1
|
Takemoto S, Onuki K, Tanimoto K, Taniguchi M, Suero T, Okamoto M, Kimura S, Osawa M, Takeshige-Amano H, Nishikawa N, Aoki S, Kuwatsuru R, Hattori N, Murakami K. Elapsed time changes of the brain radiopharmaceutical accumulation of the amyloid PET examination using 18F-flutemetamol. Ann Nucl Med 2025:10.1007/s12149-025-02046-3. [PMID: 40253573 DOI: 10.1007/s12149-025-02046-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/30/2025] [Indexed: 04/21/2025]
Abstract
OBJECTIVE The purpose of this study was to examine how the radiopharmaceutical accumulation in the brain changes with the elapsed time between the administration of 18F-flutemetamol and the start of imaging, and to determine its effect on quantitative indices. METHODS The study population consisted of 25 subjects who agreed to participate in the study. After visual evaluation by the radiologist, 14 subjects tested negative for Aβ accumulation, and 11 subjects tested positive as well. The study population was treated with 18F-flutemeamol, and list mode acquisition was performed for 50 min starting at 60 min after the time of administration. From the acquired list data, five PET images were extracted at 10-min intervals from the start to the end of acquisition, a PET image corresponding to 20 min of acquisition from 60 min after administration, and a PET image corresponding to 20 min of acquisition from 90 min after administration, respectively. Pixel values were measured for the PET images created and quantitative indices (pixel value, SUVr, Centiloid scale) were calculated and compared. RESULTS In most of the PET images, pixel values showed a decreasing trend with elapsed time after radiopharmaceutical administration. Accordingly, calculated SUVr and Centiloid Scale also changed. CONCLUSIONS Elapsed time after radiopharmaceutical administration resulted in a washout of the radiopharmaceutical accumulation in the brain. From this, it was suggested that the quantitative indices change.
Collapse
Affiliation(s)
- Shota Takemoto
- Department of Radiology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan.
| | - Koji Onuki
- Department of Radiology, Juntendo Tokyo Koto Geriatric Medical Center, 3‑3‑20, Shinsuna, Koto‑ku, Tokyo, 136‑0075, Japan
| | - Keiko Tanimoto
- Department of Radiology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Masaho Taniguchi
- Department of Radiology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Takako Suero
- Department of Radiology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Mio Okamoto
- Department of Radiology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Satoshi Kimura
- Department of Radiology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Monami Osawa
- Department of Neurology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Haruka Takeshige-Amano
- Department of Neurology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Noriko Nishikawa
- Department of Neurology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Shigeki Aoki
- Department of Radiology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Ryohei Kuwatsuru
- Department of Radiology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| | - Koji Murakami
- Department of Radiology, Juntendo University Hospital, 3‑1‑3, Hongo, Bunkyo‑ku, Tokyo, 113‑8421, Japan
| |
Collapse
|
2
|
Feizpour A, Doré V, Krishnadas N, Bourgeat P, Doecke JD, Saad ZS, Triana-Baltzer G, Laws SM, Shishegar R, Huang K, Fowler C, Ward L, Masters CL, Fripp J, Kolb HC, Villemagne VL, Rowe CC. Alzheimer's disease biological PET staging using plasma p217+tau. COMMUNICATIONS MEDICINE 2025; 5:53. [PMID: 40016526 PMCID: PMC11868538 DOI: 10.1038/s43856-025-00768-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/13/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Plasma phospho-tau biomarkers, such as p217+tau, excel at identifying Alzheimer's disease (AD) neuropathology. However, their ability to substitute for tau PET to identify AD biological stage is unclear. METHODS Participants included 248 cognitively unimpaired (CU) and 227 cognitively impaired (CI) individuals, with Janssen plasma p217+tau Simoa® assay, 18F-NAV4694 Aβ-PET (A) and 18F-MK6240 tau-PET (T) data. Biological PET stages were defined according to the Revised Criteria for Diagnosis and Staging of Alzheimer's Disease (2024): Initial (A + T-), Early (A + TMTL + ), Intermediate (A + TMOD + ), and Advanced (A + THIGH + ). The threshold for A+ was 25 Centiloid and for THIGH + , the 75th percentile SUVRtemporo-parietal in A + CI. Sixty percent were A + , 36% Intermediate/Advanced, and 9% Advanced. The performance of p217+tau in discriminating AD stages was assessed using Receiver Operating Characteristic (ROC) analysis and logistic regression. RESULTS Plasma p217+tau concentrations increase across the AD biological PET stages, except between Initial and Early stages. Screening for all AD stages (vs. A-T-), combined Intermediate/Advanced stages, or Advanced stage yields AUC of 0.92, 0.92, and 0.91, respectively (CI only: AUC 0.93, 0.89, 0.83). Plasma p217+tau Youden threshold provides sensitivity of 0.77 [0.73-0.90], specificity 0.91 [0.80-0.95], PPV 0.84 [0.71-0.89], and NPV 0.88 [0.85-0.93] for combined Intermediate/Advanced stages. For the Advanced stage alone, sensitivity is 0.89 [0.79-0.97], specificity 0.82 [0.75-0.9], NPV 0.99 [0.98-1.0], but PPV is only 0.33 [0.25-0.47]. CONCLUSIONS In addition to accurately screening for A+ individuals, plasma p217+tau is useful for identifying a combined Intermediate/Advanced stage AD cohort or pre-screening to reduce the tau-PET required to identify Advanced stage AD individuals.
Collapse
Affiliation(s)
- Azadeh Feizpour
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
| | - Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
- The Australian e-Health Research Centre, CSIRO, Melbourne, VIC, Australia
| | - Natasha Krishnadas
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
| | - Pierrick Bourgeat
- The Australian e-Health Research Centre, CSIRO, Brisbane, QLD, Australia
| | - James D Doecke
- The Australian e-Health Research Centre, CSIRO, Brisbane, QLD, Australia
- Centre for Precision Health, Edith Cowan University, Perth, WA, Australia
| | - Ziad S Saad
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, USA
| | | | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, Perth, WA, Australia
- Collaborative Genomics and Translation Group, Edith Cowan University, Perth, WA, Australia
- Curtin Medical School, Curtin University, Perth, WA, Australia
| | - Rosita Shishegar
- The Australian e-Health Research Centre, CSIRO, Melbourne, VIC, Australia
| | - Kun Huang
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
| | - Christopher Fowler
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Larry Ward
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Jurgen Fripp
- The Australian e-Health Research Centre, CSIRO, Brisbane, QLD, Australia
| | - Hartmuth C Kolb
- Neuroscience Biomarkers, Janssen Research and Development, La Jolla, CA, USA
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher C Rowe
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia.
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
3
|
Sharmin T, Chatterjee P, Doecke JD, Ashton NJ, Huynh K, Pedrini S, Sohrabi HR, Heng B, Eslick S, Zetterberg H, Blennow K, Garg M, Martins RN. Circulating medium- and long-chain acylcarnitines are associated with plasma P-tau181 in cognitively normal older adults. J Neurochem 2025; 169:10.1111/jnc.16244. [PMID: 39473263 PMCID: PMC11808462 DOI: 10.1111/jnc.16244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/15/2024] [Accepted: 09/27/2024] [Indexed: 02/11/2025]
Abstract
Alzheimer's disease (AD) pathogenesis involves dysregulation in diverse biochemical processes. Nevertheless, plasma tau phosphorylated at threonine 181 (P-tau181), a recognised AD biomarker, has been described to reflect early-stage cortical amyloid-β (Aβ) deposition in cognitively normal (CN) adults. Therefore, identifying changes in plasma metabolites associated with plasma P-tau181 at the pre-clinical stage may provide insights into underlying biochemical mechanisms to better understand initial AD pathogenesis. In the current study, plasma P-tau181, quantified via single molecule array (Simoa) technology, and plasma metabolites, quantified via targeted-mass spectrometry, were investigated for associations in CN older adults and upon stratification by positron emission tomography (PET)-Aβ load. In addition, the P-tau181-linked metabolites were evaluated for cognitive performance and neuroimaging markers of AD and the potential to distinguish between CN Aβ- and CN Aβ+ individuals. Significant positive associations of medium- and long-chain acylcarnitines (ACs) were observed with P-tau181 in the entire cohort, CN Aβ- and CN Aβ+, suggesting a link between initial Aβ pathology and fatty acid oxidation-mediated energy metabolism pathways. However, in CN Aβ-, additional linear associations of P-tau181 were observed with muscle metabolism and nitric oxide homeostasis-associated metabolites. Upon investigating the P-tau181-linked metabolites for cognitive performance, significant inverse correlations of the verbal and visual episodic memory and the global composite score were noted in CN Aβ+ with medium- and long-chain ACs, suggesting prognostic value of ACs accompanying weaker cognitive performance. While investigating neuroimaging markers, ACs had positive associations with PET-Aβ load and inverse associations with hippocampal volume in CN Aβ+, indicating connections of ACs with initial AD pathogenesis. Furthermore, based on receiver operating characteristics analysis, the associated ACs potentially classified PET-Aβ status in older adults. Therefore, plasma P-tau181-linked circulating ACs may serve as potential prognostic markers for initial AD pathogenesis in CN older adults. However, further cross-sectional and longitudinal research in highly characterised AD cohorts is needed to validate current findings.
