580] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Over 55 million people globally are living with dementia and, by 2050, this number is projected to increase to 131 million. This poses immeasurable challenges for patients and their families and a significant threat to domestic and global economies. Given this public health crisis and disappointing results from disease-modifying trials, there has been a recent shift in focus toward primary and secondary prevention strategies. Approximately 40% of Alzheimer's disease (AD) cases, which is the most common form of dementia, may be prevented or at least delayed. Success of risk reduction studies through addressing modifiable risk factors, in addition to the failure of most drug trials, lends support for personalized multidomain interventions rather than a "one-size-fits-all" approach. Evolving evidence supports early intervention in at-risk patients using individualized interventions directed at modifiable risk factors. Comprehensive risk stratification can be informed by emerging principals of precision medicine, and include expanded clinical and family history, anthropometric measurements, blood biomarkers, neurocognitive evaluation, and genetic information. Risk stratification is key in differentiating subtypes of dementia and identifies targetable areas for intervention. This article reviews a clinical approach toward dementia risk stratification and evidence-based prevention strategies, with a primary focus on AD.
Collapse
Affiliation(s)
- Kellyann Niotis
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York
| | - Kiarra Akiyoshi
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York
| | - Caroline Carlton
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York
| | - Richard Isaacson
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York.,Department of Neurology, Florida Atlantic University, Charles E. Schmidt College of Medicine, Boca Raton, Florida
| |
Collapse
|
154
|
Chen TB, Lee WJ, Chen JP, Chang SY, Lin CF, Chen HC. Imaging markers of cerebral amyloid angiopathy and hypertensive arteriopathy differentiate Alzheimer disease subtypes synergistically. Alzheimers Res Ther 2022; 14:141. [PMID: 36180874 PMCID: PMC9524061 DOI: 10.1186/s13195-022-01083-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022]
Abstract
Background Both cerebral amyloid angiopathy (CAA) and hypertensive arteriopathy (HA) are related to cognitive impairment and dementia. This study aimed to clarify CAA- and HA-related small vessel disease (SVD) imaging marker associations with cognitive dysfunction and Alzheimer disease (AD) subtypes. Methods A sample of 137 subjects with clinically diagnosed late-onset AD identified from the dementia registry of a single center from January 2017 to October 2021 were enrolled. Semi-quantitative imaging changes (visual rating scale grading) suggestive of SVD were analyzed singularly and compositely, and their correlations with cognitive domains and AD subtypes were examined. Results Patients with typical and limbic-predominant AD subtypes had worse cognitive performance and higher dementia severity than minimal-atrophy subtype patients. Deep white matter hyperintensity (WMH) presence correlated inversely with short-term memory (STM) performance. The three composite SVD scores correlated with different cognitive domains and had distinct associations with AD subtypes. After adjusting for relevant demographic factors, multivariate logistic regression (using minimal-atrophy subtype as the reference condition) revealed the following: associations of the typical subtype with periventricular WMH [odds ratio (OR) 2.62; 95% confidence interval (CI), 1.23–5.57, p = 0.012], global SVD score (OR 1.67; 95%CI, 1.11–2.52, p = 0.009), and HA-SVD score (OR 1.93; 95%CI, 1.10–3.52, p = 0.034); associations of limbic-predominant subtype with HA-SVD score (OR 2.57; 95%CI, 1.23–5.37, p = 0.012) and most global and domain-specific cognitive scores; and an association of hippocampal-sparing subtype with HA-SVD score (OR 3.30; 95%CI, 1.58–6.85, p = 0.001). Conclusion Composite SVD imaging markers reflect overall CAA and/or HA severity and may have differential associations with cognitive domains and AD subtypes. Our finding supports the possibility that the clinical AD subtypes may reflect differing burdens of underlying CAA and HA microangiopathologies. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-01083-8.
Collapse
|
155
|
Piccoli T, Blandino V, Maniscalco L, Matranga D, Graziano F, Guajana F, Agnello L, Lo Sasso B, Gambino CM, Giglio RV, La Bella V, Ciaccio M, Colletti T. Biomarkers Related to Synaptic Dysfunction to Discriminate Alzheimer's Disease from Other Neurological Disorders. Int J Mol Sci 2022; 23:10831. [PMID: 36142742 PMCID: PMC9501545 DOI: 10.3390/ijms231810831] [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: 07/26/2022] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Recently, the synaptic proteins neurogranin (Ng) and α-synuclein (α-Syn) have attracted scientific interest as potential biomarkers for synaptic dysfunction in neurodegenerative diseases. In this study, we measured the CSF Ng and α-Syn concentrations in patients affected by AD (n = 69), non-AD neurodegenerative disorders (n-AD = 50) and non-degenerative disorders (n-ND, n = 98). The concentrations of CSF Ng and α-Syn were significantly higher in AD than in n-AD and n-ND. Moreover, the Aβ42/Ng and Aβ42/α-Syn ratios showed statistically significant differences between groups and discriminated AD patients from n-AD patients, better than Ng or α-Syn alone. Regression analyses showed an association of higher Ng concentrations with MMSE < 24, pathological Aβ 42/40 ratios, pTau, tTau and the ApoEε4 genotype. Aβ 42/Ng was associated with MMSE < 24, an AD-related FDG-PET pattern, the ApoEε4 genotype, pathological Aβ 42 levels and Aβ 42/40 ratios, pTau, and tTau. Moreover, APO-Eε4 carriers showed higher Ng concentrations than non-carriers. Our results support the idea that the Aβ 42/Ng ratio is a reliable index of synaptic dysfunction/degeneration able to discriminate AD from other neurological conditions.
Collapse
Affiliation(s)
- Tommaso Piccoli
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| | - Valeria Blandino
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| | - Laura Maniscalco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Domenica Matranga
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy
| | - Fabiola Graziano
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| | - Fabrizio Guajana
- Unit of Neurology, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| | - Luisa Agnello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy
| | - Bruna Lo Sasso
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy
| | - Caterina Maria Gambino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy
| | - Rosaria Vincenza Giglio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy
- Department of Laboratory Medicine, University Hospital “P. Giaccone”, 90127 Palermo, Italy
| | - Vincenzo La Bella
- ALS Clinical Research Center and Laboratory of Neurochemistry, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| | - Marcello Ciaccio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, University of Palermo, 90127 Palermo, Italy
| | - Tiziana Colletti
- ALS Clinical Research Center and Laboratory of Neurochemistry, Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90129 Palermo, Italy
| |
Collapse
|
156
|
Contador J, Pérez-Millan A, Guillen N, Sarto J, Tort-Merino A, Balasa M, Falgàs N, Castellví M, Borrego-Écija S, Juncà-Parella J, Bosch B, Fernández-Villullas G, Ramos-Campoy O, Antonell A, Bargalló N, Sanchez-Valle R, Sala Llonch R, Lladó A. Sex differences in early-onset Alzheimer's disease. Eur J Neurol 2022; 29:3623-3632. [PMID: 36005384 DOI: 10.1111/ene.15531] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/10/2022] [Accepted: 08/13/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Sex is believed to drive heterogeneity in Alzheimer's disease (AD), although evidence in early-onset AD (<65 years, EOAD) is scarce. METHODS We included 62 EOAD patients and 44 healthy controls (HC) with cerebrospinal fluid (CSF) AD's core biomarkers and neurofilament light chain levels, neuropsychological assessment, and 3T-MRI. We measured cortical thickness (CTh) and hippocampal subfield volumes (HpS) using Freesurfer. Adjusted linear models were used to analyze sex-differences and the relationship between atrophy and cognition. RESULTS Compared to same-sex HC, female-EOAD showed greater cognitive impairment and broader atrophy burden than male-EOAD. In a direct female-EOAD and male-EOAD comparison, there were slight differences in temporal CTh, with no differences in cognition or HpS. CSF tau levels were higher in female-EOAD than in male-EOAD. Greater atrophy was associated with worse cognition in female-EOAD. CONCLUSIONS At diagnosis, there are sex-differences in the pattern of cognitive impairment, atrophy burden and CSF tau in EOAD, suggesting there is an influence of sex on pathology spreading and susceptibility to the disease in EOAD.
Collapse
Affiliation(s)
- José Contador
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Agnès Pérez-Millan
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences. Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Nuria Guillen
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Jordi Sarto
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Adrià Tort-Merino
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute
| | - Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute
| | - Magdalena Castellví
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Sergi Borrego-Écija
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Jordi Juncà-Parella
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Beatriz Bosch
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Guadalupe Fernández-Villullas
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Oscar Ramos-Campoy
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Anna Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Nuria Bargalló
- Image Diagnostic Centre Radiology Department, Hospital Clínic de Barcelona, Magnetic Resonance Image Core facility Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM., Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Roser Sala Llonch
- Institute of Neurosciences. Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, Spain.,Biomedical Imaging Group, Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
157
|
Wu R, Bao J, Kim M, Saykin AJ, Moore JH, Shen L. Mining High-Level Imaging Genetic Associations via Clustering AD Candidate Variants with Similar Brain Association Patterns. Genes (Basel) 2022; 13:1520. [PMID: 36140686 PMCID: PMC9498881 DOI: 10.3390/genes13091520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Brain imaging genetics examines associations between imaging quantitative traits (QTs) and genetic factors such as single nucleotide polymorphisms (SNPs) to provide important insights into the pathogenesis of Alzheimer's disease (AD). The individual level SNP-QT signals are high dimensional and typically have small effect sizes, making them hard to be detected and replicated. To overcome this limitation, this work proposes a new approach that identifies high-level imaging genetic associations through applying multigraph clustering to the SNP-QT association maps. Given an SNP set and a brain QT set, the association between each SNP and each QT is evaluated using a linear regression model. Based on the resulting SNP-QT association map, five SNP-SNP similarity networks (or graphs) are created using five different scoring functions, respectively. Multigraph clustering is applied to these networks to identify SNP clusters with similar association patterns with all the brain QTs. After that, functional annotation is performed for each identified SNP cluster and its corresponding brain association pattern. We applied this pipeline to an AD imaging genetic study, which yielded promising results. For example, in an association study between 54 AD SNPs and 116 amyloid QTs, we identified two SNP clusters with one responsible for amyloid beta clearances and the other regulating amyloid beta formation. These high-level findings have the potential to provide valuable insights into relevant genetic pathways and brain circuits, which can help form new hypotheses for more detailed imaging and genetics studies in independent cohorts.
Collapse
Affiliation(s)
- Ruiming Wu
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jingxuan Bao
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mansu Kim
- The Catholic University of Korea, Seoul 06591, Korea
| | | | | | - Li Shen
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
158
|
Cedres N, Aejmelaeus-Lindström A, Ekström I, Nordin S, Li X, Persson J, Olofsson JK. Subjective Impairments in Olfaction and Cognition Predict Dissociated Behavioral Outcomes. J Gerontol B Psychol Sci Soc Sci 2022; 78:1-9. [PMID: 36000774 PMCID: PMC9890914 DOI: 10.1093/geronb/gbac124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Self-rated subjective cognitive decline (SCD) and subjective olfactory impairment (SOI) are associated with objective cognitive decline and dementia. However, their relationship and co-occurrence is unknown. We aimed to (a) describe the occurrence of SOI, SCD and their overlap in the general population; (b) compare SOI and SCD in terms of longitudinal associations with corresponding objective olfactory and cognitive measures; and (c) describe how SOI and SCD may lead to distinct sensory and cognitive outcomes. METHODS Cognitively unimpaired individuals from the third wave of the Swedish population-based Betula study (n = 784, aged 35-90 years; 51% females) were split into self-rated SOI, SCD, overlapping SCD + SOI, and controls. Between-subject and within-subject repeated-measures MANCOVA were used to compare the groups regarding odor identification, cognition, age, sex, and education. Spearman correlation was used to assess the different patterns of association between olfaction and cognition across groups. RESULTS SOI was present in 21.1%, whereas SCD was present in 9.9% of participants. According to a chi-square analysis, the SCD + SOI overlap (2.7%) is on a level that could be expected if the phenomena were independent. Odor identification in SOI showed decline at the 10-year follow-up (n = 284) and was positively associated with cognition. The SOI and SCD groups showed distinct cognitive-olfactory profiles at follow-up. CONCLUSIONS SOI occur independently of SCD in the population, and these risk factors are associated with different cognitive and olfactory outcomes. The biological causes underlying SOI and SCD, as well as the risk for future cognitive impairment, need further investigation.
Collapse
Affiliation(s)
- Nira Cedres
- Address correspondence to: Nira Cedres, PhD, Department of Psychology, Sensory Cognitive Interaction Laboratory (SCI-Lab), Stockholm University, Albanovägen 12, 114 19 Stockholm, Sweden. E-mail:
| | - Andrea Aejmelaeus-Lindström
- Department of Psychology, Sensory Cognitive Interaction Laboratory (SCI-Lab), Stockholm University, Stockholm, Sweden
| | - Ingrid Ekström
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Steven Nordin
- Department of Psychology, Umeå University, Umeå, Sweden
| | - Xin Li
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Persson
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Jonas K Olofsson
- Department of Psychology, Sensory Cognitive Interaction Laboratory (SCI-Lab), Stockholm University, Stockholm, Sweden
| |
Collapse
|
159
|
Kato T, Nishita Y, Otsuka R, Inui Y, Nakamura A, Kimura Y, Ito K. Effect of cognitive reserve on amnestic mild cognitive impairment due to Alzheimer’s disease defined by fluorodeoxyglucose-positron emission tomography. Front Aging Neurosci 2022; 14:932906. [PMID: 36034127 PMCID: PMC9399434 DOI: 10.3389/fnagi.2022.932906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
This study aimed to investigate the effect of cognitive reserve (CR) on the rate of cognitive decline and cerebral glucose metabolism in amnestic mild cognitive impairment (MCI) using the Study on Diagnosis of Early Alzheimer’s Disease-Japan (SEAD-J) dataset. The patients in SEAD-J underwent cognitive tests and fluorodeoxyglucose-positron emission tomography (FDG-PET). MCI to be studied was classified as amnestic MCI due to Alzheimer’s disease (AD) with neurodegeneration. A total of 57 patients were visually interpreted as having an AD pattern (P1 pattern, Silverman’s classification). The 57 individuals showing the P1 pattern were divided into a high-education group (years of school education ≥13, N = 18) and a low-education group (years of school education ≤12, N = 39). Voxel-based statistical parametric mapping revealed more severe hypometabolism in the high-education group than in the low-education group. Glucose metabolism in the hippocampus and temporoparietal area was inversely associated with the years of school education in the high- and low-education groups (N = 57). General linear mixed model analyses demonstrated that cognitive decline was more rapid in the high-education group during 3-year follow-up. These results suggest that the cerebral glucose metabolism is lower and cognitive function declines faster in patients with high CR of amnestic MCI due to AD defined by FDG-PET.