Collapse
Affiliation(s)
- Tahmida Sharmin
- Macquarie Medical SchoolMacquarie UniversityMacquarie ParkNew South WalesAustralia
- Department of PharmacyUniversity of RajshahiRajshahiBangladesh
| | - Pratishtha Chatterjee
- Macquarie Medical SchoolMacquarie UniversityMacquarie ParkNew South WalesAustralia
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVictoriaAustralia
| | - James D. Doecke
- Australian eHealth Research CentreCSIROBrisbaneQueenslandAustralia
- School of Medical and Health SciencesEdith Cowan UniversityPerthWestern AustraliaAustralia
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Kevin Huynh
- Metabolomics LaboratoryBaker Heart and Diabetes InstituteMelbourneVictoriaAustralia
| | - Steve Pedrini
- School of Medical and Health SciencesEdith Cowan UniversityPerthWestern AustraliaAustralia
- Alzheimer's Research AustraliaNedlandsWestern AustraliaAustralia
| | - Hamid R. Sohrabi
- Macquarie Medical SchoolMacquarie UniversityMacquarie ParkNew South WalesAustralia
- Alzheimer's Research AustraliaNedlandsWestern AustraliaAustralia
- School of PsychologyMurdoch UniversityMurdochWestern AustraliaAustralia
| | - Benjamin Heng
- Macquarie Medical SchoolMacquarie UniversityMacquarie ParkNew South WalesAustralia
| | - Shaun Eslick
- Macquarie Medical SchoolMacquarie UniversityMacquarie ParkNew South WalesAustralia
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong KongChina
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
| | - Manohar Garg
- Macquarie Medical SchoolMacquarie UniversityMacquarie ParkNew South WalesAustralia
| | - Ralph N. Martins
- Macquarie Medical SchoolMacquarie UniversityMacquarie ParkNew South WalesAustralia
- School of Medical and Health SciencesEdith Cowan UniversityPerthWestern AustraliaAustralia
- Alzheimer's Research AustraliaNedlandsWestern AustraliaAustralia
| |
Collapse
|
4
|
Gillman A, Bourgeat P, Cox T, Villemagne VL, Fripp J, Huang K, Williams R, Shishegar R, O'Keefe G, Li S, Krishnadas N, Feizpour A, Bozinovski S, Rowe CC, Doré V. Digital detector PET/CT increases Centiloid measures of amyloid in Alzheimer's disease: A head-to-head comparison of cameras. J Alzheimers Dis 2025; 103:1257-1268. [PMID: 39865687 DOI: 10.1177/13872877241313063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
BACKGROUND The introduction of therapeutics for Alzheimer's disease has led to increased interest in precisely quantifying amyloid-β (Aβ) burden for diagnosis, treatment monitoring, and further clinical research. Recent positron emission tomography (PET) hardware innovations including digital detectors have led to superior resolution and sensitivity, improving quantitative accuracy. However, the effect of PET scanner on Centiloid remains relatively unexplored and is assumed to be minimized by harmonizing PET resolutions. OBJECTIVE To quantify the differences in Centiloid between scanners in a paired cohort. METHODS 36 participants from the Australian Imaging, Biomarker and Lifestyle study (AIBL) cohort were scanned within a year on two scanners. Each participant underwent 18F-NAV4694 imaging on two of the three scanners investigated, the Siemens Vision, the Siemens mCT and the Philips Gemini. We compared Aβ Centiloid quantification between scanners and assessed the effectiveness of post-reconstruction PET resolution harmonization. We further compared the scanner differences in target sub-regions and with different reference regions to assess spatial variability. RESULTS Centiloid from the Vision camera was found to be significantly higher compared to the Gemini and mCT; the difference was greater at high-Centiloid levels. Post-reconstruction resolution harmonization only accounted for and corrected ∼20% of the Centiloid (CL) difference between scanners. We further demonstrated that residual differences have effects that vary spatially between different subregions of the Centiloid mask. CONCLUSIONS We have demonstrated that the type of PET scanner that a participant is scanned on affects Centiloid quantification, even when scanner resolution is harmonized. We conclude by highlighting the need for further investigation into harmonization techniques that consider scanner differences.
Collapse
Affiliation(s)
- Ashley Gillman
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD, Australia
| | - Pierrick Bourgeat
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD, Australia
| | - Timothy Cox
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD, Australia
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
| | - Jurgen Fripp
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD, Australia
| | - Kun Huang
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
| | - Rob Williams
- Melbourne Brain Centre Imaging Unit, The University of Melbourne, Melbourne, VIC, Australia
| | - Rosita Shishegar
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD, Australia
| | - Graeme O'Keefe
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
| | - Shenpeng Li
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD, Australia
| | - Natasha Krishnadas
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Azadeh Feizpour
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Svetlana Bozinovski
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Vincent Doré
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Brisbane, QLD, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Landau SM, Harrison TM, Baker SL, Boswell MS, Lee J, Taggett J, Ward TJ, Chadwick T, Murphy A, DeCarli C, Schwarz CG, Vemuri P, Jack CR, Koeppe RA, Jagust WJ. Positron emission tomography harmonization in the Alzheimer's Disease Neuroimaging Initiative: A scalable and rigorous approach to multisite amyloid and tau quantification. Alzheimers Dement 2025; 21:e14378. [PMID: 39559932 PMCID: PMC11772732 DOI: 10.1002/alz.14378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 11/20/2024]
Abstract
INTRODUCTION A key goal of the Alzheimer's Disease NeuroImaging Initiative (ADNI) positron emission tomography (PET) Core is to harmonize quantification of β-amyloid (Aβ) and tau PET image data across multiple scanners and tracers. METHODS We developed an analysis pipeline (Berkeley PET Imaging Pipeline, B-PIP) for ADNI Aβ and tau PET images and applied it to PET data from other multisite studies. Steps include image pre-processing, refacing, magnetic resonance imaging (MRI)/PET co-registration, visual quality control (QC), quantification of tracer uptake, and standardization of Aβ and tau standardized uptake value ratios (SUVrs) across tracers. RESULTS Measurements from 10,105 cross-sectional and longitudinal Aβ and tau PET scans acquired in several studies between 2010 and 2024 can be processed, harmonized, and directly merged across tracers and cohorts. DISCUSSION The B-PIP developed in ADNI is a scalable image harmonization approach used in several observational studies and clinical trials that facilitates rigorous Aβ and tau PET quantification and data sharing. HIGHLIGHTS Quantitative results from ADNI Aβ and tau PET data are generated using a rigorous, scalable image processing pipeline This pipeline has been applied to PET data from several other large, multisite studies and trials Quantitative outcomes are harmonizable across studies and are shared with the scientific community.
Collapse
Affiliation(s)
- Susan M. Landau
- Neuroscience DepartmentUniversity of CaliforniaBerkeleyCaliforniaUSA
| | | | - Suzanne L. Baker
- Molecular Biophysics and Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Martin S. Boswell
- Molecular Biophysics and Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - JiaQie Lee
- Neuroscience DepartmentUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Jacinda Taggett
- Neuroscience DepartmentUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Tyler J. Ward
- Neuroscience DepartmentUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Trevor Chadwick
- Neuroscience DepartmentUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Alice Murphy
- Neuroscience DepartmentUniversity of CaliforniaBerkeleyCaliforniaUSA
| | | | | | | | | | - Robert A. Koeppe
- Department of RadiologyUniversity of MichiganAnn ArborMichiganUSA
| | - William J. Jagust
- Neuroscience DepartmentUniversity of CaliforniaBerkeleyCaliforniaUSA
- Molecular Biophysics and Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | | |
Collapse
|
6
|
Milligan Armstrong A, O'Brien E, Porter T, Dore V, Bourgeat P, Maruff P, Rowe CC, Villemagne VL, Rainey‐Smith SR, Laws SM. Exploring the relationship between melanopsin gene variants, sleep, and markers of brain health. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70056. [PMID: 39822292 PMCID: PMC11736627 DOI: 10.1002/dad2.70056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/30/2024] [Accepted: 11/26/2024] [Indexed: 01/19/2025]
Abstract
INTRODUCTION Melanopsin is a photopigment with roles in mediating sleep and circadian-related processes, which are often disrupted in Alzheimer's disease (AD). Melanopsin also impacts cognition and synaptogenesis. This study investigated the associations between melanopsin genetic variants, sleep, and markers of brain health. METHODS Linear regression analyses examined the relationship of single-nucleotide polymorphisms (SNPs) within the melanopsin gene (OPN4), with cortical amyloid beta (Aβ), cognition, brain volumes, and self-reported sleep traits in cognitively unimpaired older adults. Further analyses assessed whether sleep traits x OPN4 SNP interactions were associated with markers of brain health. RESULTS OPN4 SNPs rs2355009 and rs3740334 were associated with attention and processing speed and ventricular volume and language, respectively. Furthermore, rs3740334 and rs1079610 showed significant interactions with sleep traits in association with language. DISCUSSION This study shows associations of OPN4 genetic variants with markers of brain health, and suggests that these variants interact with sleep to exacerbate cognitive effects. Highlights The relationships between melanopsin gene (OPN4) variants and markers of brain health were examined cross-sectionally in cognitively unimpaired older individuals.Variation within OPN4is associated with differences in cognition and ventricular volume.rs2355009 and rs3740334 show small-moderate associations with differences in attention and processing speed. Further to this, rs2355009 and rs3740334 were associated with ventricular volumes and language performance, respectively.The interactions between rs3740334 and rs1079610 and sleep traits also showed small-moderate associations with differences in language performance.
Collapse
Affiliation(s)
- Ayeisha Milligan Armstrong
- Centre for Precision HealthEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Collaborative Genomics and Translation Group, School of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Curtin Medical SchoolCurtin University, Kent St.BentleyWestern AustraliaAustralia
| | - Eleanor O'Brien
- Centre for Precision HealthEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Collaborative Genomics and Translation Group, School of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
| | - Tenielle Porter
- Centre for Precision HealthEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Collaborative Genomics and Translation Group, School of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Curtin Medical SchoolCurtin University, Kent St.BentleyWestern AustraliaAustralia
| | - Vincent Dore
- Centre for Precision HealthEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Australian E‐Health Research CentreCSIROHerstonQueenslandAustralia
- Department of Molecular Imaging and Therapy and Centre for PETAustin HealthHeidelbergVictoriaAustralia
| | | | - Paul Maruff
- Australian E‐Health Research CentreCSIROParkvilleVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
- Cogstate Ltd.MelbourneVictoriaAustralia
| | - Christopher C. Rowe
- Department of Molecular Imaging and Therapy and Centre for PETAustin HealthHeidelbergVictoriaAustralia
- Australian E‐Health Research CentreCSIROParkvilleVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoriaAustralia
| | - Victor. L. Villemagne
- Centre for Precision HealthEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Department of Molecular Imaging and Therapy and Centre for PETAustin HealthHeidelbergVictoriaAustralia
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Stephanie R. Rainey‐Smith
- Centre for Healthy Ageing, Health Futures InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
- Alzheimer's Research AustraliaSarich Neuroscience Research InstituteNedlandsWestern AustraliaAustralia
- School of Psychological ScienceUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
- School of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
| | - Simon M. Laws
- Centre for Precision HealthEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Collaborative Genomics and Translation Group, School of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Curtin Medical SchoolCurtin University, Kent St.BentleyWestern AustraliaAustralia
| | | |
Collapse
|
7
|
Sato T, Sawai S, Shimada N. Comparison of the ability of different quantitative indices in 123I-FP-CIT single-photon emission computed tomography to differentiate dopaminergic neurodegenerative disease. Jpn J Radiol 2025; 43:78-90. [PMID: 39235674 PMCID: PMC11717878 DOI: 10.1007/s11604-024-01648-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
PURPOSE By imaging dopamine transporter (DAT) uptake in the striatum, 123I-FP-CIT SPECT can differentiate dopaminergic neurodegenerative disease (dNDD) and non-dNDD, which differ in pathophysiology and clinical management. Our aim was to compare and validate the diagnostic abilities of various 123I-FP-CIT SPECT quantitative indices for dNDD. MATERIALS AND METHODS Distribution volume ratio (DVR) and binding ratio (BR), measures of DAT uptake capacity, were measured by analyzing clinical 123I-FP-CIT SPECT images of 29 patients with dNDD, including dementia with Lewy bodies and Parkinson's disease, and 18 patients with non-dNDD, using Montreal Neurological Institute space-based anatomical standardization and an atlas template, which utilizes statistical parametric mapping. Additionally, we computed the specific binding ratio (SBR) based on Bolt's method and the maximum and mean standardized uptake values (SUVmax and SUVmean, respectively). RESULTS The caudate-to-occipital lobe, putamen-to-occipital lobe, and striatum-to-occipital lobe ratios (COR, POR, and SOR, respectively) on DVR and POR and SOR on BR were significantly lower in dNDD than in non-dNDD, with areas under the ROC curve (AUCs) of 0.941-0.960, showing high diagnostic accuracy for dNDD. However, the AUC of COR on BR was 0.839, indicating lower diagnostic performance. SBR had an AUC of 0.921, while SUVmax and SUVmean had AUCs of 0.906 and 0.900, respectively. Although striatal asymmetry on both DVR and BR exhibited AUCs of 0.728 and 0.734 and asymmetry on SBR showed an AUC of 0.757, the ratio-based DAT quantitative indices were superior. There were strong positive correlations of DVR with BR, DVR with SBR or SUVmax, BR with SBR or SUVmax, and SBR with SUVmax. CONCLUSION COR, POR, and SOR on DVR and POR and SOR on BR were the most useful DAT quantitative indices. These indices can be compared with SBR and SUV, suggesting that comprehensive evaluation improves the diagnostic accuracy of dNDD.
Collapse
Affiliation(s)
- Tomohiro Sato
- Department of Radiology, University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
- Department of Radiology, Chiba Aoba Municipal Hospital, 1273-2 Aoba-cho, Chuo-ku, Chiba City, Chiba, 260-0852, Japan.
| | - Setsu Sawai
- Department of Neurology, Chiba Aoba Municipal Hospital, 1273-2 Aoba-cho, Chuo-ku, Chiba City, Chiba, 260-0852, Japan
| | - Naokazu Shimada
- Department of Radiology, Chiba Aoba Municipal Hospital, 1273-2 Aoba-cho, Chuo-ku, Chiba City, Chiba, 260-0852, Japan
| |
Collapse
|
8
|
Takenaka A, Nihashi T, Sakurai K, Notomi K, Ono H, Inui Y, Ito S, Arahata Y, Takeda A, Ishii K, Ishii K, Ito K, Toyama H, Nakamura A, Kato T. Interrater agreement and variability in visual reading of [18F] flutemetamol PET images. Ann Nucl Med 2025; 39:68-76. [PMID: 39316332 PMCID: PMC11706841 DOI: 10.1007/s12149-024-01977-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVE The purpose of this study was to validate the concordance of visual ratings of [18F] flutemetamol amyloid positron emission tomography (PET) images and to investigate the correlation between the agreement of each rater and the Centiloid (CL) scale. METHODS A total of 192 participants, clinically classified as cognitively normal (CN) (n = 59), mild cognitive impairment (MCI) (n = 65), Alzheimer's disease (AD) (n = 55), or non-AD dementia (n = 13), participated in this study. Three experts conducted visual ratings of the amyloid PET images for all 192 patients, assigning a confidence level to each rating on a three-point scale (certain, probable, or neither). The positive or negative determination of amyloid PET results was made by majority vote. The CL value was calculated using the CapAIBL pipeline. RESULTS Overall, 101 images were determined to be positive, and 91 images were negative. Of the 101 positive images, the three raters were in complete agreement for 92 images and in disagreement for 9 images. Of the 91 negative images, the three raters were in complete agreement for 75 images and in disagreement for 16 images. Interrater reliability among the three experts was particularly high, with both Fleiss' kappa and Conger's kappa measuring 0.83 (0.76-0.89). The CL values of the unanimous positive group were significantly greater than those of the other groups, whereas the CL values of the unanimous negative group were significantly lower than those of the other groups. Images with rater disagreement had intermediate CLs. In cases with a high confidence level, the positive or negative visual ratings were in almost complete agreement. However, as confidence levels decreased, experts' visual ratings became more variable. The lower the confidence level was, the greater the number of cases with disagreement in the visual ratings. CONCLUSION Three experts independently rated 192 amyloid PET images, achieving a high level of interrater agreement. However, in patients with intermediate amyloid accumulation, visual ratings varied. Therefore, determining positive and negative decisions in these patients should be performed with caution.
Collapse
Affiliation(s)
- Akinori Takenaka
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Nihashi
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | | | - Hokuto Ono
- Micron Inc. Imaging Service Dept., Tokyo, Japan
| | - Yoshitaka Inui
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Shinji Ito
- Department of Radiology, Anjo Kosei Hospital, Anjo, Japan
| | - Yutaka Arahata
- Department of Neurology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Akinori Takeda
- Department of Neurology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kazunari Ishii
- Department of Radiology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Kenji Ishii
- Team for Neuroimaging Research, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kengo Ito
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Hiroshi Toyama
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Akinori Nakamura
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Biomarker Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Kato
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan.
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Japan.
| |
Collapse
|
9
|
Mizumura S, Tamamura N, Ebina J, Watanabe H, Hori M. Quantitative evaluation of striatal uptake ratios using an adaptive template registration method for 123I-ioflupane dopamine transporter SPECT. Ann Nucl Med 2024; 38:943-959. [PMID: 39158826 PMCID: PMC11538170 DOI: 10.1007/s12149-024-01968-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/07/2024] [Indexed: 08/20/2024]
Abstract
INTRODUCTION 123I-FP-CIT (123I-Ioflupane) SPECT shows strong accumulation in the striatum, but morphological standardization is challenging due to low accumulation outside the striatum, particularly in subjects with marked striatal decline. In this study, morphological standardization without MRI was achieved using the adaptive template registration (ATR) method to create a subject-specific optimized template with weighted images of normal-type and egg-shape-type templates. The accuracy of a quantitative method for calculating the ratio with nonspecific accumulation in the occipital lobe was evaluated by placing voxels-of-interest (VOI) on standardized images, particularly targeting the striatum. METHODS The average images of eight subjects, demonstrating normal-type and egg-shape-type tracer accumulation in 123I-Ioflupane SPECT, were utilized as normal and disease templates, respectively. The study included 300 subjects that underwent both 123I-Ioflupane SPECT and MRI for the diagnosis of suspected Parkinson's disease or for exclusion diagnosis. Morphological standardization of SPECT images using structural MRI (MRI-based method) was considered the standard of truth (SOT). Three morphological standardizations without MRI were conducted. The first involved conventional morphological standardization using a normal template (fixed template method), the second employed the ATR method, with a weighted template, and the third used the split-ATR method, processing the left and right striatum separately to address asymmetrical accumulation. VOIs were set on the striatum, caudate, putamen as regions of specific accumulation, and on the occipital lobe as a reference region for nonspecific accumulation. RESULTS Results showed significant and robust linearity in the striatal accumulation ratios for all templates when compared with the occipital lobe accumulation ratio when using the MRI-based method. Comparing intra-class correlations for different linearities, the ATR method and split-ATR method demonstrated higher linearity in the striatum, caudate, and putamen. The split-ATR method showed similar improvements, although more linearity than some of the ATR methods; the effectiveness of the Split-ATR method may vary by image quality, and further validation of its effectiveness in diverse asymmetric accumulation cases seemed warranted. CONCLUSION The use of optimized templates, such as the ATR and split-ATR methods, improved reproducibility in fully automated processing and demonstrated superior linearity compared to that of MRI-based method, in the ratio to the occipital lobe. The ATR method, which enables morphological standardization when using SPECT images only, proved highly reproducible for clinical quantitative analysis of striatal accumulation, facilitating its clinical use.