Collapse
Affiliation(s)
- Takashi Kato
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Aichi, Japan
- *Correspondence: Takashi Kato,
| | - Yukiko Nishita
- Department of Epidemiology of Aging, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Rei Otsuka
- Department of Epidemiology of Aging, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Yoshitaka Inui
- Department of Radiology, Fujita Health University School of Medicine, Aichi, Japan
| | - Akinori Nakamura
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Aichi, Japan
- Department of Biomarker Research, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Yasuyuki Kimura
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Kengo Ito
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Aichi, Japan
| | - SEAD-J Study GroupFukuyamaHidenaoSendaMichioIshiiKenjiIshiiKazunariMaedaKiyoshiYamamotoYasujiOuchiYasuomiOkamuraAyumuArahataYutakaWashimiYukihikoMeguroKenichiIkedaMitsuruKyoto University, Kyoto, Japan; Institute of Biomedical Research and Innovation, Kobe, Japan; Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan; Kindai University, Osaka, Japan; Kobe Gakuin University, Kobe, Japan; Kobe University Graduate School of Medicine, Kobe, Japan; Hamamatsu University School of Medicine, Hamamatsu, Japan; Kizawa Memorial Hospital, Gifu, Japan; National Center for Geriatrics and Gerontology, Aichi, Japan; National Center for Geriatrics and Gerontology, Aichi, Japan; Tohoku University Graduate School of Medicine, Sendai, Japan; Nagoya University School of Health Sciences, Nagoya, Japan
| |
Collapse
|
160
|
Poulakis K, Pereira JB, Muehlboeck JS, Wahlund LO, Smedby Ö, Volpe G, Masters CL, Ames D, Niimi Y, Iwatsubo T, Ferreira D, Westman E. Multi-cohort and longitudinal Bayesian clustering study of stage and subtype in Alzheimer's disease. Nat Commun 2022; 13:4566. [PMID: 35931678 PMCID: PMC9355993 DOI: 10.1038/s41467-022-32202-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/18/2022] [Indexed: 11/08/2022] Open
Abstract
Understanding Alzheimer's disease (AD) heterogeneity is important for understanding the underlying pathophysiological mechanisms of AD. However, AD atrophy subtypes may reflect different disease stages or biologically distinct subtypes. Here we use longitudinal magnetic resonance imaging data (891 participants with AD dementia, 305 healthy control participants) from four international cohorts, and longitudinal clustering to estimate differential atrophy trajectories from the age of clinical disease onset. Our findings (in amyloid-β positive AD patients) show five distinct longitudinal patterns of atrophy with different demographical and cognitive characteristics. Some previously reported atrophy subtypes may reflect disease stages rather than distinct subtypes. The heterogeneity in atrophy rates and cognitive decline within the five longitudinal atrophy patterns, potentially expresses a complex combination of protective/risk factors and concomitant non-AD pathologies. By alternating between the cross-sectional and longitudinal understanding of AD subtypes these analyses may allow better understanding of disease heterogeneity.
Collapse
Affiliation(s)
- Konstantinos Poulakis
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Joana B Pereira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmo, Sweden
| | - J-Sebastian Muehlboeck
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Lars-Olof Wahlund
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Örjan Smedby
- Department of Biomedical Engineering and Health Systems (MTH), KTH Royal Institute of Technology, Stockholm, Sweden
| | - Giovanni Volpe
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria, Australia
| | - David Ames
- Academic Unit for Psychiatry of Old Age, St George's Hospital, University of Melbourne, Melbourne, Victoria, Australia
- National Ageing Research Institute, Parkville, Victoria, Australia
| | - Yoshiki Niimi
- Unit for Early and Exploratory Clinical Development, The University of Tokyo Hospital, Tokyo, Japan
| | - Takeshi Iwatsubo
- Unit for Early and Exploratory Clinical Development, The University of Tokyo Hospital, Tokyo, Japan
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
161
|
Park J, Barahona‐Torres N, Jang S, Mok KY, Kim HJ, Han S, Cho K, Zhou X, Fu AKY, Ip NY, Seo J, Choi M, Jeong H, Hwang D, Lee DY, Byun MS, Yi D, Han JW, Mook‐Jung I, Hardy J. Multi-Omics-Based Autophagy-Related Untypical Subtypes in Patients with Cerebral Amyloid Pathology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201212. [PMID: 35694866 PMCID: PMC9376815 DOI: 10.1002/advs.202201212] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/26/2022] [Indexed: 05/05/2023]
Abstract
Recent multi-omics analyses paved the way for a comprehensive understanding of pathological processes. However, only few studies have explored Alzheimer's disease (AD) despite the possibility of biological subtypes within these patients. For this study, unsupervised classification of four datasets (genetics, miRNA transcriptomics, proteomics, and blood-based biomarkers) using Multi-Omics Factor Analysis+ (MOFA+), along with systems-biological approaches following various downstream analyses are performed. New subgroups within 170 patients with cerebral amyloid pathology (Aβ+) are revealed and the features of them are identified based on the top-rated targets constructing multi-omics factors of both whole (M-TPAD) and immune-focused models (M-IPAD). The authors explored the characteristics of subtypes and possible key-drivers for AD pathogenesis. Further in-depth studies showed that these subtypes are associated with longitudinal brain changes and autophagy pathways are main contributors. The significance of autophagy or clustering tendency is validated in peripheral blood mononuclear cells (PBMCs; n = 120 including 30 Aβ- and 90 Aβ+), induced pluripotent stem cell-derived human brain organoids/microglia (n = 12 including 5 Aβ-, 5 Aβ+, and CRISPR-Cas9 apolipoprotein isogenic lines), and human brain transcriptome (n = 78). Collectively, this study provides a strategy for precision medicine therapy and drug development for AD using integrative multi-omics analysis and network modelling.
Collapse
Affiliation(s)
- Jong‐Chan Park
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyUniversity College LondonLondonWC1N 3BGUK
- Department of Biochemistry and Biomedical SciencesCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
- Neuroscience Research InstituteMedical Research CenterCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
- SNU Korea Dementia Research CenterCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
| | - Natalia Barahona‐Torres
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyUniversity College LondonLondonWC1N 3BGUK
| | - So‐Yeong Jang
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Kin Y. Mok
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyUniversity College LondonLondonWC1N 3BGUK
| | - Haeng Jun Kim
- Department of Biochemistry and Biomedical SciencesCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
- SNU Korea Dementia Research CenterCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
| | - Sun‐Ho Han
- Department of Biochemistry and Biomedical SciencesCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
- Neuroscience Research InstituteMedical Research CenterCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
- SNU Korea Dementia Research CenterCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
| | - Kwang‐Hyun Cho
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and TechnologyDaejeon34141Republic of Korea
| | - Xiaopu Zhou
- Division of Life ScienceState Key Laboratory of Molecular NeuroscienceMolecular Neuroscience CenterThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
- Hong Kong Center for Neurodegenerative DiseasesHong Kong Science ParkHong Kong999077China
- Guangdong Provincial Key Laboratory of Brain ScienceDisease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhen‐Hong Kong Institute of Brain ScienceShenzhenGuangdong518057China
| | - Amy K. Y. Fu
- Division of Life ScienceState Key Laboratory of Molecular NeuroscienceMolecular Neuroscience CenterThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
- Hong Kong Center for Neurodegenerative DiseasesHong Kong Science ParkHong Kong999077China
- Guangdong Provincial Key Laboratory of Brain ScienceDisease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhen‐Hong Kong Institute of Brain ScienceShenzhenGuangdong518057China
| | - Nancy Y. Ip
- Division of Life ScienceState Key Laboratory of Molecular NeuroscienceMolecular Neuroscience CenterThe Hong Kong University of Science and TechnologyClear Water Bay, KowloonHong Kong999077China
- Hong Kong Center for Neurodegenerative DiseasesHong Kong Science ParkHong Kong999077China
- Guangdong Provincial Key Laboratory of Brain ScienceDisease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhen‐Hong Kong Institute of Brain ScienceShenzhenGuangdong518057China
| | - Jieun Seo
- Department of Laboratory MedicineSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Murim Choi
- Department of Biochemistry and Biomedical SciencesCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
| | - Hyobin Jeong
- European Molecular Biology LaboratoryGenome Biology UnitHeidelberg69117Germany
| | - Daehee Hwang
- Department of Biological SciencesSeoul National UniversitySeoul08826Republic of Korea
| | - Dong Young Lee
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoul03080Republic of Korea
- Department of PsychiatryCollege of medicineSeoul National UniversitySeoul03080Republic of Korea
- Department of NeuropsychiatrySeoul National University HospitalSeoul03080Republic of Korea
| | - Min Soo Byun
- Department of PsychiatryPusan National University Yangsan HospitalYangsan50612Republic of Korea
| | - Dahyun Yi
- Biomedical Research InstituteSeoul National University HospitalSeoul03082Republic of Korea
| | - Jong Won Han
- Department of Biochemistry and Biomedical SciencesCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
| | - Inhee Mook‐Jung
- Department of Biochemistry and Biomedical SciencesCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
- Neuroscience Research InstituteMedical Research CenterCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
- SNU Korea Dementia Research CenterCollege of MedicineSeoul National UniversitySeoul03080Republic of Korea
| | - John Hardy
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyUniversity College LondonLondonWC1N 3BGUK
| |
Collapse
|
162
|
Luo Q, Schnöder L, Hao W, Litzenburger K, Decker Y, Tomic I, Menger MD, Liu Y, Fassbender K. p38α‐MAPK‐deficient myeloid cells ameliorate symptoms and pathology of
APP
‐transgenic Alzheimer's disease mice. Aging Cell 2022; 21:e13679. [PMID: 35909315 PMCID: PMC9381888 DOI: 10.1111/acel.13679] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/26/2022] [Accepted: 07/07/2022] [Indexed: 11/27/2022] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia in the elderly, is pathologically characterized by extracellular deposition of amyloid‐β peptides (Aβ) and microglia‐dominated inflammatory activation in the brain. p38α‐MAPK is activated in both neurons and microglia. How p38α‐MAPK in microglia contributes to AD pathogenesis remains unclear. In this study, we conditionally knocked out p38α‐MAPK in all myeloid cells or specifically in microglia of APP‐transgenic mice, and examined animals for AD‐associated pathologies (i.e., cognitive deficits, Aβ pathology, and neuroinflammation) and individual microglia for their inflammatory activation and Aβ internalization at different disease stages (e.g., at 4 and 9 months of age). Our experiments showed that p38α‐MAPK‐deficient myeloid cells were more effective than p38α‐MAPK‐deficient microglia in reducing cerebral Aβ and neuronal impairment in APP‐transgenic mice. Deficiency of p38α‐MAPK in myeloid cells inhibited inflammatory activation of individual microglia at 4 months but enhanced it at 9 months. Inflammatory activation promoted microglial internalization of Aβ. Interestingly, p38α‐MAPK‐deficient myeloid cells reduced IL‐17a‐expressing CD4‐positive lymphocytes in 9 but not 4‐month‐old APP‐transgenic mice. By cross‐breeding APP‐transgenic mice with Il‐17a‐knockout mice, we observed that IL‐17a deficiency potentially activated microglia and reduced Aβ deposition in the brain as shown in 9‐month‐old myeloid p38α‐MAPK‐deficient AD mice. Thus, p38α‐MAPK deficiency in all myeloid cells, but not only in microglia, prevents AD progression. IL‐17a‐expressing lymphocytes may partially mediate the pathogenic role of p38α‐MAPK in peripheral myeloid cells. Our study supports p38α‐MAPK as a therapeutic target for AD patients.
Collapse
Affiliation(s)
- Qinghua Luo
- Department of Neurology Saarland University Homburg Germany
- German Institute for Dementia Prevention (DIDP) Saarland University Homburg Germany
| | - Laura Schnöder
- Department of Neurology Saarland University Homburg Germany
- German Institute for Dementia Prevention (DIDP) Saarland University Homburg Germany
| | - Wenlin Hao
- Department of Neurology Saarland University Homburg Germany
- German Institute for Dementia Prevention (DIDP) Saarland University Homburg Germany
| | - Kathrin Litzenburger
- Department of Neurology Saarland University Homburg Germany
- German Institute for Dementia Prevention (DIDP) Saarland University Homburg Germany
| | - Yann Decker
- Department of Neurology Saarland University Homburg Germany
- German Institute for Dementia Prevention (DIDP) Saarland University Homburg Germany
| | - Inge Tomic
- Department of Neurology Saarland University Homburg Germany
- German Institute for Dementia Prevention (DIDP) Saarland University Homburg Germany
| | - Michael D. Menger
- Institute for Clinical and Experimental Surgery Saarland University Homburg Germany
| | - Yang Liu
- Department of Neurology Saarland University Homburg Germany
- German Institute for Dementia Prevention (DIDP) Saarland University Homburg Germany
| | - Klaus Fassbender
- Department of Neurology Saarland University Homburg Germany
- German Institute for Dementia Prevention (DIDP) Saarland University Homburg Germany
| |
Collapse
|
163
|
Kuchcinski G, Patin L, Lopes R, Leroy M, Delbeuck X, Rollin-Sillaire A, Lebouvier T, Wang Y, Spincemaille P, Tourdias T, Hacein-Bey L, Devos D, Pasquier F, Leclerc X, Pruvo JP, Verclytte S. Quantitative susceptibility mapping demonstrates different patterns of iron overload in subtypes of early-onset Alzheimer's disease. Eur Radiol 2022; 33:184-195. [PMID: 35881183 DOI: 10.1007/s00330-022-09014-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES We aimed to define brain iron distribution patterns in subtypes of early-onset Alzheimer's disease (EOAD) by the use of quantitative susceptibility mapping (QSM). METHODS EOAD patients prospectively underwent MRI on a 3-T scanner and concomitant clinical and neuropsychological evaluation, between 2016 and 2019. An age-matched control group was constituted of cognitively healthy participants at risk of developing AD. Volumetry of the hippocampus and cerebral cortex was performed on 3DT1 images. EOAD subtypes were defined according to the hippocampal to cortical volume ratio (HV:CTV). Limbic-predominant atrophy (LPMRI) is referred to HV:CTV ratios below the 25th percentile, hippocampal-sparing (HpSpMRI) above the 75th percentile, and typical-AD between the 25th and 75th percentile. Brain iron was estimated using QSM. QSM analyses were made voxel-wise and in 7 regions of interest within deep gray nuclei and limbic structures. Iron distribution in EOAD subtypes and controls was compared using an ANOVA. RESULTS Sixty-eight EOAD patients and 43 controls were evaluated. QSM values were significantly higher in deep gray nuclei (p < 0.001) and limbic structures (p = 0.04) of EOAD patients compared to controls. Among EOAD subtypes, HpSpMRI had the highest QSM values in deep gray nuclei (p < 0.001) whereas the highest QSM values in limbic structures were observed in LPMRI (p = 0.005). QSM in deep gray nuclei had an AUC = 0.92 in discriminating HpSpMRI and controls. CONCLUSIONS In early-onset Alzheimer's disease patients, we observed significant variations of iron distribution reflecting the pattern of brain atrophy. Iron overload in deep gray nuclei could help to identify patients with atypical presentation of Alzheimer's disease. KEY POINTS • In early-onset AD patients, QSM indicated a significant brain iron overload in comparison with age-matched controls. • Iron load in limbic structures was higher in participants with limbic-predominant subtype. • Iron load in deep nuclei was more important in participants with hippocampal-sparing subtype.