Collapse
Affiliation(s)
- Sunao Mizumura
- Department of Radiology, Toho University Omori Medical Center, 1‑1‑5, Omori‑nishi, Ota‑ku, Tokyo, 143‑8541, Japan.
| | - Naoyuki Tamamura
- Nihon Medi-Physics Co., Ltd., 3‑4‑10, Shinsuna, Koto‑ku, Tokyo, 136‑0075, Japan
| | - Junya Ebina
- Department of Neurology, Toho University Omori Medical Center, 1-1-5 Omori‑nishi, Ota‑ku, Tokyo, Japan
| | - Hikaru Watanabe
- Department of Radiology, Toho University Omori Medical Center, 1‑1‑5, Omori‑nishi, Ota‑ku, Tokyo, 143‑8541, Japan
| | - Masaaki Hori
- Department of Radiology, Toho University Omori Medical Center, 1‑1‑5, Omori‑nishi, Ota‑ku, Tokyo, 143‑8541, Japan
| |
Collapse
|
10
|
Feizpour A, Doecke JD, Doré V, Krishnadas N, Huang K, Bourgeat P, Laws SM, Fowler C, Robertson J, Mackintosh L, Ayton S, Martins R, Rainey-Smith SR, Taddei K, Ward L, Stage E, Bannon AW, Masters CL, Fripp J, Villemagne VL, Rowe CC. Detection and staging of Alzheimer's disease by plasma pTau217 on a high throughput immunoassay platform. EBioMedicine 2024; 109:105405. [PMID: 39437657 PMCID: PMC11536028 DOI: 10.1016/j.ebiom.2024.105405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Plasma phospho-tau 217 (pTau217) assays can accurately detect Alzheimer's disease (AD) pathology, but clinical application is limited by the need for specialised equipment. This study tests the performance of a plasma pTau217 assay performed on the Lumipulse-G® platform, that is in widespread clinical use, for selecting patients for therapy based on β-amyloid (Aβ) status and tau staging. METHODS Participants included 388 individuals with 18F-NAV4694 Aβ-PET and 18F-MK6240 tau-PET. Association of pTau217 with PET was examined using Spearman's correlation. Discriminative performance for Aβ and tau PET status as well as tau staging was assessed using Receiver Operating Characteristic analysis. FINDINGS Plasma pTau217 had a high correlation with both Aβ Centiloid (r = 0.76) and tau SUVRmeta-temporal (r = 0.78). Area under curve (AUC) was 0.93 for Aβ- vs Aβ+ and 0.94 for tau- vs tau+. Applying one threshold (Youden's index), pTau217 was 87% accurate in classification of participants to Aβ- vs Aβ+. Applying two thresholds to classify participants into Low, Indeterminate, and High zones, 17.8% had Indeterminate results and among Low/High zone participants, 92% were correctly classified as Aβ- or Aβ+. The assay accurately discriminated moderate/high neocortical tau from no tau or tau limited to mesial-temporal lobe (AUC 0.97) and high neocortical tau from all others (AUC 0.94). INTERPRETATION Plasma pTau217, measured by the widely-available, fully-automated Lumipulse®, was a strong predictor of both Aβ and tau PET status and demonstrated strong predictive power in identifying individuals likely to benefit the most from anti-Aβ treatments. FUNDING NHMRC grants 1132604, 1140853, 1152623 and AbbVie.
Collapse
Affiliation(s)
- Azadeh Feizpour
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia; Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Victoria, Australia
| | - James David Doecke
- The Australian e-Health Research Centre, CSIRO, Brisbane, Queensland, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Victoria, Australia; The Australian e-Health Research Centre, CSIRO, Melbourne, Victoria, Australia
| | - Natasha Krishnadas
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia; Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Victoria, Australia
| | - Kun Huang
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Victoria, Australia
| | - Pierrick Bourgeat
- The Australian e-Health Research Centre, CSIRO, Brisbane, Queensland, Australia
| | - Simon Matthew Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia; Collaborative Genomics and Translation Group, Edith Cowan University, Joondalup, Western Australia, Australia; Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Christopher Fowler
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Joanne Robertson
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Lucy Mackintosh
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Scott Ayton
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ralph Martins
- Australian Alzheimer's Research Foundation, Nedlands, Perth, Australia
| | - Stephanie Ruth Rainey-Smith
- Australian Alzheimer's Research Foundation, Nedlands, Perth, Australia; Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia
| | - Kevin Taddei
- Australian Alzheimer's Research Foundation, Nedlands, Perth, Australia
| | - Larry Ward
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | | | | | - Colin Louis Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Jurgen Fripp
- The Australian e-Health Research Centre, CSIRO, Brisbane, Queensland, Australia
| | - Victor Luis Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Victoria, Australia; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher Cleon Rowe
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia; Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Diniz E, Santini T, Helmet K, Aizenstein HJ, Ibrahim TS. Cross-modality image translation of 3 Tesla Magnetic Resonance Imaging to 7 Tesla using Generative Adversarial Networks. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.16.24315609. [PMID: 39484249 PMCID: PMC11527090 DOI: 10.1101/2024.10.16.24315609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The rapid advancements in magnetic resonance imaging (MRI) technology have precipitated a new paradigm wherein cross-modality data translation across diverse imaging platforms, field strengths, and different sites is increasingly challenging. This issue is particularly accentuated when transitioning from 3 Tesla (3T) to 7 Tesla (7T) MRI systems. This study proposes a novel solution to these challenges using generative adversarial networks (GANs)-specifically, the CycleGAN architecture-to create synthetic 7T images from 3T data. Employing a dataset of 1112 and 490 unpaired 3T and 7T MR images, respectively, we trained a 2-dimensional (2D) CycleGAN model, evaluating its performance on a paired dataset of 22 participants scanned at 3T and 7T. Independent testing on 22 distinct participants affirmed the model's proficiency in accurately predicting various tissue types, encompassing cerebral spinal fluid, gray matter, and white matter. Our approach provides a reliable and efficient methodology for synthesizing 7T images, achieving a median Dice of 6.82%,7,63%, and 4.85% for Cerebral Spinal Fluid (CSF), Gray Matter (GM), and White Matter (WM), respectively, in the testing dataset, thereby significantly aiding in harmonizing heterogeneous datasets. Furthermore, it delineates the potential of GANs in amplifying the contrast-to-noise ratio (CNR) from 3T, potentially enhancing the diagnostic capability of the images. While acknowledging the risk of model overfitting, our research underscores a promising progression towards harnessing the benefits of 7T MR systems in research investigations while preserving compatibility with existent 3T MR data. This work was previously presented at the ISMRM 2021 conference (Diniz, Helmet, Santini, Aizenstein, & Ibrahim, 2021).
Collapse
Affiliation(s)
- Eduardo Diniz
- Department of Electrical and Computer Engineering, University of Pittsburgh, Pennsylvania, United States
| | - Tales Santini
- Department of Bioengineering, University of Pittsburgh, Pennsylvania, United States
| | - Karim Helmet
- Department of Bioengineering, University of Pittsburgh, Pennsylvania, United States
- Department of Psychiatry, University of Pittsburgh, Pennsylvania, United States
| | - Howard J. Aizenstein
- Department of Bioengineering, University of Pittsburgh, Pennsylvania, United States
- Department of Psychiatry, University of Pittsburgh, Pennsylvania, United States
| | - Tamer S. Ibrahim
- Department of Bioengineering, University of Pittsburgh, Pennsylvania, United States
| |
Collapse
|
12
|
Huang X, Fowler C, Li Y, Li QX, Sun J, Pan Y, Jin L, Perez KA, Dubois C, Lim YY, Drysdale C, Rumble RL, Chinnery HR, Rowe CC, Martins RN, Maruff P, Doecke JD, Lin Y, Belaidi AA, Barnham KJ, Masters CL, Gu BJ. Clearance and transport of amyloid β by peripheral monocytes correlate with Alzheimer's disease progression. Nat Commun 2024; 15:7998. [PMID: 39266542 PMCID: PMC11393069 DOI: 10.1038/s41467-024-52396-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Impaired clearance of amyloid β (Aβ) in late-onset Alzheimer's disease (AD) affects disease progression. The role of peripheral monocytes in Aβ clearance from the central nervous system (CNS) is unclear. We use a flow cytometry assay to identify Aβ-binding monocytes in blood, validated by confocal microscopy, Western blotting, and mass spectrometry. Flow cytometry immunophenotyping and correlation with AD biomarkers are studied in 150 participants from the AIBL study. We also examine monocytes in human cerebrospinal fluid (CSF) and their migration in an APP/PS1 mouse model. The assay reveals macrophage-like Aβ-binding monocytes with high phagocytic potential in both the periphery and CNS. We find lower surface Aβ levels in mild cognitive impairment (MCI) and AD-dementia patients compared to cognitively unimpaired individuals. Monocyte infiltration from blood to CSF and migration from CNS to peripheral lymph nodes and blood are observed. Here we show that Aβ-binding monocytes may play a role in CNS Aβ clearance, suggesting their potential as a biomarker for AD diagnosis and monitoring.
Collapse
Affiliation(s)
- Xin Huang
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
- The Innate Phagocytosis Laboratory, Level 11, Melbourne, Victoria, Australia
| | - Chris Fowler
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Yihan Li
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Qiao-Xin Li
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
- National Dementia Diagnostics Laboratory, The University of Melbourne, Parkville, VIC, Australia
| | - Jiaqi Sun
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Yijun Pan
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Liang Jin
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Keyla A Perez
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Céline Dubois
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Yen Y Lim
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| | - Candace Drysdale
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Rebecca L Rumble
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Holly R Chinnery
- Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
- Lions Eye Institute, Perth, Western Australia, Australia
- Optometry, School of Allied Health, The University of Western Australia, Perth, Australia
| | - Christopher C Rowe
- Department of Nuclear Medicine and Center for PET, Austin Health, Heidelberg, VIC, Australia
| | - Ralph N Martins
- Center of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, WA, Australia
| | - Paul Maruff
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
- Cogstate Ltd., Melbourne, VIC, Australia
| | - James D Doecke
- Health and Biosecurity, Australian E-Health Research Center, CSIRO, Brisbane, QLD, Australia
| | - Yong Lin
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Abdel A Belaidi
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Kevin J Barnham
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.
| | - Ben J Gu
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.