Collapse
Affiliation(s)
- Grégory Kuchcinski
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France. .,UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, Univ Lille, F-59000, Lille, France. .,Department of Neuroradiology, CHU Lille, Rue Emile Laine, F-59000, Lille, France.
| | - Lucas Patin
- Department of Neuroradiology, CHU Lille, Rue Emile Laine, F-59000, Lille, France
| | - Renaud Lopes
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, Univ Lille, F-59000, Lille, France
| | - Mélanie Leroy
- Memory Center - CNR MAJ, DISTALZ-LICEND, F-59000, Lille, France
| | - Xavier Delbeuck
- Memory Center - CNR MAJ, DISTALZ-LICEND, F-59000, Lille, France
| | - Adeline Rollin-Sillaire
- Memory Center - CNR MAJ, DISTALZ-LICEND, F-59000, Lille, France.,Department of Neurology, CHU Lille, F-59000, Lille, France
| | - Thibaud Lebouvier
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,Memory Center - CNR MAJ, DISTALZ-LICEND, F-59000, Lille, France.,Department of Neurology, CHU Lille, F-59000, Lille, France
| | - Yi Wang
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | | | - Thomas Tourdias
- Neuroimagerie diagnostique et thérapeutique, CHU de Bordeaux, F-33000, Bordeaux, France.,Neurocentre Magendie, Inserm, U1215, Université de Bordeaux, F-33000, Bordeaux, France
| | - Lotfi Hacein-Bey
- Radiology Department, University of California Davis School of Medicine, Sacramento, CA, USA
| | - David Devos
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,Department of Pharmacology, CHU Lille, F-59000, Lille, France
| | - Florence Pasquier
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,Memory Center - CNR MAJ, DISTALZ-LICEND, F-59000, Lille, France.,Department of Neurology, CHU Lille, F-59000, Lille, France
| | - Xavier Leclerc
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, Univ Lille, F-59000, Lille, France.,Department of Neuroradiology, CHU Lille, Rue Emile Laine, F-59000, Lille, France
| | - Jean-Pierre Pruvo
- Inserm, U1172 - LilNCog - Lille Neuroscience & Cognition, Univ Lille, F-59000, Lille, France.,UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, Univ Lille, F-59000, Lille, France.,Department of Neuroradiology, CHU Lille, Rue Emile Laine, F-59000, Lille, France
| | - Sébastien Verclytte
- Department of Imaging, Lille Catholic Hospitals, Lille Catholic University, F-59000, Lille, France
| |
Collapse
|
164
|
Ma X, Zhang Y, Gou D, Ma J, Du J, Wang C, Li S, Cui H. Metabolic Reprogramming of Microglia Enhances Proinflammatory Cytokine Release through EphA2/p38 MAPK Pathway in Alzheimer’s Disease. J Alzheimers Dis 2022; 88:771-785. [DOI: 10.3233/jad-220227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background: The activation of microglia and neuroinflammation has been implicated in the pathogenesis of Alzheimer’s disease (AD), but the exact roles of microglia and the underlying mechanisms remain unclear. Objective: To clarify how the metabolic reprogramming of microglia induce by amyloid-β (Aβ)1-42 to affect the release of proinflammatory cytokines in AD. Methods: MTS assay was used to detect the viability of BV2 cells treated with different concentrations of Aβ1-42 for different periods of time. The expression levels of proinflammatory cytokines were determined by qRT-PCR and western blot assay in BV2 cells and hippocampus of mice. RNA sequencing was applied to evaluate the gene expression profiles in response to HK2 knockdown in BV2 cells treated with Aβ1-42. Results: Low concentrations of Aβ1-42 increased the viability of BV2 cells and promoted the release of proinflammatory cytokines, and this process is accompanied by increased glycolysis. Inhibition of glycolysis significantly downregulated the release of proinflammatory cytokines in BV2 cells and hippocampus of mice treated with Aβ1-42. The results of RNA sequencing revealed the expression of chemokine ligand 2 (Cxcl2) and ephrin receptor tyrosine kinase A2 (EphA2) were significantly downregulated when knocked down HK2 in BV2 cells. Subsequently, the expression of proinflammatory cytokines was downregulated in BV2 cell after knocking down EphA2. Conclusion: This study demonstrated that EphA2/p38 MAPK pathway is involved the release of proinflammatory cytokines in microglia induced by Aβ1-42 in AD, which is accompanied by metabolic reprogramming from oxidative phosphorylation (OXPHOS) to glycolysis.
Collapse
Affiliation(s)
- Xiaowei Ma
- Department of Anatomy, Hebei Medical University, Shijiazhuang, P.R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P.R. China
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Yizhou Zhang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, P.R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P.R. China
| | - Dongyun Gou
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Jingle Ma
- Department of Anatomy, Hebei Medical University, Shijiazhuang, P.R. China
| | - Juan Du
- Department of Anatomy, Hebei Medical University, Shijiazhuang, P.R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P.R. China
| | - Chang Wang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, P.R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P.R. China
| | - Sha Li
- Department of Anatomy, Hebei Medical University, Shijiazhuang, P.R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P.R. China
| | - Huixian Cui
- Department of Anatomy, Hebei Medical University, Shijiazhuang, P.R. China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, P.R. China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, P.R. China
| |
Collapse
|
165
|
Kobayashi R, Hayashi H, Kawakatsu S, Shibuya Y, Morioka D, Ohba M, Yoshioka M, Sakamoto K, Kanoto M, Otani K. Comparing Medial Temporal Atrophy Between Early-Onset Semantic Dementia and Early-Onset Alzheimer's Disease Using Voxel-Based Morphometry: A Multicenter MRI Study. Curr Alzheimer Res 2022; 19:503-510. [PMID: 35996258 DOI: 10.2174/1567205019666220820145429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Early-onset Semantic dementia (EOSD) and early-onset Alzheimer's disease (EOAD) are often difficult to clinically differentiate in the early stages of the diseases because of the overlaps of clinical symptoms such as language symptoms. We compared the degree of atrophy in medial temporal structures between the two types of dementia using the voxel-based specific regional analysis system for Alzheimer's disease (VSRAD). METHODS The participants included 29 (age: 61.7±4.5 years) and 39 (age: 60.2±4.9 years) patients with EOSD and EOAD, respectively. The degree of atrophy in medial temporal structures was quantified using the VSRAD for magnetic resonance imaging data. Receiver operating characteristic (ROC) analysis was performed to distinguish patients with EOSD and EOAD using the mean Z score (Z-score) in bilateral medial temporal structures and the absolute value (laterality score) of the laterality of Z-score (| right-left |) for indicating the degree of asymmetrical atrophy in medial temporal structures. RESULTS The EOSD group had significantly higher Z and laterality scores than the EOAD group (Zscores: mean ± standard deviation: 3.74±1.05 vs. 1.56±0.81, respectively; P<0.001; laterality score: mean ± standard deviation: 2.35±1.23 vs. 0.68±0.51, respectively; P<0.001). In ROC analysis, the sensitivity and specificity to differentiate EOSD from EOAD by a Z-score of 2.29 were 97% and 85%, respectively and by the laterality score of 1.05 were 93% and 85%, respectively. CONCLUSION EOSD leads to more severe and asymmetrical atrophy in medial temporal structures than EOAD. The VSRAD may be useful to distinguish between these dementias that have several clinically similar symptoms.
Collapse
Affiliation(s)
- Ryota Kobayashi
- Department of Psychiatry, Yamagata University School of Medicine, Iidanishi 2-2-2, Yamagata 990-9585, Japan
| | - Hiroshi Hayashi
- Department of Occupational Therapy, Fukushima Medical University School of Health Sciences, Sakaemachi 10-6, Fukushima 960-8516, Japan
| | - Shinobu Kawakatsu
- Department of Neuropsychiatry, Aizu Medical Center, Fukushima Medical University, Kawahigashi 21-2, Aizuwakamatsu 969-3492, Japan
| | - Yuzuru Shibuya
- Department of Psychiatry, Nihonkai General Hospital, Akihocho 30, Sakata 998-8501, Japan
| | - Daichi Morioka
- Department of Psychiatry, Yamagata University School of Medicine, Iidanishi 2-2-2, Yamagata 990-9585, Japan
| | - Makoto Ohba
- Department of Radiology, Yamagata University Hospital, Iidanishi 2-2-2, Yamagata 990- 9585, Japan
| | - Masanori Yoshioka
- Department of Radiology, Yamagata University Hospital, Iidanishi 2-2-2, Yamagata 990- 9585, Japan
| | - Kazutaka Sakamoto
- Department of Psychiatry, Yamagata University School of Medicine, Iidanishi 2-2-2, Yamagata 990-9585, Japan.,Department of Neuropsychiatry, Aizu Medical Center, Fukushima Medical University, Kawahigashi 21-2, Aizuwakamatsu 969-3492, Japan
| | - Masafumi Kanoto
- Department of Radiology, Division of Diagnostic Radiology, Yamagata University School of Medicine, Iidanishi 2-2-2, Yamagata 990-9585, Japan
| | - Koichi Otani
- Department of Psychiatry, Yamagata University School of Medicine, Iidanishi 2-2-2, Yamagata 990-9585, Japan
| |
Collapse
|
166
|
Toledo JB, Liu H, Grothe MJ, Rashid T, Launer L, Shaw LM, Snoussi H, Heckbert S, Weiner M, Trojanwoski JQ, Seshadri S, Habes M. Disentangling tau and brain atrophy cluster heterogeneity across the Alzheimer's disease continuum. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12305. [PMID: 35619830 PMCID: PMC9127251 DOI: 10.1002/trc2.12305] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 12/01/2022]
Abstract
Introduction Neuroimaging heterogeneity in dementia has been examined using single modalities. We evaluated the associations of magnetic resonance imaging (MRI) atrophy and flortaucipir positron emission tomography (PET) clusters across the Alzheimer's disease (AD) spectrum. Methods We included 496 Alzheimer's Disease Neuroimaging Initiative participants with brain MRI, flortaucipir PET scan, and amyloid beta biomarker measures obtained. We applied a novel robust collaborative clustering (RCC) approach on the MRI and flortaucipir PET scans. We derived indices for AD-like (SPARE-AD index) and brain age (SPARE-BA) atrophy. Results We identified four tau (I-IV) and three atrophy clusters. Tau clusters were associated with the apolipoprotein E genotype. Atrophy clusters were associated with white matter hyperintensity volumes. Only the hippocampal sparing atrophy cluster showed a specific association with brain aging imaging index. Tau clusters presented stronger clinical associations than atrophy clusters. Tau and atrophy clusters were partially associated. Conclusions Each neuroimaging modality captured different aspects of brain aging, genetics, vascular changes, and neurodegeneration leading to individual multimodal phenotyping.
Collapse
Affiliation(s)
- Jon B. Toledo
- Department of NeurologyUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Hangfan Liu
- Center for Biomedical Image Computing and Analytics (CBICA)University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Michel J. Grothe
- Unidad de Trastornos del Movimiento, Instituto de Bioedicina de Sevilla (IBiS)Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevilleSpain
| | - Tanweer Rashid
- Center for Biomedical Image Computing and Analytics (CBICA)University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexasUSA
| | - Lenore Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research ProgramNational Institute on AgingBethesdaMarylandUSA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Haykel Snoussi
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexasUSA
| | - Susan Heckbert
- Department of Epidemiology and Cardiovascular Health Research UnitUniversity of WashingtonSeattleWashingtonUSA
- Department of RadiologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of PsychiatryUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Michael Weiner
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Alzheimer's Disease Core Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - John Q. Trojanwoski
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute on AgingPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sudha Seshadri
- Udall Parkinson's Research CenterPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUniversity of Texas Health Sciences CenterSan AntonioTexasUSA
| | - Mohamad Habes
- Center for Biomedical Image Computing and Analytics (CBICA)University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexasUSA
| | | |
Collapse
|
167
|
Fu Z, Zhao M, He Y, Wang X, Li X, Kang G, Han Y, Li S. Aberrant topological organization and age-related differences in the human connectome in subjective cognitive decline by using regional morphology from magnetic resonance imaging. Brain Struct Funct 2022; 227:2015-2033. [PMID: 35579698 DOI: 10.1007/s00429-022-02488-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 03/24/2022] [Indexed: 11/25/2022]
Abstract
Subjective cognitive decline (SCD) is characterized by self-experienced deficits in cognitive capacity with normal performance in objective cognitive tests. Previous structural covariance studies showed specific insights into understanding the structural alterations of the brain in neurodegenerative diseases. Moreover, in subjects with neurodegenerative diseases, accelerated brain degeneration with aging was shown. However, the age-related variations in coordinated topological patterns of morphological networks in individuals with SCD remain poorly understood. In this study, 77 individual morphological networks were constructed, including 42 normal controls (NCs) and 35 SCD individuals, from structural magnetic resonance imaging (sMRI). A stepwise linear regression model and partial correlation analysis were constructed to evaluate the differences in age-related alterations of the network properties in individuals with SCD compared with NCs. Compared with NC, the properties of integration and segregation in individuals with SCD were lower, and the aberrant metrics were negatively correlated with age in SCD. The rich-club connections persevered, but the paralimbic system connections were disrupted in individuals with SCD compared with NCs. In addition, age-related differences in nodal global efficiency are distributed mainly in prefrontal cortex regions. In conclusion, the age-related disruption of topological organizations in individuals with SCD may indicate that the degeneration of brain efficiency with aging was accelerated in individuals with SCD.
Collapse
Affiliation(s)
- Zhenrong Fu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science & Medical Engineering, Beihang University, Beijing, China
| | - Mingyan Zhao
- Department of Neurology, Tangshan Gongren Hospital, Tangshan, Hebei, China
- Department of Neurology, XuanWu Hospital of Capital Medical University, Beijing, China
| | - Yirong He
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science & Medical Engineering, Beihang University, Beijing, China
| | - Xuetong Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science & Medical Engineering, Beihang University, Beijing, China
| | - Xin Li
- School of Electrical Engineering, Yanshan University, Qinhuangdao, China
- Measurement Technology and Instrumentation Key Lab of Hebei Province, Qinhuangdao, China
| | - Guixia Kang
- School of Information and Communication Engineering, Beijing University of Posts and Telecommunications, Beijing, China
| | - Ying Han
- Department of Neurology, XuanWu Hospital of Capital Medical University, Beijing, China
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
- Biomedical Engineering Institute, Hainan University, Haikou, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Shuyu Li
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science & Medical Engineering, Beihang University, Beijing, China.
| |
Collapse
|
168
|
Long-term use of pharmacological treatment in Alzheimer's disease: a retrospective cohort study in real-world clinical practice. Eur J Clin Pharmacol 2022; 78:1155-1163. [PMID: 35484251 DOI: 10.1007/s00228-022-03325-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 04/18/2022] [Indexed: 01/26/2023]
Abstract
PURPOSE To assess the impact of long-term use of different drugs commonly prescribed in Alzheimer's disease (AD) on its clinical course and to identify clinical and therapeutic factors associated with a delay in AD progression. METHODS We retrospectively enrolled 50 patients visited at the Neurology Unit, Careggi University Hospital (Florence), followed for at least 24 months. AD diagnosis was made according to clinical diagnostic criteria for probable/possible AD dementia, always supported at least by one biomarker. Clinical features, MMSE scores evaluated at diagnosis and every 6 months, and AD drugs used for at least 6 months, were recorded. Cox regression analysis was performed to estimate the hazard ratio (HR) for AD progression, assuming as the "final event," the progression to a more severe disease stage, defined as the achievement of an MMSE score less than 10. RESULTS At baseline, the median MMSE score was 22. During follow-up (median of 41 months), 56% of patients progressed to a more severe disease stage. The use of memantine, either alone (HR 0.24; 95% CI 0.09-0.60) or combined with acetylcholinesterase inhibitors (HR 0.35; 95% CI 0.14-0.88) and a higher MMSE score at baseline (HR 0.82; 95% CI 0.70-0.96) were associated with a significantly lower risk of AD progression. CONCLUSION Nowadays, effective disease-modifying therapy for AD is missing. Nevertheless, when the diagnosis is established, our results support the advantage of long-term use of available pharmacological treatments, especially in combination, in delaying AD progression to its more severe disease stage.