- The Innate Phagocytosis Laboratory, Level 11, Melbourne, Victoria, Australia.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Xia Y, Dore V, Fripp J, Bourgeat P, Laws SM, Fowler CJ, Rainey-Smith SR, Martins RN, Rowe C, Masters CL, Coulson EJ, Maruff P. Association of Basal Forebrain Atrophy With Cognitive Decline in Early Alzheimer Disease. Neurology 2024; 103:e209626. [PMID: 38885444 PMCID: PMC11254448 DOI: 10.1212/wnl.0000000000209626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/09/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVES In early Alzheimer disease (AD), β-amyloid (Aβ) deposition is associated with volume loss in the basal forebrain (BF) and cognitive decline. However, the extent to which Aβ-related BF atrophy manifests as cognitive decline is not understood. This study sought to characterize the relationship between BF atrophy and the decline in memory and attention in patients with early AD. METHODS Participants from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study who completed Aβ-PET imaging and repeated MRI and cognitive assessments were included. At baseline, participants were classified based on their clinical dementia stage and Aβ status, yielding groups that were cognitively unimpaired (CU) Aβ-, CU Aβ+, and mild cognitive impairment (MCI) Aβ+. Linear mixed-effects models were used to assess changes in volumetric measures of BF subregions and the hippocampus and changes in AIBL memory and attention composite scores for each group compared with CU Aβ- participants. Associations between Aβ burden, brain atrophy, and cognitive decline were evaluated and explored further using mediation analyses. RESULTS The cohort included 476 participants (72.6 ± 5.9 years, 55.0% female) with longitudinal data from a median follow-up period of 6.1 years. Compared with the CU Aβ- group (n = 308), both CU Aβ+ (n = 107) and MCI Aβ+ (n = 61) adults showed faster decline in BF and hippocampal volumes and in memory and attention (Cohen d = 0.73-1.74). Rates of atrophy in BF subregions and the hippocampus correlated with cognitive decline, and each individually mediated the impact of Aβ burden on memory and attention decline. When all mediators were considered simultaneously, hippocampal atrophy primarily influenced the effect of Aβ burden on memory decline (β [SE] = -0.139 [0.032], proportion mediated [PM] = 28.0%) while the atrophy of the posterior nucleus basalis of Meynert in the BF (β [SE] = -0.068 [0.029], PM = 13.1%) and hippocampus (β [SE] = -0.121 [0.033], PM = 23.4%) distinctively influenced Aβ-related attention decline. DISCUSSION These findings highlight the significant role of BF atrophy in the complex pathway linking Aβ to cognitive impairment in early stages of AD. Volumetric assessment of BF subregions could be essential in elucidating the relationships between the brain structure and behavior in AD.
Collapse
Affiliation(s)
- Ying Xia
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Vincent Dore
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Jurgen Fripp
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Pierrick Bourgeat
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Simon M Laws
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Christopher J Fowler
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Stephanie R Rainey-Smith
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Ralph N Martins
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Christopher Rowe
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Colin L Masters
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Elizabeth J Coulson
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Paul Maruff
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| |
Collapse
|
14
|
Kim S, Wang SM, Kang DW, Um YH, Han EJ, Park SY, Ha S, Choe YS, Kim HW, Kim REY, Kim D, Lee CU, Lim HK. A Comparative Analysis of Two Automated Quantification Methods for Regional Cerebral Amyloid Retention: PET-Only and PET-and-MRI-Based Methods. Int J Mol Sci 2024; 25:7649. [PMID: 39062892 PMCID: PMC11276670 DOI: 10.3390/ijms25147649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Accurate quantification of amyloid positron emission tomography (PET) is essential for early detection of and intervention in Alzheimer's disease (AD) but there is still a lack of studies comparing the performance of various automated methods. This study compared the PET-only method and PET-and-MRI-based method with a pre-trained deep learning segmentation model. A large sample of 1180 participants in the Catholic Aging Brain Imaging (CABI) database was analyzed to calculate the regional standardized uptake value ratio (SUVR) using both methods. The logistic regression models were employed to assess the discriminability of amyloid-positive and negative groups through 10-fold cross-validation and area under the receiver operating characteristics (AUROC) metrics. The two methods showed a high correlation in calculating SUVRs but the PET-MRI method, incorporating MRI data for anatomical accuracy, demonstrated superior performance in predicting amyloid-positivity. The parietal, frontal, and cingulate importantly contributed to the prediction. The PET-MRI method with a pre-trained deep learning model approach provides an efficient and precise method for earlier diagnosis and intervention in the AD continuum.
Collapse
Affiliation(s)
- Sunghwan Kim
- Department of Psychiatry, College of Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sheng-Min Wang
- Department of Psychiatry, College of Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Dong Woo Kang
- Department of Psychiatry, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yoo Hyun Um
- Department of Psychiatry, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Eun Ji Han
- Division of Nuclear Medicine, Department of Radiology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sonya Youngju Park
- Division of Nuclear Medicine, Department of Radiology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yeong Sim Choe
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Hye Weon Kim
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Regina EY Kim
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Donghyeon Kim
- Research Institute, Neurophet Inc., Seoul 06234, Republic of Korea (R.E.K.)
| | - Chang Uk Lee
- Department of Psychiatry, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, College of Medicine, Yeouido St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
- CMC Institute for Basic Medical Science, The Catholic Medical Center of The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
15
|
Shang C, Sakurai K, Nihashi T, Arahata Y, Takeda A, Ishii K, Ishii K, Matsuda H, Ito K, Kato T, Toyama H, Nakamura A. Comparison of consistency in centiloid scale among different analytical methods in amyloid PET: the CapAIBL, VIZCalc, and Amyquant methods. Ann Nucl Med 2024; 38:460-467. [PMID: 38512444 PMCID: PMC11108942 DOI: 10.1007/s12149-024-01919-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
OBJECTIVE The Centiloid (CL) scale is a standardized measure for quantifying amyloid deposition in amyloid positron emission tomography (PET) imaging. We aimed to assess the agreement among 3 CL calculation methods: CapAIBL, VIZCalc, and Amyquant. METHODS This study included 192 participants (mean age: 71.5 years, range: 50-87 years), comprising 55 with Alzheimer's disease, 65 with mild cognitive impairment, 13 with non-Alzheimer's dementia, and 59 cognitively normal participants. All the participants were assessed using the three CL calculation methods. Spearman's rank correlation, linear regression, Friedman tests, Wilcoxon signed-rank tests, and Bland-Altman analysis were employed to assess data correlations, linear associations, method differences, and systematic bias, respectively. RESULTS Strong correlations (rho = 0.99, p < .001) were observed among the CL values calculated using the three methods. Scatter plots and regression lines visually confirmed these strong correlations and met the validation criteria. Despite the robust correlations, a significant difference in CL value between CapAIBL and Amyquant was observed (36.1 ± 39.7 vs. 34.9 ± 39.4; p < .001). In contrast, no significant differences were found between CapAIBL and VIZCalc or between VIZCalc and Amyquant. The Bland-Altman analysis showed no observable systematic bias between the methods. CONCLUSIONS The study demonstrated strong agreement among the three methods for calculating CL values. Despite minor variations in the absolute values of the Centiloid scores obtained using these methods, the overall agreement suggests that they are interchangeable.
Collapse
Affiliation(s)
- Cong Shang
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Takashi Nihashi
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Yutaka Arahata
- Department of Neurology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Akinori Takeda
- Department of Neurology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kazunari Ishii
- Department of Radiology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Kenji Ishii
- Team for Neuroimaging Research, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Hiroshi Matsuda
- Department of Biofunctional Imaging, Fukushima Medical University, Fukushima, Japan
- Drug Discovery and Cyclotron Research Center, Southern Tohoku Research Institute for Neuroscience, Koriyama, Japan
| | - Kengo Ito
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Kato
- Department of Radiology, National Center for Geriatrics and Gerontology, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan.
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Japan.
| | - Hiroshi Toyama
- Department of Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Akinori Nakamura
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Biomarker Research, National Center for Geriatrics and Gerontology, Obu, Japan
| |
Collapse
|
16
|
Xia Y, Maruff P, Doré V, Bourgeat P, Laws SM, Fowler C, Rainey-Smith SR, Martins RN, Villemagne VL, Rowe CC, Masters CL, Coulson EJ, Fripp J. Longitudinal trajectories of basal forebrain volume in normal aging and Alzheimer's disease. Neurobiol Aging 2023; 132:120-130. [PMID: 37801885 DOI: 10.1016/j.neurobiolaging.2023.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/03/2023] [Accepted: 09/07/2023] [Indexed: 10/08/2023]
Abstract
Dysfunction of the cholinergic basal forebrain (BF) system and amyloid-β (Aβ) deposition are early pathological features in Alzheimer's disease (AD). However, their association in early AD is not well-established. This study investigated the nature and magnitude of volume loss in the BF, over an extended period, in 516 older adults who completed Aβ-PET and serial magnetic resonance imaging scans. Individuals were grouped at baseline according to the presence of cognitive impairment (CU, CI) and Aβ status (Aβ-, Aβ+). Longitudinal volumetric changes in the BF and hippocampus were assessed across groups. The results indicated that high Aβ levels correlated with faster volume loss in the BF and hippocampus, and the effect of Aβ varied within BF subregions. Compared to CU Aβ+ individuals, Aβ-related loss among CI Aβ+ adults was much greater in the predominantly cholinergic subregion of Ch4p, whereas no difference was observed for the Ch1/Ch2 region. The findings support early and substantial vulnerability of the BF and further reveal distinctive degeneration of BF subregions during early AD.