Collapse
|
169
|
Castillo-Passi RI, Vergara RC, Rogers NK, Ponce D, Bennett M, Behrens MI. Cancer History Is Associated with Slower Speed of Cognitive Decline in Patients with Amnestic Cognitive Impairment. J Alzheimers Dis 2022; 87:1695-1711. [DOI: 10.3233/jad-215660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Several epidemiological studies report a negative association between Cancer and Alzheimer’s disease (AD). Objective: To characterize the trajectories of memory loss in individuals with early amnestic cognitive impairment with and without history of previous cancer. Methods: Cognitive deterioration was assessed using the Montreal Cognitive Assessment (MoCA) or MoCA-Memory Index Score (MoCA-MIS) biannually in subjects with early amnestic cognitive impairment followed-up retrospectively from 2007 to 2021. History of Cancer was obtained from clinical records. Simple linear regressions of MoCA-MIS scores were calculated for each subject and analyzed with K-means cluster analysis to identify subgroups with different cognitive decline trajectories. χ 2 and t tests were used for descriptive categorical and continuous variables and mixed multiple linear regressions to determine cognitive decline covariates. Results: Analysis of the trajectory of cognitive decline in 141 subjects with early amnestic cognitive impairment identified two subgroups: Fast (n = 60) and Slow (n = 81) progressors. At baseline Fast progressors had better MoCA-MIS (p < 0.001) and functionality (CDR p = 0.02, AD8 p = 0.05), took less anti-dementia medications (p = 0.005), and had higher depression rates (p = 0.02). Interestingly, Fast progressors slowed their speed of memory decline (from 1.6 to 1.1 MoCA-MIS points/year) and global cognitive decline (from 2.0 to 1.4 total MoCA points/year) when Cancer history was present. Conclusion: Two trajectories of amnestic cognitive decline were identified, possibly derived from different neurophysiopathologies or clinical stages. This study suggests that a history of previous Cancer slows down amnestic cognitive decline, specifically in a subgroup of subjects with depression at baseline and accelerated deterioration at follow-up.
Collapse
Affiliation(s)
- Rolando I. Castillo-Passi
- Centro de Investigación Clínica Avanzada (CICA), Hospital Clínico de la Universidad de Chile, Independencia, Santiago, RM, Chile
- Departamento de Neurología y Psiquiatría, CAS, Clínica Alemana Universidad del Desarrollo, Santiago, RM, Chile
- Millennium Nucleus to Improve the Mental Health of Adolescents and Youths, Imhay, Chile
| | - Rodrigo C. Vergara
- Departamento de Kinesiología, Facultad de Artes y Educación Física, Universidad Metropolitana de Ciencias de la Educación, Santiago, RM, Chile
| | - Nicole K. Rogers
- Departamento de Neurociencia, Facultad de Medicina Universidad de Chile, Independencia Santiago, RM, Chile
- Instituto de Neurocirugía Dr. Alfonso Asenjo, Providencia, Santiago, RM, Chile
| | - Daniela Ponce
- Centro de Investigación Clínica Avanzada (CICA), Hospital Clínico de la Universidad de Chile, Independencia, Santiago, RM, Chile
| | - Magdalena Bennett
- IROM Department, McCombs School of Business, The University of Texas at Austin, Austin, TX, USA
| | - María Isabel Behrens
- Centro de Investigación Clínica Avanzada (CICA), Hospital Clínico de la Universidad de Chile, Independencia, Santiago, RM, Chile
- Departamento de Neurología y Neurocirugía, Hospital Clínico, Universidad de Chile, Independencia, Santiago, RM, Chile
- Departamento de Neurociencia, Facultad de Medicina Universidad de Chile, Independencia Santiago, RM, Chile
- Departamento de Neurología y Psiquiatría, CAS, Clínica Alemana Universidad del Desarrollo, Santiago, RM, Chile
| |
Collapse
|
170
|
Persson K, Edwin TH, Knapskog AB, Tangen GG, Selbæk G, Engedal K. Hippocampal Atrophy Subtypes of Alzheimer's Disease Using Automatic MRI in a Memory Clinic Cohort: Clinical Implications. Dement Geriatr Cogn Disord 2022; 51:80-89. [PMID: 35344967 DOI: 10.1159/000522382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/28/2022] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION One pathological hallmark of Alzheimer's disease (AD) is atrophy of medial temporal brain regions that can be visualized on magnetic resonance imaging (MRI), but not all patients will have atrophy. The aim was to use MRI to categorize patients according to their hippocampal atrophy status and to present prevalence of the subtypes, difference in clinical symptomatology and progression, and factors associated with hippocampal subtypes. METHODS We included 215 patients with AD who had been assessed with the clinically available MRI software NeuroQuant (NQ; CorTechs labs/University of California, San Diego, CA, USA). NQ measures the hippocampus volume and calculates a normative percentile. Atrophy was regarded to be present if the percentile was ≤5. Demographics, cognitive measurements, AD phenotypes, apolipoprotein E status, and results from cerebrospinal fluid and amyloid positron emission tomography analyses were included as explanatory variables of the hippocampal subtypes. RESULTS Of all, 60% had no hippocampal atrophy. These patients were younger and less cognitively impaired concerning global measures, memory function, and abstraction but impaired concerning executive, visuospatial, and semantic fluency, and more of them had nonamnestic AD, compared to those with hippocampal atrophy. No difference in progression rate was observed between the two groups. In mild cognitive impairment patients, amyloid pathology was associated with the no hippocampal atrophy group. CONCLUSION The results have clinical implications. Clinicians should be aware of the large proportion of AD patients presenting without atrophy of the hippocampus as measured with this clinical MRI method in the diagnostic set up and that nonamnestic phenotypes are more common in this group as compared to those with atrophy. Furthermore, the findings are relevant in clinical trials.
Collapse
Affiliation(s)
- Karin Persson
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway.,Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Trine H Edwin
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway.,Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Gro G Tangen
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway.,Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Geir Selbæk
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway.,Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Knut Engedal
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway.,Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
171
|
Golriz Khatami S, Salimi Y, Hofmann-Apitius M, Oxtoby NP, Birkenbihl C. Comparison and aggregation of event sequences across ten cohorts to describe the consensus biomarker evolution in Alzheimer's disease. Alzheimers Res Ther 2022; 14:55. [PMID: 35443691 PMCID: PMC9020023 DOI: 10.1186/s13195-022-01001-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/06/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Previous models of Alzheimer's disease (AD) progression were primarily hypothetical or based on data originating from single cohort studies. However, cohort datasets are subject to specific inclusion and exclusion criteria that influence the signals observed in their collected data. Furthermore, each study measures only a subset of AD-relevant variables. To gain a comprehensive understanding of AD progression, the heterogeneity and robustness of estimated progression patterns must be understood, and complementary information contained in cohort datasets be leveraged. METHODS We compared ten event-based models that we fit to ten independent AD cohort datasets. Additionally, we designed and applied a novel rank aggregation algorithm that combines partially overlapping, individual event sequences into a meta-sequence containing the complementary information from each cohort. RESULTS We observed overall consistency across the ten event-based model sequences (average pairwise Kendall's tau correlation coefficient of 0.69 ± 0.28), despite variance in the positioning of mainly imaging variables. The changes described in the aggregated meta-sequence are broadly consistent with the current understanding of AD progression, starting with cerebrospinal fluid amyloid beta, followed by tauopathy, memory impairment, FDG-PET, and ultimately brain deterioration and impairment of visual memory. CONCLUSION Overall, the event-based models demonstrated similar and robust disease cascades across independent AD cohorts. Aggregation of data-driven results can combine complementary strengths and information of patient-level datasets. Accordingly, the derived meta-sequence draws a more complete picture of AD pathology compared to models relying on single cohorts.
Collapse
Affiliation(s)
- Sepehr Golriz Khatami
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53757, Sankt Augustin, Germany.
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany.
| | - Yasamin Salimi
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53757, Sankt Augustin, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53757, Sankt Augustin, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany
| | - Neil P Oxtoby
- Centre for Medical Image Computing and Department of Computer Science, University College London, Gower St, London, WC1E 6BT, UK
| | - Colin Birkenbihl
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), 53757, Sankt Augustin, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany
| |
Collapse
|
172
|
Young CB, Winer JR, Younes K, Cody KA, Betthauser TJ, Johnson SC, Schultz A, Sperling RA, Greicius MD, Cobos I, Poston KL, Mormino EC. Divergent Cortical Tau Positron Emission Tomography Patterns Among Patients With Preclinical Alzheimer Disease. JAMA Neurol 2022; 79:592-603. [PMID: 35435938 PMCID: PMC9016616 DOI: 10.1001/jamaneurol.2022.0676] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Importance Characterization of early tau deposition in individuals with preclinical Alzheimer disease (AD) is critical for prevention trials that aim to select individuals at risk for AD and halt the progression of disease. Objective To evaluate the prevalence of cortical tau positron emission tomography (PET) heterogeneity in a large cohort of clinically unimpaired older adults with elevated β-amyloid (A+). Design, Setting, and Participants This cross-sectional study examined prerandomized tau PET, amyloid PET, structural magnetic resonance imaging, demographic, and cognitive data from the Anti-Amyloid Treatment in Asymptomatic AD (A4) Study from April 2014 to December 2017. Follow-up analyses used observational tau PET data from the Alzheimer's Disease Neuroimaging Initiative (ADNI), the Harvard Aging Brain Study (HABS), and the Wisconsin Registry for Alzheimer's Prevention and the Wisconsin Alzheimer's Disease Research Center (together hereinafter referred to as Wisconsin) to evaluate consistency. Participants were clinically unimpaired at the study visit closest to the tau PET scan and had available amyloid and tau PET data (A4 Study, n = 447; ADNI, n = 433; HABS, n = 190; and Wisconsin, n = 328). No participants who met eligibility criteria were excluded. Data were analyzed from May 11, 2021, to January 25, 2022. Main Outcomes and Measures Individuals with preclinical AD with heterogeneous cortical tau PET patterns (A+T cortical+) were identified by examining asymmetrical cortical tau signal and disproportionate cortical tau signal relative to medial temporal lobe (MTL) tau. Voxelwise tau patterns, amyloid, neurodegeneration, cognition, and demographic characteristics were examined. Results The 447 A4 participants (A+ group, 392; and normal β-amyloid group, 55), with a mean (SD) age of 71.8 (4.8) years, included 239 women (54%). A total of 36 individuals in the A+ group (9% of the A+ group) exhibited heterogeneous cortical tau patterns and were further categorized into 3 subtypes: asymmetrical left, precuneus dominant, and asymmetrical right. A total of 116 individuals in the A+ group (30% of the A+ group) showed elevated MTL tau (A+T MTL+). Individuals in the A+T cortical+ group were younger than those in the A+T MTL+ group (t61.867 = -2.597; P = .03). Across the A+T cortical+ and A+T MTL+ groups, increased regional tau was associated with reduced hippocampal volume and MTL thickness but not with cortical thickness. Memory scores were comparable between the A+T cortical+ and A+T MTL+ groups, whereas executive functioning scores were lower for the A+T cortical+ group than for the A+T MTL+ group. The prevalence of the A+T cortical+ group and tau patterns within the A+T cortical+ group were consistent in ADNI, HABS, and Wisconsin. Conclusions and Relevance This study suggests that early tau deposition may follow multiple trajectories during preclinical AD and may involve several cortical regions. Staging procedures, especially those based on neuropathology, that assume a uniform trajectory across individuals are insufficient for disease monitoring with tau imaging.
Collapse
Affiliation(s)
- Christina B Young
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Joseph R Winer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Kyan Younes
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Karly A Cody
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison.,Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison.,Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison.,Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison.,Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison
| | - Aaron Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Inma Cobos
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | | |
Collapse
|
173
|
Hanyu H, Koyama Y, Horita H, Aoki T, Sato T, Takenoshita N, Kanetaka H, Shimizu S, Hirao K, Watanabe S. Characterization of Alzheimer’s Disease Subtypes Based on Magnetic Resonance Imaging and Perfusion Single-Photon Emission Computed Tomography. J Alzheimers Dis 2022; 87:781-789. [DOI: 10.3233/jad-215674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Alzheimer’s disease (AD) is a biologically heterogenous disease. Previous studies have reported the existence of various AD subtypes, and the various clinical features of the subtypes. However, inconsistent results have been obtained. Objective: To clarify the clinical characteristics of the various AD subtypes, by classifying probable AD into subtypes based on magnetic resonance imaging (MRI) and single-photon emission computed tomography (SPECT) findings. Methods: A total of 245 patients with probable AD were classified into the typical AD (TAD) subtype, limbic-predominant (LP) subtype, hippocampal-sparing (HS) subtype, and minimal-change (MC) subtype, based on the presence of medial temporal lobe atrophy on MRI and posterior cerebral hypoperfusion on SPECT. Demographics, including age, sex, body mass index, disease duration, education years, comorbidities, frailty, leisure activity, and neuropsychological findings were compared between the AD subtypes. Results: he frequency of TAD, LP, HS, and MC subtypes was 49%, 20%, 18%, and 13%, respectively. Patients with the LP subtype were older and characterized by fewer major comorbidities, higher frailty, and slower progression of disease. Patients with the HS subtype were younger and characterized by shorter disease duration, lower frailty, and preserved memory, but had prominent constructional dysfunction. Patients of the MC subtype were characterized by shorter disease duration, lower education level, less leisure activity, less impaired memory and orientation, and slower progression. Conclusion: Patients with different AD subtypes differed in their demographic and clinical features. The characterization of patients’ AD subtypes may provide effective support for the diagnosis, treatment, and care of AD patients.
Collapse
Affiliation(s)
- Haruo Hanyu
- Dementia Research Center, Tokyo General Hospital, Tokyo, Japan
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Yumi Koyama
- Department of Rehabilitation, Tokyo General Hospital, Tokyo, Japan
| | - Haruka Horita
- Department of Rehabilitation, Tokyo General Hospital, Tokyo, Japan
| | - Toshinori Aoki
- Department of Radiology, Tokyo General Hospital, Tokyo, Japan
| | - Tomohiko Sato
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Naoto Takenoshita
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Hidekazu Kanetaka
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Soichiro Shimizu
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Kentaro Hirao
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | | |
Collapse
|
174
|
Duong MT, Das SR, Lyu X, Xie L, Richardson H, Xie SX, Yushkevich PA, Wolk DA, Nasrallah IM. Dissociation of tau pathology and neuronal hypometabolism within the ATN framework of Alzheimer's disease. Nat Commun 2022; 13:1495. [PMID: 35314672 PMCID: PMC8938426 DOI: 10.1038/s41467-022-28941-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/11/2022] [Indexed: 11/08/2022] Open
Abstract
Alzheimer's disease (AD) is defined by amyloid (A) and tau (T) pathologies, with T better correlated to neurodegeneration (N). However, T and N have complex regional relationships in part related to non-AD factors that influence N. With machine learning, we assessed heterogeneity in 18F-flortaucipir vs. 18F-fluorodeoxyglucose positron emission tomography as markers of T and neuronal hypometabolism (NM) in 289 symptomatic patients from the Alzheimer's Disease Neuroimaging Initiative. We identified six T/NM clusters with differing limbic and cortical patterns. The canonical group was defined as the T/NM pattern with lowest regression residuals. Groups resilient to T had less hypometabolism than expected relative to T and displayed better cognition than the canonical group. Groups susceptible to T had more hypometabolism than expected given T and exhibited worse cognitive decline, with imaging and clinical measures concordant with non-AD copathologies. Together, T/NM mismatch reveals distinct imaging signatures with pathobiological and prognostic implications for AD.
Collapse
Affiliation(s)
- Michael Tran Duong
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Sandhitsu R Das
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xueying Lyu
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Long Xie
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hayley Richardson
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sharon X Xie
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A Yushkevich
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Wolk
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Ilya M Nasrallah
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
- Alzheimer's Disease Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
175
|
Raikes AC, Hernandez GD, Matthews DC, Lukic AS, Law M, Shi Y, Schneider LS, Brinton RD. Exploratory imaging outcomes of a phase 1b/2a clinical trial of allopregnanolone as a regenerative therapeutic for Alzheimer's disease: Structural effects and functional connectivity outcomes. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12258. [PMID: 35310526 PMCID: PMC8919249 DOI: 10.1002/trc2.12258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 01/14/2023]
Abstract
Introduction Allopregnanolone (ALLO), an endogenous neurosteroid, promoted neurogenesis and oligogenesis and restored cognitive function in animal models of Alzheimer's disease (AD). Based on these discovery research findings, we conducted a randomized-controlled phase 1b/2a multiple ascending dose trial of ALLO in persons with early AD (NCT02221622) to assess safety, tolerability, and pharmacokinetics. Exploratory imaging outcomes to determine whether ALLO impacted hippocampal structure, white matter integrity, and functional connectivity are reported. Methods Twenty-four individuals participated in the trial (n = 6 placebo; n = 18 ALLO) and underwent brain magnetic resonance imaging (MRI) before and after 12 weeks of treatment. Hippocampal atrophy rate was determined from volumetric MRI, computed as rate of change, and qualitatively assessed between ALLO and placebo sex, apolipoprotein E (APOE) ε4 allele, and ALLO dose subgroups. White matter microstructural integrity was compared between placebo and ALLO using fractional and quantitative anisotropy (QA). Changes in local, inter-regional, and network-level functional connectivity were also compared between groups using resting-state functional MRI. Results Rate of decline in hippocampal volume was slowed, and in some cases reversed, in the ALLO group compared to placebo. Gain of hippocampal volume was evident in APOE ε4 carriers (range: 0.6% to 7.8% increased hippocampal volume). Multiple measures of white matter integrity indicated evidence of preserved or improved integrity. ALLO significantly increased fractional anisotropy (FA) in 690 of 690 and QA in 1416 of 1888 fiber tracts, located primarily in the corpus callosum, bilateral thalamic radiations, and bilateral corticospinal tracts. Consistent with structural changes, ALLO strengthened local, inter-regional, and network level functional connectivity in AD-vulnerable regions, including the precuneus and posterior cingulate, and network connections between the default mode network and limbic system. Discussion Indicators of regeneration from previous preclinical studies and these exploratory MRI-based outcomes from this phase 1b/2a clinical cohort support advancement to a phase 2 proof-of-concept efficacy clinical trial of ALLO as a regenerative therapeutic for mild AD (REGEN-BRAIN study; NCT04838301).
Collapse
Affiliation(s)
- Adam C. Raikes
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
| | | | - Dawn C. Matthews
- Departments of Pharmacology and Neurology, College of MedicineADM DiagnosticsNorthbrookIllinoisUSA
| | - Ana S. Lukic
- Departments of Pharmacology and Neurology, College of MedicineADM DiagnosticsNorthbrookIllinoisUSA
| | - Meng Law
- Department of RadiologyAlfred HealthDepartment of Neuroscience and Computer Systems EngineeringMonash UniversityMelbourneAustralia
| | - Yonggang Shi
- Stevens Neuroimaging and Informatics InstituteKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Lon S. Schneider
- Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Roberta D. Brinton
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
176
|
Ezzati A, Davatzikos C, Wolk DA, Hall CB, Habeck C, Lipton RB. Application of predictive models in boosting power of Alzheimer's disease clinical trials: A post hoc analysis of phase 3 solanezumab trials. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12223. [PMID: 35310531 PMCID: PMC8919041 DOI: 10.1002/trc2.12223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 07/15/2021] [Accepted: 11/01/2021] [Indexed: 01/18/2023]
Abstract
Background The ideal participants for Alzheimer's disease (AD) clinical trials would show cognitive decline in the absence of treatment (i.e., placebo arm) and would also respond to the therapeutic intervention. Objective To investigate if predictive models can be an effective tool for identifying and excluding people unlikely to show cognitive decline as an enrichment strategy in AD trials. Method We used data from the placebo arms of two phase 3, double-blind trials, EXPEDITION and EXPEDITION2. Patients had 18 months of follow-up. Based on the longitudinal data from the placebo arm, we classified participants into two groups: one showed cognitive decline (any negative slope) and the other showed no cognitive decline (slope is zero or positive) on the Alzheimer's Disease Assessment Scale-Cognitive subscale (ADAS-cog). We used baseline data for EXPEDITION to train regression-based classifiers and machine learning classifiers to estimate probability of cognitive decline. Models were applied to EXPEDITION2 data to assess predicted performance in an independent sample. Features used in predictive models included baseline demographics, apolipoprotein E ε4 genotype, neuropsychological scores, functional scores, and volumetric magnetic resonance imaging. Result In EXPEDITION, 46.3% of placebo-treated patients showed no cognitive decline and the proportion was similar in EXPEDITION2 (45.6%). Models had high sensitivity and modest specificity in both the training (EXPEDITION) and replication samples (EXPEDITION2) for detecting the stable group. Positive predictive value of models was higher than the base prevalence of cognitive decline, and negative predictive value of models were higher than the base rate of participants who had stable cognition. Conclusion Excluding persons with AD unlikely to decline from the active and placebo arms of clinical trials using predictive models may boost the power of AD trials through selective inclusion of participants expected to decline.
Collapse
Affiliation(s)
- Ali Ezzati
- Department of NeurologyAlbert Einstein College of Medicine and Montefiore Medical CenterBronxNew YorkUSA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and AnalyticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - David A. Wolk
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Memory CenterPerelman Center for Advanced MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Charles B. Hall
- Department of Department of Epidemiology and Population HealthAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Christian Habeck
- Department of NeurologyCognitive Neuroscience DivisionColumbia UniversityNew YorkNew YorkUSA
| | - Richard B. Lipton
- Department of NeurologyAlbert Einstein College of Medicine and Montefiore Medical CenterBronxNew YorkUSA
- Department of Department of Epidemiology and Population HealthAlbert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
177
|
Chung SJ, Chang Y, Jeon J, Shin JI, Song TJ, Kim J. Association of Alzheimer's Disease with COVID-19 Susceptibility and Severe Complications: A Nationwide Cohort Study. J Alzheimers Dis 2022; 87:701-710. [PMID: 35275548 DOI: 10.3233/jad-220031] [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] [Indexed: 12/21/2022]
Abstract
BACKGROUND Identification of patients at high susceptibility and high risk of developing serious complications related to coronavirus disease 2019 (COVID-19) infection is clinically important in the face of the COVID-19 pandemic. OBJECTIVE To investigate whether patients with Alzheimer's disease (AD) are more susceptible to COVID-19 infection and whether they have a higher risk of developing serious complications. METHODS We retrospectively reviewed the Korean nationwide population-based COVID-19 dataset for participants who underwent real-time reverse transcription polymerase chain reaction assays for COVID-19 between January 1 and June 4, 2020. A 1 : 3 ratio propensity score matching and binary logistic regression analysis were performed to investigate the association between AD and the susceptibility or severe complications (i.e., mechanical ventilation, intensive care unit admission, or death) of COVID-19. RESULTS Among 195,643 study participants, 5,725 participants had AD and 7,334 participants were diagnosed with COVID-19. The prevalence of participants testing positive for COVID-19 did not differ according to the presence of AD (p = 0.234). Meanwhile, AD was associated with an increased risk of severe COVID-19 complications (OR 2.25 [95% CI 1.54-3.28]). Secondary outcome analyses showed that AD patients had an increased risk for mortality (OR 3.09 [95% CI 2.00-4.78]) but were less likely to receive mechanical ventilation (OR 0.42 [95% CI 0.20-0.87]). CONCLUSION AD was not associated with increased susceptibility to COVID-19 infection, but was associated with severe COVID-19 complications, especially with mortality. Early diagnosis and active intervention are necessary for patients with AD suspected COVID-19 infection.
Collapse
Affiliation(s)
- Seok Jong Chung
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Republic of Korea.,Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoonkyung Chang
- Department of Neurology, Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Jimin Jeon
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Republic of Korea.,Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Jin Song
- Department of Neurology, Seoul Hospital, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Jinkwon Kim
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, Republic of Korea.,Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
178
|
Wu J, Zhao K, Li Z, Wang D, Ding Y, Wei Y, Zhang H, Liu Y. A systematic analysis of diagnostic performance for Alzheimer's disease using structural MRI. PSYCHORADIOLOGY 2022; 2:287-295. [PMID: 38665142 PMCID: PMC10939341 DOI: 10.1093/psyrad/kkac001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/13/2022] [Accepted: 02/14/2022] [Indexed: 04/28/2024]
Abstract
Background Alzheimer's disease (AD) is one of the most common neurodegenerative disorders in the elderly. Although numerous structural magnetic resonance imaging (sMRI) studies have reported diagnostic models that could distinguish AD from normal controls (NCs) with 80-95% accuracy, limited efforts have been made regarding the clinically practical computer-aided diagnosis (CAD) system for AD. Objective To explore the potential factors that hinder the clinical translation of the AD-related diagnostic models based on sMRI. Methods To systematically review the diagnostic models for AD based on sMRI, we identified relevant studies published in the past 15 years on PubMed, Web of Science, Scopus, and Ovid. To evaluate the heterogeneity and publication bias among those studies, we performed subgroup analysis, meta-regression, Begg's test, and Egger's test. Results According to our screening criterion, 101 studies were included. Our results demonstrated that high diagnostic accuracy for distinguishing AD from NC was obtained in recently published studies, accompanied by significant heterogeneity. Meta-analysis showed that many factors contributed to the heterogeneity of high diagnostic accuracy of AD using sMRI, which included but was not limited to the following aspects: (i) different datasets; (ii) different machine learning models, e.g. traditional machine learning or deep learning model; (iii) different cross-validation methods, e.g. k-fold cross-validation leads to higher accuracies than leave-one-out cross-validation, but both overestimate the accuracy when compared to validation in independent samples; (iv) different sample sizes; and (v) the publication times. We speculate that these complicated variables might be the adverse factor for developing a clinically applicable system for the early diagnosis of AD. Conclusions Our findings proved that previous studies reported promising results for classifying AD from NC with different models using sMRI. However, considering the many factors hindering clinical radiology practice, there would still be a long way to go to improve.
Collapse
Affiliation(s)
- Jiangping Wu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| | - Kun Zhao
- Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Zhuangzhuang Li
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| | - Dong Wang
- School of Information Science and Engineering, Shandong Normal University, Ji'nan, 250014, China
| | - Yanhui Ding
- School of Information Science and Engineering, Shandong Normal University, Ji'nan, 250014, China
| | - Yongbin Wei
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| | - Han Zhang
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, 100876, China
- Center for Artificial Intelligence in Medical Imaging, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| |
Collapse
|
179
|
Hanyu H, Koyama Y, Horita H, Aoki T, Sato T, Kanetaka H, Shimizu S, Hirao K. Discrepancy between cognitive test and brain imaging results in Alzheimer's disease associated with diabetes. Curr Alzheimer Res 2022; 19:95-103. [PMID: 35227184 DOI: 10.2174/1567205019666220228152655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/16/2021] [Accepted: 01/14/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND/OBJECTIVE Although a large number of studies have been performed on the association between Alzheimer's disease (AD) and type 2 diabetes mellitus (DM), the underlying pathophysiology of AD associated with DM has not been fully elucidated to date. We compared cognitive functions and brain imaging findings between AD patients with and without DM to characterize the association between cognition and imaging findings in AD patients with DM. METHODS Cognitive functions and brain imaging findings, including medial temporal lobe atrophy analyzed by magnetic resonance imaging, and hypoperfusion in the parietal, posterior cingulate, and frontal regions analyzed by single-photon emission computed tomography were compared between 126 AD patients without DM ([AD-DM]) and 51 AD patients with DM ([AD+DM]). Factors associated with cognitive-imaging associations, including education, occupation, leisure activity, comorbidity, frailty, and other demographics, were analyzed. RESULTS The [AD+DM] group showed significantly more severe cognitive dysfunction than the [AD- DM] group, despite a similar degree of brain imaging abnormalities. Among the factors associated with cognitive-imaging associations, the level of leisure activity was significantly lower in the [AD+DM] group than in the [AD-DM] group, but no significant differences in other factors were observed between the 2 groups. CONCLUSION The cognitive-imaging discrepancy observed in AD patients with DM may be associated with their low cognitive reserve, possibly caused by their low amount of leisure activities. Our findings suggest that lifestyle interventions, including physical, cognitive, and social activities, may reduce cognitive decline in AD patients with DM.
Collapse
Affiliation(s)
- Haruo Hanyu
- Dementia Research Center, Tokyo General Hospital, Tokyo, Japan
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Yumi Koyama
- Rehabilitation Center, Tokyo General Hospital, Tokyo, Japan
| | - Haruka Horita
- Rehabilitation Center, Tokyo General Hospital, Tokyo, Japan
| | - Toshinori Aoki
- Department of Radiology, Tokyo General Hospital, Tokyo, Japan
| | - Tomohiko Sato
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Hidekazu Kanetaka
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Soichiro Shimizu
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Kentaro Hirao
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
180
|
Tremblay C, Serrano GE, Intorcia AJ, Curry J, Sue LI, Nelson CM, Walker JE, Glass MJ, Arce RA, Fleisher AS, Pontecorvo MJ, Atri A, Montine TJ, Chen K, Beach TG. Hemispheric Asymmetry and Atypical Lobar Progression of Alzheimer-Type Tauopathy. J Neuropathol Exp Neurol 2022; 81:158-171. [PMID: 35191506 DOI: 10.1093/jnen/nlac008] [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: 11/13/2022] Open
Abstract
The spread of neurofibrillary tau pathology in Alzheimer disease (AD) mostly follows a stereotypical pattern of topographical progression but atypical patterns associated with interhemispheric asymmetry have been described. Because histopathological studies that used bilateral sampling are limited, this study aimed to assess interhemispheric tau pathology differences and the presence of topographically atypical cortical spreading patterns. Immunohistochemical staining for detection of tau pathology was performed in 23 regions of interest in 57 autopsy cases comparing bilateral cortical regions and hemispheres. Frequent mild (82% of cases) and occasional moderate (32%) interhemispheric density discrepancies were observed, whereas marked discrepancies were uncommon (7%) and restricted to occipital regions. Left and right hemispheric tau pathology dominance was observed with similar frequencies, except in Braak Stage VI that favored a left dominance. Interhemispheric Braak stage differences were observed in 16% of cases and were more frequent in advanced (IV-VI) versus early (I-III) stages. One atypical lobar topographical pattern in which occipital tau pathology density exceeded frontal lobe scores was identified in 4 cases favoring a left dominant asymmetry. We speculate that asymmetry and atypical topographical progression patterns may be associated with atypical AD clinical presentations and progression characteristics, which should be tested by comprehensive clinicopathological correlations.