Collapse
Affiliation(s)
- Ying Xia
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, Queensland, Australia.
| | - Paul Maruff
- Cogstate Ltd, Melbourne, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Vincent Doré
- Department of Nuclear Medicine and Centre for PET, Austin Health, Melbourne, Victoria, Australia; The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Melbourne, Victoria, Australia
| | - Pierrick Bourgeat
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, Queensland, Australia
| | - Simon M Laws
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia; Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Christopher Fowler
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Stephanie R Rainey-Smith
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia; Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Ralph N Martins
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Department of Biomedical Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Victor L Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, Melbourne, Victoria, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher C Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Melbourne, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth J Coulson
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Jurgen Fripp
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, Queensland, Australia
| |
Collapse
|
17
|
Jovalekic A, Roé-Vellvé N, Koglin N, Quintana ML, Nelson A, Diemling M, Lilja J, Gómez-González JP, Doré V, Bourgeat P, Whittington A, Gunn R, Stephens AW, Bullich S. Validation of quantitative assessment of florbetaben PET scans as an adjunct to the visual assessment across 15 software methods. Eur J Nucl Med Mol Imaging 2023; 50:3276-3289. [PMID: 37300571 PMCID: PMC10542295 DOI: 10.1007/s00259-023-06279-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023]
Abstract
PURPOSE Amyloid positron emission tomography (PET) with [18F]florbetaben (FBB) is an established tool for detecting Aβ deposition in the brain in vivo based on visual assessment of PET scans. Quantitative measures are commonly used in the research context and allow continuous measurement of amyloid burden. The aim of this study was to demonstrate the robustness of FBB PET quantification. METHODS This is a retrospective analysis of FBB PET images from 589 subjects. PET scans were quantified with 15 analytical methods using nine software packages (MIMneuro, Hermes BRASS, Neurocloud, Neurology Toolkit, statistical parametric mapping (SPM8), PMOD Neuro, CapAIBL, non-negative matrix factorization (NMF), AmyloidIQ) that used several metrics to estimate Aβ load (SUVR, centiloid, amyloid load, and amyloid index). Six analytical methods reported centiloid (MIMneuro, standard centiloid, Neurology Toolkit, SPM8 (PET only), CapAIBL, NMF). All results were quality controlled. RESULTS The mean sensitivity, specificity, and accuracy were 96.1 ± 1.6%, 96.9 ± 1.0%, and 96.4 ± 1.1%, respectively, for all quantitative methods tested when compared to histopathology, where available. The mean percentage of agreement between binary quantitative assessment across all 15 methods and visual majority assessment was 92.4 ± 1.5%. Assessments of reliability, correlation analyses, and comparisons across software packages showed excellent performance and consistent results between analytical methods. CONCLUSION This study demonstrated that quantitative methods using both CE marked software and other widely available processing tools provided comparable results to visual assessments of FBB PET scans. Software quantification methods, such as centiloid analysis, can complement visual assessment of FBB PET images and could be used in the future for identification of early amyloid deposition, monitoring disease progression and treatment effectiveness.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia
| | | | | | | | | | | |
Collapse
|
18
|
Gandia-Ferrero MT, Torres-Espallardo I, Martínez-Sanchis B, Morera-Ballester C, Muñoz E, Sopena-Novales P, González-Pavón G, Martí-Bonmatí L. Objective Image Quality Comparison Between Brain-Dedicated PET and PET/CT Scanners. J Med Syst 2023; 47:88. [PMID: 37589893 DOI: 10.1007/s10916-023-01984-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
As part of a clinical validation of a new brain-dedicated PET system (CMB), image quality of this scanner has been compared to that of a whole-body PET/CT scanner. To that goal, Hoffman phantom and patient data were obtined with both devices. Since CMB does not use a CT for attenuation correction (AC) which is crucial for PET images quality, this study includes the evaluation of CMB PET images using emission-based or CT-based attenuation maps. PET images were compared using 34 image quality metrics. Moreover, a neural network was used to evaluate the degree of agreement between both devices on the patients diagnosis prediction. Overall, results showed that CMB images have higher contrast and recovery coefficient but higher noise than PET/CT images. Although SUVr values presented statistically significant differences in many brain regions, relative differences were low. An asymmetry between left and right hemispheres, however, was identified. Even so, the variations between the two devices were minor. Finally, there is a greater similarity between PET/CT and CMB CT-based AC PET images than between PET/CT and the CMB emission-based AC PET images.
Collapse
Affiliation(s)
- Maria Teresa Gandia-Ferrero
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell, València, 46026, Spain.
| | - Irene Torres-Espallardo
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell, València, 46026, Spain
- Nuclear Medicine Department, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell, València, 46026, Spain
| | - Begoña Martínez-Sanchis
- Nuclear Medicine Department, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell, València, 46026, Spain
| | | | - Enrique Muñoz
- Oncovision, Carrer de Jeroni de Montsoriu, 92, València, 46022, Spain
| | - Pablo Sopena-Novales
- Nuclear Medicine Department, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell, València, 46026, Spain
| | | | - Luis Martí-Bonmatí
- Biomedical Imaging Research Group (GIBI230), La Fe Health Research Institute (IIS La Fe), Avenida Fernando Abril Martorell, València, 46026, Spain
- Radiology Department, La Fe University and Polytechnic Hospital, Avenida Fernando Abril Martorell, València, 46026, Spain
| |
Collapse
|
19
|
Kurihara M, Komatsu H, Sengoku R, Shibukawa M, Morimoto S, Matsubara T, Arakawa A, Orita M, Ishibashi K, Mitsutake A, Shibata S, Ishiura H, Adachi K, Ohse K, Hatano K, Ihara R, Higashihara M, Nishina Y, Tokumaru AM, Ishii K, Saito Y, Murayama S, Kanemaru K, Iwata A. CSF P-Tau181 and Other Biomarkers in Patients With Neuronal Intranuclear Inclusion Disease. Neurology 2023; 100:e1009-e1019. [PMID: 36517236 PMCID: PMC9990848 DOI: 10.1212/wnl.0000000000201647] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/11/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES CSF tau phosphorylated at threonine 181 (p-tau181) is a widely used biomarker for Alzheimer disease (AD) and has recently been regarded to reflect β-amyloid and/or p-tau deposition in the AD brain. Neuronal intranuclear inclusion disease (NIID) is a neurodegenerative disease characterized by intranuclear inclusions in neurons, glial cells, and other somatic cells. Symptoms include dementia, neuropathy, and others. CSF biomarkers were not reported. The objective of this study was to investigate whether CSF biomarkers including p-tau181 are altered in patients with NIID. METHODS This was a retrospective observational study. CSF concentrations of p-tau181, total tau, amyloid-beta 1-42 (Aβ42), monoamine metabolites homovanillic acid (HVA), and 5-hydroxyindole acetic acid (5-HIAA) were compared between 12 patients with NIID, 120 patients with Alzheimer clinical syndrome biologically confirmed based on CSF biomarker profiles, and patients clinically diagnosed with other neurocognitive disorders (dementia with Lewy bodies [DLB], 24; frontotemporal dementia [FTD], 13; progressive supranuclear palsy [PSP], 21; and corticobasal syndrome [CBS], 13). Amyloid PET using Pittsburgh compound B (PiB) was performed in 6 patients with NIID. RESULTS The mean age of patients with NIID, AD, DLB, FTD, PSP, and CBS was 71.3, 74.6, 76.8, 70.2, 75.5, and 71.9 years, respectively. CSF p-tau181 was significantly higher in NIID (72.7 ± 24.8 pg/mL) compared with DLB, PSP, and CBS and was comparable between NIID and AD. CSF p-tau181 was above the cutoff value (50.0 pg/mL) in 11 of 12 patients with NIID (91.7%). Within these patients, only 2 patients showed decreased CSF Aβ42, and these patients showed negative or mild local accumulation in PiB PET, respectively. PiB PET scans were negative in the remaining 4 patients tested. The proportion of patients with increased CSF p-tau181 and normal Aβ42 (A-T+) was significantly higher in NIID (75%) compared with DLB, PSP, and CBS (4.2%, 4.8%, and 7.7%, respectively). CSF HVA and 5-HIAA concentrations were significantly higher in patients with NIID compared with disease controls. DISCUSSION CSF p-tau181 was increased in patients with NIID without amyloid accumulation. Although the deposition of p-tau has not been reported in NIID brains, the molecular mechanism of tau phosphorylation or secretion of p-tau may be altered in NIID.