Collapse
Affiliation(s)
- Cécilia Tremblay
- From the Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Geidy E Serrano
- From the Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Jasmine Curry
- From the Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Lucia I Sue
- From the Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Courtney M Nelson
- From the Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Jessica E Walker
- From the Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Michael J Glass
- From the Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Richard A Arce
- From the Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | - Alireza Atri
- From the Banner Sun Health Research Institute, Sun City, Arizona, USA.,Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, Arizona, USA.,School of Mathematics and Statistics, Arizona State University, Tempe, Arizona, USA.,Department of Neurology, College of Medicine Phoenix, University of Arizona, Tucson, Arizona, USA
| | - Thomas G Beach
- From the Banner Sun Health Research Institute, Sun City, Arizona, USA
| |
Collapse
|
181
|
Concordance of Alzheimer’s Disease Subtypes Produced from Different Representative Morphological Measures: A Comparative Study. Brain Sci 2022; 12:brainsci12020187. [PMID: 35203950 PMCID: PMC8869952 DOI: 10.3390/brainsci12020187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Gray matter (GM) density and cortical thickness (CT) obtained from structural magnetic resonance imaging are representative GM morphological measures that have been commonly used in Alzheimer’s disease (AD) subtype research. However, how the two measures affect the definition of AD subtypes remains unclear. Methods: A total of 180 AD patients from the ADNI database were used to identify AD subgroups. The subtypes were identified via a data-driven strategy based on the density features and CT features, respectively. Then, the similarity between the two features in AD subtype definition was analyzed. Results: Four distinct subtypes were discovered by both density and CT features: diffuse atrophy AD, minimal atrophy AD (MAD), left temporal dominant atrophy AD (LTAD), and occipital sparing AD. The matched subtypes exhibited relatively high similarity in atrophy patterns and neuropsychological and neuropathological characteristics. They differed only in MAD and LTAD regarding the carrying of apolipoprotein E ε2. Conclusions: The results verified that different representative morphological GM measurement methods could produce similar AD subtypes. Meanwhile, the influences of apolipoprotein E genotype, asymmetric disease progression, and their interactions should be considered and included in the AD subtype definition. This study provides a valuable reference for selecting features in future studies of AD subtypes.
Collapse
|
182
|
Abdelnour C, Ferreira D, van de Beek M, Cedres N, Oppedal K, Cavallin L, Blanc F, Bousiges O, Wahlund LO, Pilotto A, Padovani A, Boada M, Pagonabarraga J, Kulisevsky J, Aarsland D, Lemstra AW, Westman E. Parsing heterogeneity within dementia with Lewy bodies using clustering of biological, clinical, and demographic data. Alzheimers Res Ther 2022; 14:14. [PMID: 35063023 PMCID: PMC8783432 DOI: 10.1186/s13195-021-00946-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/06/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) includes various core clinical features that result in different phenotypes. In addition, Alzheimer's disease (AD) and cerebrovascular pathologies are common in DLB. All this increases the heterogeneity within DLB and hampers clinical diagnosis. We addressed this heterogeneity by investigating subgroups of patients with similar biological, clinical, and demographic features. METHODS We studied 107 extensively phenotyped DLB patients from the European DLB consortium. Factorial analysis of mixed data (FAMD) was used to identify dimensions in the data, based on sex, age, years of education, disease duration, Mini-Mental State Examination (MMSE), cerebrospinal fluid (CSF) levels of AD biomarkers, core features of DLB, and regional brain atrophy. Subsequently, hierarchical clustering analysis was used to subgroup individuals based on the FAMD dimensions. RESULTS We identified 3 dimensions using FAMD that explained 38% of the variance. Subsequent hierarchical clustering identified 4 clusters. Cluster 1 was characterized by amyloid-β and cerebrovascular pathologies, medial temporal atrophy, and cognitive fluctuations. Cluster 2 had posterior atrophy and showed the lowest frequency of visual hallucinations and cognitive fluctuations and the worst cognitive performance. Cluster 3 had the highest frequency of tau pathology, showed posterior atrophy, and had a low frequency of parkinsonism. Cluster 4 had virtually normal AD biomarkers, the least regional brain atrophy and cerebrovascular pathology, and the highest MMSE scores. CONCLUSIONS This study demonstrates that there are subgroups of DLB patients with different biological, clinical, and demographic characteristics. These findings may have implications in the diagnosis and prognosis of DLB, as well as in the treatment response in clinical trials.
Collapse
Affiliation(s)
- Carla Abdelnour
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
- Department of Medicine of the Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Marleen van de Beek
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Nira Cedres
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Psychology, Sensory Cognitive Interaction Laboratory (SCI-lab), Stockholm University, Stockholm, Sweden
| | - Ketil Oppedal
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Radiology, Stavanger University Hospital, Stavanger, Norway
- Department of Electrical Engineering and Computer Science, University of Stavanger, Stavanger, Norway
| | - Lena Cavallin
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology Karolinska University Hospital, Stockholm, Sweden
| | - Frédéric Blanc
- Service, Memory Resources and Research Centre, University Hospital of Strasbourg, Strasbourg, France
- Team IMIS/Neurocrypto, French National Center for Scientific Research, ICube Laboratory and Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, Strasbourg, France
- Centre Mémoire, de Ressources et de Recherche d'Alsace (Strasbourg-Colmar), Strasbourg, France
| | - Olivier Bousiges
- Centre Mémoire, de Ressources et de Recherche d'Alsace (Strasbourg-Colmar), Strasbourg, France
- Laboratory of Biochemistry and Molecular Biology, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives, UMR7364, University Hospital of Strasbourg, Strasbourg, France
| | - Lars-Olof Wahlund
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Mercè Boada
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau. Biomedical Research Institute (IIB-Sant Pau), Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau. Biomedical Research Institute (IIB-Sant Pau), Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Dag Aarsland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Eric Westman
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
183
|
Restifo LL. Unraveling the Gordian knot: genetics and the troubled road to effective therapeutics for Alzheimer's disease. Genetics 2022; 220:iyab185. [PMID: 34718566 PMCID: PMC8733445 DOI: 10.1093/genetics/iyab185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
In the late 20th century, identification of the major protein components of amyloid plaques and neurofibrillary tangles provided a window into the molecular pathology of Alzheimer's disease, ushering in an era of optimism that targeted therapeutics would soon follow. The amyloid-cascade hypothesis took hold very early, supported by discoveries that dominant mutations in APP, PSEN1, and PSEN2 cause the very rare, early-onset, familial forms of the disease. However, in the past decade, a stunning series of failed Phase-3 clinical trials, testing anti-amyloid antibodies or processing-enzyme inhibitors, prompts the question, What went wrong? The FDA's recent controversial approval of aducanumab, despite widespread concerns about efficacy and safety, only amplifies the question. The assumption that common, late-onset Alzheimer's is a milder form of familial disease was not adequately questioned. The differential timing of discoveries, including blood-brain-barrier-penetrant tracers for imaging of plaques and tangles, made it easy to focus on amyloid. Furthermore, the neuropathology community initially implemented Alzheimer's diagnostic criteria based on plaques only. The discovery that MAPT mutations cause frontotemporal dementia with tauopathy made it even easier to overlook the tangles in Alzheimer's. Many important findings were simply ignored. The accepted mouse models did not predict the human clinical trials data. Given this lack of pharmacological validity, input from geneticists in collaboration with neuroscientists is needed to establish criteria for valid models of Alzheimer's disease. More generally, scientists using genetic model organisms as whole-animal bioassays can contribute to building the pathogenesis network map of Alzheimer's disease.
Collapse
Affiliation(s)
- Linda L Restifo
- Department of Neurology, University of Arizona Health Sciences, Tucson, AZ 85724, USA
- Department of Cellular and Molecular Medicine, University of Arizona Health Sciences, Tucson, AZ 85724, USA
- Department of Neuroscience and Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ 85721, USA
- Graduate Interdisciplinary Program in Genetics, University of Arizona, Tucson, AZ 85719, USA
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, Arizona 85724, USA
- BIO5 Interdisciplinary Research Institute, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
184
|
Llamas-Rodríguez J, Oltmer J, Greve DN, Williams E, Slepneva N, Wang R, Champion S, Lang-Orsini M, Fischl B, Frosch MP, van der Kouwe AJ, Augustinack JC. Entorhinal Subfield Vulnerability to Neurofibrillary Tangles in Aging and the Preclinical Stage of Alzheimer's Disease. J Alzheimers Dis 2022; 87:1379-1399. [PMID: 35491780 PMCID: PMC9198759 DOI: 10.3233/jad-215567] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Neurofibrillary tangle (NFT) accumulation in the entorhinal cortex (EC) precedes the transformation from cognitive controls to mild cognitive impairment and Alzheimer's disease (AD). While tauopathy has been described in the EC before, the order and degree to which the individual subfields within the EC are engulfed by NFTs in aging and the preclinical AD stage is unknown. OBJECTIVE We aimed to investigate substructures within the EC to map the populations of cortical neurons most vulnerable to tau pathology in aging and the preclinical AD stage. METHODS We characterized phosphorylated tau (CP13) in 10 cases at eight well-defined anterior-posterior levels and assessed NFT density within the eight entorhinal subfields (described by Insausti and colleagues) at the preclinical stages of AD. We validated with immunohistochemistry and labeled the NFT density ratings on ex vivo MRIs. We measured subfield cortical thickness and reconstructed the labels as three-dimensional isosurfaces, resulting in anatomically comprehensive, histopathologically validated tau "heat maps." RESULTS We found the lateral EC subfields ELc, ECL, and ECs (lateral portion) to have the highest tau density in semi-quantitative scores and quantitative measurements. We observed significant stepwise higher tau from anterior to posterior levels (p < 0.001). We report an age-dependent anatomically-specific vulnerability, with all cases showing posterior tau pathology, yet older individuals displaying an additional anterior tau burden. Finally, cortical thickness of each subfield negatively correlated with respective tau scores (p < 0.05). CONCLUSION Our findings indicate that posterior-lateral subfields within the EC are the most vulnerable to early NFTs and atrophy in aging and preclinical AD.
Collapse
Affiliation(s)
- Josué Llamas-Rodríguez
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Jan Oltmer
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Douglas N. Greve
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Emily Williams
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Natalya Slepneva
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Ruopeng Wang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Samantha Champion
- Department of Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Melanie Lang-Orsini
- Department of Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Bruce Fischl
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- CSAIL/HST, MIT, Cambridge, MA, USA
| | - Matthew P. Frosch
- Department of Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - André J.W. van der Kouwe
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Jean C. Augustinack
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
185
|
Peng S, Zeng L, Haure-Mirande JV, Wang M, Huffman DM, Haroutunian V, Ehrlich ME, Zhang B, Tu Z. Transcriptomic Changes Highly Similar to Alzheimer's Disease Are Observed in a Subpopulation of Individuals During Normal Brain Aging. Front Aging Neurosci 2021; 13:711524. [PMID: 34924992 PMCID: PMC8675870 DOI: 10.3389/fnagi.2021.711524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is a major risk factor for late-onset Alzheimer’s disease (LOAD). How aging contributes to the development of LOAD remains elusive. In this study, we examined multiple large-scale transcriptomic datasets from both normal aging and LOAD brains to understand the molecular interconnection between aging and LOAD. We found that shared gene expression changes between aging and LOAD are mostly seen in the hippocampal and several cortical regions. In the hippocampus, the expression of phosphoprotein, alternative splicing and cytoskeleton genes are commonly changed in both aging and AD, while synapse, ion transport, and synaptic vesicle genes are commonly down-regulated. Aging-specific changes are associated with acetylation and methylation, while LOAD-specific changes are more related to glycoprotein (both up- and down-regulations), inflammatory response (up-regulation), myelin sheath and lipoprotein (down-regulation). We also found that normal aging brain transcriptomes from relatively young donors (45–70 years old) clustered into several subgroups and some subgroups showed gene expression changes highly similar to those seen in LOAD brains. Using brain transcriptomic datasets from another cohort of older individuals (>70 years), we found that samples from cognitively normal older individuals clustered with the “healthy aging” subgroup while AD samples mainly clustered with the “AD similar” subgroups. This may imply that individuals in the healthy aging subgroup will likely remain cognitively normal when they become older and vice versa. In summary, our results suggest that on the transcriptome level, aging and LOAD have strong interconnections in some brain regions in a subpopulation of cognitively normal aging individuals. This supports the theory that the initiation of LOAD occurs decades earlier than the manifestation of clinical phenotype and it may be essential to closely study the “normal brain aging” to identify the very early molecular events that may lead to LOAD development.
Collapse
Affiliation(s)
- Shouneng Peng
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Lu Zeng
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | | | - Minghui Wang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Derek M Huffman
- Department of Medicine, Albert Einstein College of Medicine, New York City, NY, United States.,Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, United States.,Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York City, NY, United States
| | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Mental Illness Research, Education and Clinical Center (MIRECC), James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, United States
| | - Michelle E Ehrlich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Bin Zhang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Zhidong Tu
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| |
Collapse
|
186
|
Jellinger KA. Recent update on the heterogeneity of the Alzheimer’s disease spectrum. J Neural Transm (Vienna) 2021; 129:1-24. [DOI: 10.1007/s00702-021-02449-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/25/2021] [Indexed: 02/03/2023]
|
187
|
Warren A. Preserved Consciousness in Alzheimer's Disease and Other Dementias: Caregiver Awareness and Communication Strategies. Front Psychol 2021; 12:790025. [PMID: 34950092 PMCID: PMC8688803 DOI: 10.3389/fpsyg.2021.790025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease is an insidious onset neurodegenerative syndrome without effective treatment or cure. It is rapidly becoming a global health crisis that is overwhelming healthcare, society, and individuals. The clinical nature of neurocognitive decline creates significant challenges in bidirectional communication between caregivers and persons with Alzheimer's disease (AD) that can negatively impact quality-of-life. This paper sought to understand how and to what extent would awareness training about the levels of consciousness in AD influence the quality-of-life interactions in the caregiver-patient dyad. A literature review of multiple databases was conducted utilizing a transdisciplinary approach. The sum of findings indicates a positive relationship between enhanced caregiver awareness and training, positive interactions, and improved QOL measures among patients and caregivers. A multidirectional relationship was found among healthcare policies, training and education resources, caregivers, and persons with AD. Specifically, the current lack of policy and inadequate training and educational resources has various detrimental effects on patients and caregivers, while improvements in training and education of caregivers yields positive outcomes in communication and QOL. Furthermore, evidence of preserved consciousness in persons with AD was demonstrated from multiple disciplines, including neurobiological, psychological, and biopsychosocial models. The literature further revealed several methods to access the preserved consciousness in persons with AD and related dementias, including sensory, emotional, and cognitive stimulations. The evidence from the literature suggests a reframed approach to our understanding and treatment of persons with AD is not only warranted, but crucial to address the needs of those affected by AD.
Collapse
Affiliation(s)
- Alison Warren
- The Department of Clinical Research and Leadership, The George Washington University, Washington, DC, United States
| |
Collapse
|
188
|
Aksman LM, Wijeratne PA, Oxtoby NP, Eshaghi A, Shand C, Altmann A, Alexander DC, Young AL. pySuStaIn: a Python implementation of the Subtype and Stage Inference algorithm. SOFTWAREX 2021; 16:100811. [PMID: 34926780 PMCID: PMC8682799 DOI: 10.1016/j.softx.2021.100811] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Progressive disorders are highly heterogeneous. Symptom-based clinical classification of these disorders may not reflect the underlying pathobiology. Data-driven subtyping and staging of patients has the potential to disentangle the complex spatiotemporal patterns of disease progression. Tools that enable this are in high demand from clinical and treatment-development communities. Here we describe the pySuStaIn software package, a Python-based implementation of the Subtype and Stage Inference (SuStaIn) algorithm. SuStaIn unravels the complexity of heterogeneous diseases by inferring multiple disease progression patterns (subtypes) and individual severity (stages) from cross-sectional data. The primary aims of pySuStaIn are to enable widespread application and translation of SuStaIn via an accessible Python package that supports simple extension and generalization to novel modelling situations within a single, consistent architecture.
Collapse
Affiliation(s)
- Leon M Aksman
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
- Centre for Medical Image Computing, Departments of Computer Science and Medical Physics, University College London
| | - Peter A Wijeratne
- Centre for Medical Image Computing, Departments of Computer Science and Medical Physics, University College London
| | - Neil P Oxtoby
- Centre for Medical Image Computing, Departments of Computer Science and Medical Physics, University College London
| | - Arman Eshaghi
- Centre for Medical Image Computing, Departments of Computer Science and Medical Physics, University College London
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London
| | - Cameron Shand
- Centre for Medical Image Computing, Departments of Computer Science and Medical Physics, University College London
| | - Andre Altmann
- Centre for Medical Image Computing, Departments of Computer Science and Medical Physics, University College London
| | - Daniel C Alexander
- Centre for Medical Image Computing, Departments of Computer Science and Medical Physics, University College London
| | - Alexandra L Young
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London
| |
Collapse
|
189
|
Kawakatsu S, Kobayashi R, Hayashi H, Morioka D, Utsunomiya A, Kabasawa T, Ohe R, Otani K. Clinicopathological heterogeneity of Alzheimer's disease with pure Alzheimer's disease pathology: Cases associated with dementia with Lewy bodies, very early-onset dementia, and primary progressive aphasia. Neuropathology 2021; 41:427-449. [PMID: 34816507 DOI: 10.1111/neup.12765] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/05/2021] [Accepted: 05/28/2021] [Indexed: 12/31/2022]
Abstract
We report four cases depicting the heterogeneity of Alzheimer's disease (AD) associated with pure AD pathology. Case 1 was a 77-year-old man with a false positive diagnosis of dementia with Lewy bodies with reduced dopamine transporter uptake activity of the striatum but no Lewy body pathology. There were tau deposits in the large neurons in the putamen, which may be related to the development of parkinsonism. Case 2 was an AD patient in his early 30s who presented with a psychotic episode and a cognitive decline, and later developed myoclonus and seizures. He demonstrated considerable amyloid-beta deposits in the cerebral cortex, including cotton wool plaques, basal ganglia, and cerebellum. Tau deposits were also abundant in the cerebral neocortex, hippocampus, basal ganglia, and brain stem. Case 3 was a 60-year-old woman who exhibited typical symptoms characteristic of the logopenic variant of primary progressive aphasia (lvPPA). Case 4 was a 68-year-old man who exhibited the semantic variant of primary progressive aphasia (svPPA) plus repetition impairment, a rare case associated with AD pathology. In addition to tau pathology, astrocytic pathology was prominent in the white matter and cortical layers of the left temporoparietal cortices. While the main AD lesion in case 4 was evaluated by tau accumulation and astrogliosis in the left temporal lobe, that in case 3 in was evaluated by the same points in the left parietal lobe. Within the spectrum of lvPPA, case 4 may be regarded as a temporal variant of lvPPA presenting svPPA. The pathology of PPA associated with AD may have broader clinical manifestations than that in previously described cases. Case 4 also showed pathological features characteristic of cerebral amyloid angiopathy throughout the cerebral cortex. The distribution of tau and astrocytic pathologies in the cerebral cortex, basal ganglia, brain stem, and cerebellum may explain the various symptoms of atypical pure AD patients.
Collapse
Affiliation(s)
- Shinobu Kawakatsu
- Department of Neuropsychiatry, Aizu Medical Center, Fukushima Medical University, Fukushima, Japan
| | - Ryota Kobayashi
- Department of Psychiatry, Yamagata University School of Medicine, Yamagata, Japan
| | - Hiroshi Hayashi
- Department of Psychiatry, Yamagata University School of Medicine, Yamagata, Japan
| | - Daichi Morioka
- Department of Psychiatry, Yamagata University School of Medicine, Yamagata, Japan
| | - Aya Utsunomiya
- Department of Pathology, Yamagata University School of Medicine, Yamagata, Japan
| | - Takanobu Kabasawa
- Department of Pathology, Yamagata University School of Medicine, Yamagata, Japan
| | - Rintaro Ohe
- Department of Pathology, Yamagata University School of Medicine, Yamagata, Japan
| | - Koichi Otani
- Department of Psychiatry, Yamagata University School of Medicine, Yamagata, Japan
| |
Collapse
|
190
|
White matter variability, cognition, and disorders: a systematic review. Brain Struct Funct 2021; 227:529-544. [PMID: 34731328 PMCID: PMC8844174 DOI: 10.1007/s00429-021-02382-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 09/03/2021] [Indexed: 11/23/2022]
Abstract
Inter-individual differences can inform treatment procedures and—if accounted for—have the potential to significantly improve patient outcomes. However, when studying brain anatomy, these inter-individual variations are commonly unaccounted for, despite reports of differences in gross anatomical features, cross-sectional, and connectional anatomy. Brain connections are essential to facilitate functional organization and, when severed, cause impairments or complete loss of function. Hence, the study of cerebral white matter may be an ideal compromise to capture inter-individual variability in structure and function. We reviewed the wealth of studies that associate cognitive functions and clinical symptoms with individual tracts using diffusion tractography. Our systematic review indicates that tractography has proven to be a sensitive method in neurology, psychiatry, and healthy populations to identify variability and its functional correlates. However, the literature may be biased, as the most commonly studied tracts are not necessarily those with the highest sensitivity to cognitive functions and pathologies. Additionally, the hemisphere of the studied tract is often unreported, thus neglecting functional laterality and asymmetries. Finally, we demonstrate that tracts, as we define them, are not correlated with one, but multiple cognitive domains or pathologies. While our systematic review identified some methodological caveats, it also suggests that tract–function correlations might still be a promising tool in identifying biomarkers for precision medicine. They can characterize variations in brain anatomy, differences in functional organization, and predicts resilience and recovery in patients.
Collapse
|
191
|
Gautherot M, Kuchcinski G, Bordier C, Sillaire AR, Delbeuck X, Leroy M, Leclerc X, Pruvo JP, Pasquier F, Lopes R. Longitudinal Analysis of Brain-Predicted Age in Amnestic and Non-amnestic Sporadic Early-Onset Alzheimer's Disease. Front Aging Neurosci 2021; 13:729635. [PMID: 34803654 PMCID: PMC8596466 DOI: 10.3389/fnagi.2021.729635] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/27/2021] [Indexed: 01/28/2023] Open
Abstract
Objective: Predicted age difference (PAD) is a score computed by subtracting chronological age from "brain" age, which is estimated using neuroimaging data. The goal of this study was to evaluate the PAD as a marker of phenotypic heterogeneity and severity among early-onset Alzheimer's disease (EOAD) patients. Methods: We first used 3D T1-weighted (3D-T1) magnetic resonance images (MRI) of 3,227 healthy subjects aged between 18 and 85 years to train, optimize, and evaluate the brain age model. A total of 123 participants who met the criteria for early-onset (<65 years) sporadic form of probable Alzheimer's disease (AD) and presented with two distinctive clinical presentations [an amnestic form (n = 74) and a non-amnestic form (n = 49)] were included at baseline and followed-up for a maximum period of 4 years. All the participants underwent a work-up at baseline and every year during the follow-up period, which included clinical examination, neuropsychological testing and genotyping, and structural MRI. In addition, cerebrospinal fluid biomarker assay was recorded at baseline. PAD score was calculated by applying brain age model to 3D-T1 images of the EOAD patients and healthy controls, who were matched based on age and sex. At baseline, between-group differences for neuropsychological and PAD scores were assessed using linear models. Regarding longitudinal analysis of neuropsychological and PAD scores, differences between amnestic and non-amnestic participants were analyzed using linear mixed-effects modeling. Results: PAD score was significantly higher for non-amnestic patients (2.35 ± 0.91) when compared to amnestic patients (2.09 ± 0.74) and controls (0.00 ± 1). Moreover, PAD score was linearly correlated with the Mini-Mental State Examination (MMSE) and the Clinical Dementia Rating Sum of Boxes (CDR-SB), for both amnestic and non-amnestic sporadic forms. Longitudinal analyses showed that the gradual development of the disease in patients was accompanied by a significant increase in PAD score over time, for both amnestic and non-amnestic patients. Conclusion: PAD score was able to separate amnestic and non-amnestic sporadic forms. Regardless of the clinical presentation, as PAD score was a way of quantifying an early brain age acceleration, it was an appropriate method to detect the development of AD and follow the evolution of the disease as a marker of severity as MMSE and CDR-SB.
Collapse
Affiliation(s)
- Morgan Gautherot
- UMS 2014–US 41–PLBS–Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
| | - Grégory Kuchcinski
- UMS 2014–US 41–PLBS–Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
- Neuroradiology Department, Lille University Medical Centre, Lille, France
| | - Cécile Bordier
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
| | - Adeline Rollin Sillaire
- Memory Center, DISTALZ, Lille, France
- Neurology Department, Lille University Medical Centre, Lille, France
| | | | - Mélanie Leroy
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
- Memory Center, DISTALZ, Lille, France
| | - Xavier Leclerc
- UMS 2014–US 41–PLBS–Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
- Neuroradiology Department, Lille University Medical Centre, Lille, France
| | - Jean-Pierre Pruvo
- UMS 2014–US 41–PLBS–Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
- Neuroradiology Department, Lille University Medical Centre, Lille, France
| | - Florence Pasquier
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
- Memory Center, DISTALZ, Lille, France
- Neurology Department, Lille University Medical Centre, Lille, France
| | - Renaud Lopes
- UMS 2014–US 41–PLBS–Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
| |
Collapse
|
192
|
Md S, Alhakamy NA, Alfaleh MA, Afzal O, Altamimi ASA, Iqubal A, Shaik RA. Mechanisms Involved in Microglial-Interceded Alzheimer's Disease and Nanocarrier-Based Treatment Approaches. J Pers Med 2021; 11:1116. [PMID: 34834468 PMCID: PMC8619529 DOI: 10.3390/jpm11111116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder accountable for dementia and cognitive dysfunction. The etiology of AD is complex and multifactorial in origin. The formation and deposition of amyloid-beta (Aβ), hyperphosphorylated tau protein, neuroinflammation, persistent oxidative stress, and alteration in signaling pathways have been extensively explored among the various etiological hallmarks. However, more recently, the immunogenic regulation of AD has been identified, and macroglial activation is considered a limiting factor in its etiological cascade. Macroglial activation causes neuroinflammation via modulation of the NLRP3/NF-kB/p38 MAPKs pathway and is also involved in tau pathology via modulation of the GSK-3β/p38 MAPK pathways. Additionally, microglial activation contributes to the discrete release of neurotransmitters and an altered neuronal synaptic plasticity. Therefore, activated microglial cells appear to be an emerging target for managing and treating AD. This review article discussed the pathology of microglial activation in AD and the role of various nanocarrier-based anti-Alzeihmenr's therapeutic approaches that can either reverse or inhibit this activation. Thus, as a targeted drug delivery system, nanocarrier approaches could emerge as a novel means to overcome existing AD therapy limitations.
Collapse
Affiliation(s)
- Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed A. Alfaleh
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.)
| | - Abdulmalik S. A. Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.)
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
| | - Rasheed A. Shaik
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
193
|
Smailovic U, Johansson C, Koenig T, Kåreholt I, Graff C, Jelic V. Decreased Global EEG Synchronization in Amyloid Positive Mild Cognitive Impairment and Alzheimer's Disease Patients-Relationship to APOE ε4. Brain Sci 2021; 11:brainsci11101359. [PMID: 34679423 PMCID: PMC8533770 DOI: 10.3390/brainsci11101359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
The apolipoprotein E (APOE) ε4 allele is a risk factor for Alzheimer's disease (AD) that has been linked to changes in brain structure and function as well as to different biological subtypes of the disease. The present study aimed to investigate the association of APOE ε4 genotypes with brain functional impairment, as assessed by quantitative EEG (qEEG) in patients on the AD continuum. The study population included 101 amyloid positive patients diagnosed with mild cognitive impairment (MCI) (n = 50) and AD (n = 51) that underwent resting-state EEG recording and CSF Aβ42 analysis. In total, 31 patients were APOE ε4 non-carriers, 42 were carriers of one, and 28 were carriers of two APOE ε4 alleles. Quantitative EEG analysis included computation of the global field power (GFP) and global field synchronization (GFS) in conventional frequency bands. Amyloid positive patients who were carriers of APOE ε4 allele(s) had significantly higher GFP beta and significantly lower GFS in theta and beta bands compared to APOE ε4 non-carriers. Increased global EEG power in beta band in APOE ε4 carriers may represent a brain functional compensatory mechanism that offsets global EEG slowing in AD patients. Our findings suggest that decreased EEG measures of global synchronization in theta and beta bands reflect brain functional deficits related to the APOE ε4 genotype in patients that are on a biomarker-verified AD continuum.
Collapse
Affiliation(s)
- Una Smailovic
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14152 Huddinge, Sweden;
- Department of Clinical Neurophysiology, Karolinska University Hospital, 14186 Huddinge, Sweden
- Correspondence:
| | - Charlotte Johansson
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14152 Huddinge, Sweden; (C.J.); (C.G.)
- Clinic for Cognitive Disorders, Karolinska University Hospital, 14186 Huddinge, Sweden
| | - Thomas Koenig
- Translational Research Center, University Hospital of Psychiatry, University of Bern, 3012 Bern, Switzerland;
| | - Ingemar Kåreholt
- Aging Research Centre, Karolinska Institutet and Stockholm University, 17165 Solna, Sweden;
- School of Health and Welfare, Aging Research Network—Jönköping (ARN-J), Institute for Gerontology, Jönköping University, 55111 Jönköping, Sweden
| | - Caroline Graff
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14152 Huddinge, Sweden; (C.J.); (C.G.)
- Unit for Hereditary Dementia, Karolinska University Hospital-Solna, 17176 Solna, Sweden
| | - Vesna Jelic
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14152 Huddinge, Sweden;
- Clinic for Cognitive Disorders, Karolinska University Hospital, 14186 Huddinge, Sweden
| |
Collapse
|
194
|
Shiino A, Shirakashi Y, Ishida M, Tanigaki K. Machine learning of brain structural biomarkers for Alzheimer's disease (AD) diagnosis, prediction of disease progression, and amyloid beta deposition in the Japanese population. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12246. [PMID: 34692983 PMCID: PMC8515359 DOI: 10.1002/dad2.12246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 06/11/2021] [Indexed: 12/30/2022]
Abstract
INTRODUCTION We developed machine learning (ML) designed to analyze structural brain magnetic resonance imaging (MRI), and trained it on the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. In this study, we verified its utility in the Japanese population. METHODS A total of 535 participants were enrolled from the Japanese ADNI database, including 148 AD, 152 normal, and 235 mild cognitive impairment (MCI). Probability of AD was expressed as AD likelihood scores (ADLS). RESULTS The accuracy of AD diagnosis was 88.0% to 91.2%. The accuracy of predicting the disease progression in non-dementia participants over a 3-year observation was 76.0% to 79.3%. More than 90% of the participants with low ADLS did not progress to AD within 3 years. In the amyloid positron emission tomography (PET)-positive MCI, the hazard ratio of progression was 2.39 with low ADLS, and 5.77 with high ADLS. When high ADLS was defined as N+ and Pittsburgh compound B (PiB) PET positivity was defined as A+, the time to disease progression for 50% of MCI participants was 23.7 months in A+N+, whereas it was 52.3 months in A+N-. CONCLUSION These results support the feasibility of our ML for the diagnosis of AD and prediction of the disease progression.