Collapse
Affiliation(s)
- Masanori Kurihara
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Hiroki Komatsu
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Renpei Sengoku
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Mari Shibukawa
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Satoru Morimoto
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Tomoyasu Matsubara
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Akira Arakawa
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Makoto Orita
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Kenji Ishibashi
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Akihiko Mitsutake
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Shota Shibata
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Hiroyuki Ishiura
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Kaori Adachi
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Kensuke Ohse
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Keiko Hatano
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Ryoko Ihara
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Mana Higashihara
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Yasushi Nishina
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Aya Midori Tokumaru
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Kenji Ishii
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Yuko Saito
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Shigeo Murayama
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Kazutomi Kanemaru
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Atsushi Iwata
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan.
| |
Collapse
|
20
|
Tarawneh HY, Jayakody DMP, Verma S, Doré V, Xia Y, Mulders WHAM, Martins RN, Sohrabi HR. Auditory Event-Related Potentials in Older Adults with Subjective Memory Complaints. J Alzheimers Dis 2023; 92:1093-1109. [PMID: 36847006 DOI: 10.3233/jad-221119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
BACKGROUND Auditory event-related potentials (AERPs) have been suggested as possible biomarkers for the early diagnosis of Alzheimer's disease (AD). However, no study has investigated AERP measures in individuals with subjective memory complaints (SMCs), who have been suggested to be at a pre-clinical stage of AD. OBJECTIVE This study investigated whether AERPs in older adults with SMC can be used to objectively identify those at high risk of developing AD. METHODS AERPs were measured in older adults. Presence of SMC was determined using the Memory Assessment Clinics Questionnaire (MAC-Q). Hearing thresholds using pure-tone audiometry, neuropsychological data, levels of amyloid-β burden and Apolipoprotein E (APOE)ɛ genotype were also obtained A classic two-tone discrimination (oddball) paradigm was used to elicit AERPs (i.e., P50, N100, P200, N200, and P300). RESULTS Sixty-two individuals (14 male, mean age 71.9±5.2 years) participated in this study, of which, 43 (11 male, mean age 72.4±5.5 years) were SMC and 19 (3 male, mean age 70.8±4.3 years) were non-SMC (controls). P50 latency was weakly but significantly correlated with MAC-Q scores. In addition, P50 latencies were significantly longer in Aβ+ individuals compared to Aβ- individuals. CONCLUSION Results suggest that P50 latencies may be a useful tool to identify individuals at higher risk (i.e., participants with high Aβ burden) of developing measurable cognitive decline. Further longitudinal and cross-sectional studies in a larger cohort on SMC individuals are warranted to determine if AERP measures could be of significance for the detection of pre-clinical AD.
Collapse
Affiliation(s)
- Hadeel Y Tarawneh
- School of Human Sciences, The University of Western Australia, Perth, Australia.,Ear Science Institute Australia, Perth, Australia
| | - Dona M P Jayakody
- Ear Science Institute Australia, Perth, Australia.,Ear Science Centre, School of Surgery, The University of Western Australia, Perth, Australia
| | - Shipra Verma
- Department of Geriatric Medicine, Fiona Stanley and Fremantle Hospital, Perth, Australia.,Department of Nuclear Medicine, Fiona Stanley and Royal Perth Hospital, Perth, Australia
| | - Vincent Doré
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Melbourne, Victoria, Australia.,Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Victoria, Australia
| | - Ying Xia
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, Queensland, Australia
| | | | - Ralph N Martins
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Hamid R Sohrabi
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.,Centre for Healthy Ageing, The Health Futures Institute, Murdoch University, Perth, Australia
| |
Collapse
|
21
|
Chatterjee P, Doré V, Pedrini S, Krishnadas N, Thota R, Bourgeat P, Ikonomovic MD, Rainey-Smith SR, Burnham SC, Fowler C, Taddei K, Mulligan R, Ames D, Masters CL, Fripp J, Rowe CC, Martins RN, Villemagne VL. Plasma Glial Fibrillary Acidic Protein Is Associated with 18F-SMBT-1 PET: Two Putative Astrocyte Reactivity Biomarkers for Alzheimer's Disease. J Alzheimers Dis 2023; 92:615-628. [PMID: 36776057 PMCID: PMC10041433 DOI: 10.3233/jad-220908] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
BACKGROUND Astrocyte reactivity is an early event along the Alzheimer's disease (AD) continuum. Plasma glial fibrillary acidic protein (GFAP), posited to reflect astrocyte reactivity, is elevated across the AD continuum from preclinical to dementia stages. Monoamine oxidase-B (MAO-B) is also elevated in reactive astrocytes observed using 18F-SMBT-1 PET in AD. OBJECTIVE The objective of this study was to evaluate the association between the abovementioned astrocyte reactivity biomarkers. METHODS Plasma GFAP and Aβ were measured using the Simoa ® platform in participants who underwent brain 18F-SMBT-1 and Aβ-PET imaging, comprising 54 healthy control (13 Aβ-PET+ and 41 Aβ-PET-), 11 mild cognitively impaired (3 Aβ-PET+ and 8 Aβ-PET-) and 6 probable AD (5 Aβ-PET+ and 1 Aβ-PET-) individuals. Linear regressions were used to assess associations of interest. RESULTS Plasma GFAP was associated with 18F-SMBT-1 signal in brain regions prone to early Aβ deposition in AD, such as the supramarginal gyrus (SG), posterior cingulate (PC), lateral temporal (LT) and lateral occipital cortex (LO). After adjusting for age, sex, APOE ɛ4 genotype, and soluble Aβ (plasma Aβ 42/40 ratio), plasma GFAP was associated with 18F-SMBT-1 signal in the SG, PC, LT, LO, and superior parietal cortex (SP). On adjusting for age, sex, APOE ɛ4 genotype and insoluble Aβ (Aβ-PET), plasma GFAP was associated with 18F-SMBT-1 signal in the SG. CONCLUSION There is an association between plasma GFAP and regional 18F-SMBT-1 PET, and this association appears to be dependent on brain Aβ load.
Collapse
Affiliation(s)
- Pratishtha Chatterjee
- Macquarie Medical School, Macquarie University, North Ryde, New South Wales, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Vincent Doré
- The Australian eHealth Research Centre, CSIRO, Brisbane, Queensland, Australia.,Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia
| | - Steve Pedrini
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | - Natasha Krishnadas
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Rohith Thota
- Macquarie Medical School, Macquarie University, North Ryde, New South Wales, Australia.,School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, New South Wales, Australia
| | - Pierrick Bourgeat
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Queensland, Australia
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, Pennsylvania, PA, USA.,Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, PA, USA
| | - Stephanie R Rainey-Smith
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia.,School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia
| | - Samantha C Burnham
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Queensland, Australia
| | - Christopher Fowler
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Kevin Taddei
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| | - Rachel Mulligan
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia
| | - David Ames
- National Ageing Research Institute, Parkville, Victoria, Australia.,Academic Unit for Psychiatry of Old Age, University of Melbourne, Melbourne, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Jürgen Fripp
- The Australian eHealth Research Centre, CSIRO, Brisbane, Queensland, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia.,The Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ralph N Martins
- Macquarie Medical School, Macquarie University, North Ryde, New South Wales, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia.,School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Victoria, Australia.,Department of Psychiatry, University of Pittsburgh, Pennsylvania, PA, USA
| | | |
Collapse
|
22
|
Landau SM, Ward TJ, Murphy A, Iaccarino L, Harrison TM, La Joie R, Baker S, Koeppe RA, Jagust WJ. Quantification of amyloid beta and tau PET without a structural MRI. Alzheimers Dement 2023; 19:444-455. [PMID: 35429219 DOI: 10.1002/alz.12668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Relying on magnetic resonance imaging (MRI) for quantification of positron emission tomography (PET) images may limit generalizability of the results. We evaluated several MRI-free approaches for amyloid beta (Aβ) and tau PET quantification relative to MRI-dependent quantification cross-sectionally and longitudinally. METHODS We compared baseline MRI-free and MRI-dependent measurements of Aβ PET ([18F]florbetapir [FBP], N = 1290, [18F]florbetaben [FBB], N = 290) and tau PET ([18F]flortaucipir [FTP], N = 768) images with respect to continuous and dichotomous agreement, effect sizes of Aβ+ impaired versus Aβ- unimpaired groups, and longitudinal standardized uptake value ratio (SUVR) slopes in a subset of individuals. RESULTS The best-performing MRI-free approaches had high continuous and dichotomous agreement with MRI-dependent SUVRs for Aβ PET and temporal flortaucipir (R2 ≥0.95; ± agreement ≥92%) and for Alzheimer's disease-related effect sizes; agreement was slightly lower for entorhinal flortaucipir and longitudinal slopes. DISCUSSION There is no consistent loss of baseline or longitudinal AD-related signal with MRI-free Aβ and tau PET image quantification.
Collapse
Affiliation(s)
- Susan M Landau
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, USA
| | - Tyler J Ward
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, USA
| | - Alice Murphy
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, USA
| | - Leonardo Iaccarino
- Memory and Aging Center, University of California, San Francisco, California, USA
| | - Theresa M Harrison
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, USA
| | - Renaud La Joie
- Memory and Aging Center, University of California, San Francisco, California, USA
| | - Suzanne Baker
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Robert A Koeppe
- Division of Nuclear Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, California, USA.,Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | | |
Collapse
|
23
|
Dhiman K, Villemagne VL, Fowler C, Bourgeat P, Li QX, Collins S, Rowe CC, Masters CL, Ames D, Blennow K, Zetterberg H, Martins RN, Gupta V. Cerebrospinal fluid levels of fatty acid-binding protein 3 are associated with likelihood of amyloidopathy in cognitively healthy individuals. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12377. [PMID: 36479019 PMCID: PMC9719998 DOI: 10.1002/dad2.12377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 12/12/2022]
Abstract
Introduction Fatty acid-binding protein 3 (FABP3) is a biomarker of neuronal membrane disruption, associated with lipid dyshomeostasis-a notable Alzheimer's disease (AD) pathophysiological change. We assessed the association of cerebrospinal fluid (CSF) FABP3 levels with brain amyloidosis and the likelihood/risk of developing amyloidopathy in cognitively healthy individuals. Methods FABP3 levels were measured in CSF samples of cognitively healthy participants, > 60 years of age (n = 142), from the Australian Imaging, Biomarkers & Lifestyle Flagship Study of Ageing (AIBL). Results FABP3 levels were positively associated with baseline brain amyloid beta (Aβ) load as measured by standardized uptake value ratio (SUVR, standardized β = 0.22, P = .009) and predicted the change in brain Aβ load (standardized β = 0.32, P = .004). Higher levels of CSF FABP3 (above median) were associated with a likelihood of amyloidopathy (odds ratio [OR] 2.28, 95% confidence interval [CI] 1.12 to 4.65, P = .023). Discussion These results support inclusion of CSF FABP3 as a biomarker in risk-prediction models of AD.