Collapse
Affiliation(s)
- Akihiko Shiino
- Molecular Neuroscience Research CenterShiga University of Medical ScienceShigaJapan
| | - Yoshitomo Shirakashi
- Molecular Neuroscience Research CenterShiga University of Medical ScienceShigaJapan
| | - Manabu Ishida
- Department of NeurologyShimane UniversityShimaneJapan
| | | | | |
Collapse
|
195
|
Katayama T, Sawada J, Takahashi K, Yahara O, Hasebe N. Meta-analysis of cerebrospinal fluid neuron-specific enolase levels in Alzheimer's disease, Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Alzheimers Res Ther 2021; 13:163. [PMID: 34610837 PMCID: PMC8493707 DOI: 10.1186/s13195-021-00907-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 09/23/2021] [Indexed: 12/29/2022]
Abstract
Background This study examined the usefulness of cerebrospinal fluid (CSF) neuron-specific enolase (NSE) levels as a candidate biomarker of neurodegeneration in Alzheimer’s disease (AD), Parkinson’s disease (PD), PD with dementia (PDD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Methods We performed a systematic search of PubMed, the Cochrane Library, Scopus, and Google Scholar to find studies that measured CSF NSE levels in AD, PD, DLB, and/or MSA. For each disease, we pooled all available data and performed a meta-analysis, and meta-regression analyses of age and sex were conducted if the main analysis found a significant association. Results Twenty studies were included (13 for AD, 8 for PD/PDD/DLB, and 4 for MSA). Significantly elevated CSF NSE levels were detected in AD (Hedges’ g = 0.822, 95% confidence interval [95% CI] 0.332 to 1.311, p = 0.0010), but the data exhibited high heterogeneity (I2 = 88.43%, p < 0.001). The meta-regression analysis of AD showed that age (p < 0.001), but not sex, had a significant effect on CSF NSE levels. A meta-analysis of the pooled data for PD/PDD/DLB did not show any significant changes in the CSF NSE level, but a sub-group analysis of PDD/DLB revealed significantly elevated CSF NSE levels (Hedges’ g = 0.507, 95% CI 0.020 to 0.993, p = 0.0412). No significant changes in CSF NSE levels were detected in MSA. Conclusions The CSF NSE level may be a useful biomarker of neurodegeneration in AD and PDD/DLB. Age was found to affect the CSF NSE levels of AD patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00907-3.
Collapse
Affiliation(s)
- Takayuki Katayama
- Department of Neurology, Asahikawa City Hospital, 1-1-65 Kinseicho, Asahikawa, 070-8610, Japan.
| | - Jun Sawada
- Division of Neurology, First Department of Internal Medicine, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Kae Takahashi
- Department of Neurology, Asahikawa City Hospital, 1-1-65 Kinseicho, Asahikawa, 070-8610, Japan
| | - Osamu Yahara
- Department of Neurology, Asahikawa City Hospital, 1-1-65 Kinseicho, Asahikawa, 070-8610, Japan
| | - Naoyuki Hasebe
- Division of Neurology, First Department of Internal Medicine, Asahikawa Medical University Hospital, Asahikawa, Japan
| |
Collapse
|
196
|
Uretsky M, Gibbons LE, Mukherjee S, Trittschuh EH, Fardo DW, Boyle PA, Keene CD, Saykin AJ, Crane PK, Schneider JA, Mez J. Longitudinal cognitive performance of Alzheimer's disease neuropathological subtypes. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12201. [PMID: 34604500 PMCID: PMC8474122 DOI: 10.1002/trc2.12201] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/03/2021] [Accepted: 06/17/2021] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) neuropathological subtypes (limbic predominant [lpAD], hippocampal sparing [HpSpAD], and typical [tAD]), defined by relative neurofibrillary tangle (NFT) burden in limbic and cortical regions, have not been studied in prospectively characterized epidemiological cohorts with robust cognitive assessments. METHODS Two hundred ninety-two participants with neuropathologically confirmed AD from the Religious Orders Study and Memory and Aging Project were categorized by neuropathological subtype based on previously specified diagnostic criteria using quantitative regional NFT counts. Rates of cognitive decline were compared across subtypes using linear mixed-effects models that included subtype, time, and a subtype-time interaction as predictors and four cognitive domain factor scores (memory, executive function, language, visuospatial) and a global score as outcomes. To assess if memory was relatively preserved in HpSpAD, non-memory factor scores were included as covariates in the mixed-effects model with memory as the outcome. RESULTS There were 57 (20%) with lpAD, 22 (8%) with HpSpAD and 213 (73%) with tAD. LpAD died significantly later than the participants with tAD (2.4 years, P = .01) and with HpSpAD (3.8 years, P = .03). Compared to tAD, HpSpAD, but not lpAD, performed significantly worse in all cognitive domains at the time of initial impairment and declined significantly faster in memory, language, and globally. HpSpAD did not have relatively preserved memory performance at any time point. CONCLUSION The relative frequencies of AD neuropathological subtypes in an epidemiological sample were consistent with a previous report in a convenience sample. People with HpSpAD decline rapidly, but may not have a memory-sparing clinical syndrome. Cohort-specific differences in regional tau burden and comorbid neuropathology may explain the lack of clinicopathological correlation.
Collapse
Affiliation(s)
- Madeline Uretsky
- Boston University Alzheimer's Disease and CTE CentersBoston University School of MedicineBostonMassachusettsUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| | - Laura E. Gibbons
- Department of General Internal MedicineUniversity of Washington School of Medicine, University of WashingtonSeattleWashingtonUSA
| | - Shubhabrata Mukherjee
- Department of General Internal MedicineUniversity of Washington School of Medicine, University of WashingtonSeattleWashingtonUSA
| | - Emily H. Trittschuh
- Geriatric Research, Education, and Clinical CenterPuget Sound Veterans Affairs Health Care SystemSeattleWashingtonUSA
- Department of Psychiatry and Behavioral SciencesUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - David W. Fardo
- Sanders‐Brown Center on AgingUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- College of Public Health and Department of BiostatisticsUniversity of KentuckyLexingtonKentuckyUSA
| | - Patricia A. Boyle
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Division of Behavioral SciencesRush Medical CollegeChicagoIllinoisUSA
| | - C. Dirk Keene
- University of Washington Alzheimer's Disease Research CenterUniversity of Washington School of MedicineSeattleWashingtonUSA
- Department of Laboratory Medicine and PathologyUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Andrew J. Saykin
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Radiology and Imaging ServicesIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Paul K. Crane
- Department of General Internal MedicineUniversity of Washington School of Medicine, University of WashingtonSeattleWashingtonUSA
- University of Washington Alzheimer's Disease Research CenterUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Julie A. Schneider
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush Medical College, ChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Jesse Mez
- Boston University Alzheimer's Disease and CTE CentersBoston University School of MedicineBostonMassachusettsUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| |
Collapse
|
197
|
Wu BS, Zhang YR, Li HQ, Kuo K, Chen SD, Dong Q, Liu Y, Yu JT. Cortical structure and the risk for Alzheimer's disease: a bidirectional Mendelian randomization study. Transl Psychiatry 2021; 11:476. [PMID: 34526483 PMCID: PMC8443658 DOI: 10.1038/s41398-021-01599-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/16/2021] [Accepted: 08/27/2021] [Indexed: 02/08/2023] Open
Abstract
Progressive loss of neurons in a specific brain area is one of the manifestations of Alzheimer's disease (AD). Much effort has been devoted to investigating brain atrophy and AD. However, the causal relationship between cortical structure and AD is not clear. We conducted a bidirectional two-sample Mendelian randomization analysis to investigate the causal relationship between cortical structure (surface area and thickness of the whole cortex and 34 cortical regions) and AD risk. Genetic variants used as instruments came from a large genome-wide association meta-analysis of cortical structure (33,992 participants of European ancestry) and AD (AD and AD-by-proxy, 71,880 cases, 383,378 controls). We found suggestive associations of the decreased surface area of the temporal pole (OR (95% CI): 0.95 (0.9, 0.997), p = 0.04), and decreased thickness of cuneus (OR (95% CI): 0.93 (0.89, 0.98), p = 0.006) with higher AD risk. We also found a suggestive association of vulnerability to AD with the decreased surface area of precentral (β (SE): -43.4 (21.3), p = 0.042) and isthmus cingulate (β (SE): -18.5 (7.3), p = 0.011). However, none of the Bonferroni-corrected p values of the causal relationship between cortical structure and AD met the threshold. We show suggestive evidence of an association of the atrophy of the temporal pole and cuneus with higher AD risk. In the other direction, there was a suggestive causal relationship between vulnerability to AD and the decreased surface area of the precentral and isthmus cingulate. Our findings shed light on the associations of cortical structure with the occurrence of AD.
Collapse
Affiliation(s)
- Bang-Sheng Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong-Qi Li
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kevin Kuo
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
198
|
Li D, Zhang L, Nelson NW, Mielke MM, Yu F. Plasma Neurofilament Light and Future Declines in Cognition and Function in Alzheimer's Disease in the FIT-AD Trial. J Alzheimers Dis Rep 2021; 5:601-611. [PMID: 34514342 PMCID: PMC8385429 DOI: 10.3233/adr-210302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2021] [Indexed: 01/27/2023] Open
Abstract
Background: Utilities of blood-based biomarkers in Alzheimer’s disease (AD) clinical trials remain unknown. Objective: To evaluate the ability of plasma neurofilament light chain (NfL) to predict future declines in cognition and activities of daily living (ADL) outcomes in 26 older adults with mild-to-moderate AD dementia from the FIT-AD Trial. Methods: Plasma NfL was measured at baseline and 3 and 6 months. Cognition and ADL were assessed using the AD Assessment Scale-Cognition (ADAS-Cog) and AD Uniform Dataset Instruments and Disability Assessment for Dementia (DAD), respectively, at baseline, 3, 6, 9, and 12 months. Linear mixed effects models were used to examine the associations between baseline or change in plasma NfL and changes in outcomes. Results: Higher baseline plasma NfL was associated with greater rate of decline in ADAS-Cog from baseline to 6 months (standardized estimate of 0.00462, p = 0.02853) and in ADL from baseline to 12 months (standardized estimate of –0.00284, p = 0.03338). Greater increase in plasma NfL in short term from baseline to 3 months was associated with greater rate of decline in memory and ADL from 3 to 6 months (standardized estimate of –0.04638 [0.003], p = 0.01635; standardized estimate of –0.03818, p = 0.0435) and greater rate of decline in ADL from 3 to 12 month (standardized estimate of –0.01492, p = 0.01082). Conclusion: This study demonstrated that plasma NfL might have the potential to predict cognitive and function decline up to 12 months. However, future studies with bigger sample sizes need to confirm the findings.
Collapse
Affiliation(s)
- Danni Li
- Department of Lab Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Lin Zhang
- School of Public Health, Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Nathaniel W Nelson
- Graduate School of Professional Psychology, University of St. Thomas, Minnesota, St. Paul, MN, USA
| | - Michelle M Mielke
- Departments of Health Sciences Research and Neurology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Fang Yu
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| |
Collapse
|
199
|
Yu M, Sporns O, Saykin AJ. The human connectome in Alzheimer disease - relationship to biomarkers and genetics. Nat Rev Neurol 2021; 17:545-563. [PMID: 34285392 PMCID: PMC8403643 DOI: 10.1038/s41582-021-00529-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
The pathology of Alzheimer disease (AD) damages structural and functional brain networks, resulting in cognitive impairment. The results of recent connectomics studies have now linked changes in structural and functional network organization in AD to the patterns of amyloid-β and tau accumulation and spread, providing insights into the neurobiological mechanisms of the disease. In addition, the detection of gene-related connectome changes might aid in the early diagnosis of AD and facilitate the development of personalized therapeutic strategies that are effective at earlier stages of the disease spectrum. In this article, we review studies of the associations between connectome changes and amyloid-β and tau pathologies as well as molecular genetics in different subtypes and stages of AD. We also highlight the utility of connectome-derived computational models for replicating empirical findings and for tracking and predicting the progression of biomarker-indicated AD pathophysiology.
Collapse
Affiliation(s)
- Meichen Yu
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Network Science Institute, Bloomington, IN, USA
| | - Olaf Sporns
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Network Science Institute, Bloomington, IN, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana University Network Science Institute, Bloomington, IN, USA.
| |
Collapse
|
200
|
Zangrossi A, Montemurro S, Altoè G, Mondini S. Heterogeneity and Factorial Structure in Alzheimer's Disease: A Cognitive Perspective. J Alzheimers Dis 2021; 83:1341-1351. [PMID: 34420975 DOI: 10.3233/jad-210719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) patients show heterogeneous cognitive profiles which suggest the existence of cognitive subgroups. A deeper comprehension of this heterogeneity could contribute to move toward a precision medicine perspective. OBJECTIVE In this study, we aimed 1) to investigate AD cognitive heterogeneity as a product of the combination of within- (factors) and between-patients (sub-phenotypes) components, and 2) to promote its assessment in clinical practice by defining a small set of critical tests for this purpose. METHODS We performed factor mixture analysis (FMA) on neurocognitive assessment results of N = 230 patients with a clinical diagnosis of AD. This technique allowed to investigate the structure of cognitive heterogeneity in this sample and to characterize the core features of cognitive sub-phenotypes. Subsequently, we performed a tests selection based on logistic regression to highlight the best tests to detect AD patients in our sample. Finally, the accuracy of the same tests in the discrimination of sub-phenotypes was evaluated. RESULTS FMA revealed a structure characterized by five latent factors and four groups, which were identifiable by means of a few cognitive tests and were mainly characterized by memory deficits with visuospatial difficulties ("Visuospatial AD"), typical AD cognitive pattern ("Typical AD"), less impaired memory ("Mild AD"), and language/praxis deficits with relatively spared memory ("Nonamnestic AD"). CONCLUSION The structure of cognitive heterogeneity in our sample of AD patients, as studied by FMA, could be summarized by four sub-phenotypes with distinct cognitive characteristics easily identifiable in clinical practice. Clinical implications under the precision medicine framework are discussed.
Collapse
Affiliation(s)
- Andrea Zangrossi
- Department of Neuroscience, University of Padua, Padua, Italy.,Padova Neuroscience Center (PNC), University of Padua, Padua, Italy
| | | | - Gianmarco Altoè
- Department of Developmental and Social Psychology, University of Padua, Padua, Italy
| | - Sara Mondini
- Department of Philosophy, Sociology, Pedagogy and Applied Psychology, University of Padua, Padua, Italy.,Human Inspired Technology Research Centre, University of Padua, Padua, Italy
| |
Collapse
|