Collapse
Affiliation(s)
- Kunal Dhiman
- IMPACT - The Institute for Mental and Physical Health and Clinical Translation School of Medicine Deakin University Geelong Victoria Australia
- Western Health Partnership School of Nursing and Midwifery (Centre for Quality and Patient Safety Research in the Institute of Health Transformation) Faculty of Health Deakin University Melbourne Victoria Australia
- School of Medical and Health Sciences Edith Cowan University Joondalup Western Australia Australia
| | - Victor L Villemagne
- Department of Psychiatry University of Pittsburgh Pittsburgh Pennsylvania USA
- Department of Molecular Imaging & Therapy and Centre for PET Austin Health Heidelberg Victoria Australia
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
| | - Christopher Fowler
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria Australia
| | - Pierrick Bourgeat
- Australian e-Health Research Centre CSIRO Health and Biosecurity Brisbane Queensland Australia
| | - Qiao-Xin Li
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria Australia
| | - Steven Collins
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy and Centre for PET Austin Health Heidelberg Victoria Australia
- Department of Medicine The University of Melbourne Melbourne Victoria Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health The University of Melbourne Parkville Victoria Australia
| | - David Ames
- National Ageing Research Institute Parkville Victoria Australia
- Academic Unit for Psychiatry of Old age St. George's Hospital The University of Melbourne Melbourne Victoria Australia
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology the Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Gothenburg Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry Institute of Neuroscience and Physiology the Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
- Clinical Neurochemistry Laboratory Sahlgrenska University Hospital Gothenburg Sweden
- Department of Neurodegenerative Disease UCL Queen Square Institute of Neurology London UK
- UK Dementia Research Institute at UCL London UK
- Hong Kong Center for Neurodegenerative Diseases Hong Kong China
| | - Ralph N Martins
- School of Medical and Health Sciences Edith Cowan University Joondalup Western Australia Australia
- Australian Alzheimer's Research Foundation Ralph and Patricia Sarich Neuroscience Research Institute Nedlands Western Australia Australia
- Department of Biomedical Sciences Macquarie University Sydney New South Wales Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia Perth Western Australia Australia
- KaRa Institute of Neurological Diseases Sydney New South Wales Australia
- Co-operative Research Centre for Mental Health Carlton Victoria Australia
| | - Veer Gupta
- IMPACT - The Institute for Mental and Physical Health and Clinical Translation School of Medicine Deakin University Geelong Victoria Australia
- School of Medical and Health Sciences Edith Cowan University Joondalup Western Australia Australia
| |
Collapse
|
24
|
Bourgeat P, Doré V, Burnham SC, Benzinger T, Tosun D, Li S, Goyal M, LaMontagne P, Jin L, Rowe CC, Weiner MW, Morris JC, Masters CL, Fripp J, Villemagne VL. β-amyloid PET harmonisation across longitudinal studies: Application to AIBL, ADNI and OASIS3. Neuroimage 2022; 262:119527. [PMID: 35917917 PMCID: PMC9550562 DOI: 10.1016/j.neuroimage.2022.119527] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 10/31/2022] Open
Abstract
INTRODUCTION The Centiloid scale was developed to harmonise the quantification of β-amyloid (Aβ) PET images across tracers, scanners, and processing pipelines. However, several groups have reported differences across tracers and scanners even after centiloid conversion. In this study, we aim to evaluate the impact of different pre and post-processing harmonisation steps on the robustness of longitudinal Centiloid data across three large international cohort studies. METHODS All Aβ PET data in AIBL (N = 3315), ADNI (N = 3442) and OASIS3 (N = 1398) were quantified using the MRI-based Centiloid standard SPM pipeline and the PET-only pipeline CapAIBL. SUVR were converted into Centiloids using each tracer's respective transform. Global Aβ burden from pre-defined target cortical regions in Centiloid units were quantified for both raw PET scans and PET scans smoothed to a uniform 8 mm full width half maximum (FWHM) effective smoothness. For Florbetapir, we assessed the performance of using both the standard Whole Cerebellum (WCb) and a composite white matter (WM)+WCb reference region. Additionally, our recently proposed quantification based on Non-negative Matrix Factorisation (NMF) was applied to all spatially and SUVR normalised images. Correlation with clinical severity measured by the Mini-Mental State Examination (MMSE) and effect size, as well as tracer agreement in 11C-PiB-18F-Florbetapir pairs and longitudinal consistency were evaluated. RESULTS The smoothing to a uniform resolution partially reduced longitudinal variability, but did not improve inter-tracer agreement, effect size or correlation with MMSE. Using a Composite reference region for 18F-Florbetapir improved inter-tracer agreement, effect size, correlation with MMSE, and longitudinal consistency. The best results were however obtained when using the NMF method which outperformed all other quantification approaches in all metrics used. CONCLUSIONS FWHM smoothing has limited impact on longitudinal consistency or outliers. A Composite reference region including subcortical WM should be used for computing both cross-sectional and longitudinal Florbetapir Centiloid. NMF improves Centiloid quantification on all metrics examined.
Collapse
Affiliation(s)
| | - Vincent Doré
- CSIRO Health and Biosecurity, Brisbane, Australia; Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia
| | | | | | - Duygu Tosun
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA,; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Shenpeng Li
- CSIRO Health and Biosecurity, Brisbane, Australia
| | - Manu Goyal
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, USA
| | - Liang Jin
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Michael W Weiner
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA,; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - John C Morris
- Washington University in St. Louis, St. Louis, MO, USA
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Brisbane, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia; Department of Psychiatry, The University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Ruwanpathirana GP, Williams RC, Masters CL, Rowe CC, Johnston LA, Davey CE. Mapping the association between tau-PET and Aβ-amyloid-PET using deep learning. Sci Rep 2022; 12:14797. [PMID: 36042256 PMCID: PMC9427855 DOI: 10.1038/s41598-022-18963-6] [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: 02/04/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022] Open
Abstract
In Alzheimer’s disease, the molecular pathogenesis of the extracellular Aβ-amyloid (Aβ) instigation of intracellular tau accumulation is poorly understood. We employed a high-resolution PET scanner, with low detection thresholds, to examine the Aβ-tau association using a convolutional neural network (CNN), and compared results to a standard voxel-wise linear analysis. The full range of Aβ Centiloid values was highly predicted by the tau topography using the CNN (training R2 = 0.86, validation R2 = 0.75, testing R2 = 0.72). Linear models based on tau-SUVR identified widespread positive correlations between tau accumulation and Aβ burden throughout the brain. In contrast, CNN analysis identified focal clusters in the bilateral medial temporal lobes, frontal lobes, precuneus, postcentral gyrus and middle cingulate. At low Aβ levels, information from the middle cingulate, frontal lobe and precuneus regions was more predictive of Aβ burden, while at high Aβ levels, the medial temporal regions were more predictive of Aβ burden. The data-driven CNN approach revealed new associations between tau topography and Aβ burden.
Collapse
Affiliation(s)
- Gihan P Ruwanpathirana
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC, Australia.,Melbourne Brain Centre Imaging Unit, The University of Melbourne, Melbourne, VIC, Australia
| | - Robert C Williams
- Melbourne Brain Centre Imaging Unit, The University of Melbourne, Melbourne, VIC, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia.,Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.,Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Leigh A Johnston
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC, Australia.,Melbourne Brain Centre Imaging Unit, The University of Melbourne, Melbourne, VIC, Australia
| | - Catherine E Davey
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC, Australia. .,Melbourne Brain Centre Imaging Unit, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
26
|
Li Y, Huang X, Fowler C, Lim YY, Laws SM, Faux N, Doecke JD, Trounson B, Pertile K, Rumble R, Doré V, Villemagne VL, Rowe CC, Wiley JS, Maruff P, Masters CL, Gu BJ. Identification of Leukocyte Surface P2X7 as a Biomarker Associated with Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23147867. [PMID: 35887215 PMCID: PMC9322488 DOI: 10.3390/ijms23147867] [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: 06/17/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) has shown altered immune responses in the periphery. We studied P2X7 (a proinflammatory receptor and a scavenger receptor) and two integrins, CD11b and CD11c, on the surface of circulating leukocytes and analysed their associations with Aβ-PET, brain atrophy, neuropsychological assessments, and cerebrospinal fluid (CSF) biomarkers. Total 287 age-matched, sex-balanced participants were recruited in a discovery cohort and two validation cohorts through the AIBL study and studied using tri-colour flow cytometry. Our results demonstrated reduced expressions of P2X7, CD11b, and CD11c on leukocytes, particularly monocytes, in Aβ +ve cases compared with Aβ -ve controls. P2X7 and integrin downregulation was observed at pre-clinical stage of AD and stayed low throughout disease course. We further constructed a polygenic risk score (PRS) model based on 12 P2RX7 risk alleles to assess the genetic impact on P2X7 function in AIBL and ADNI cohorts. No significant association was identified between the P2RX7 gene and AD, indicating that P2X7 downregulation in AD is likely caused by environmental changes rather than genetic factors. In conclusion, the downregulation of P2X7 and integrins at pre-clinical stage of AD indicates altered pro-inflammatory responses, phagocytic functions, and migrating capabilities of circulating monocytes in early AD pathogenesis. Our study not only improves our understanding of peripheral immune involvement in early stage of AD but also provides more insights into novel biomarker development, diagnosis, and prognosis of AD.
Collapse
Affiliation(s)
- Yihan Li
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Xin Huang
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Christopher Fowler
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Yen Y. Lim
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC 3168, Australia; (Y.Y.L.); (V.D.)
| | - Simon M. Laws
- School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA 6027, Australia;
| | - Noel Faux
- Melbourne Data Analytics Platform, Petascale Campus Initiative, The University of Melbourne, 21 Bedford St., North Melbourne, VIC 3051, Australia;
| | - James D. Doecke
- The Australian e-Health Research Centre, CSIRO, Brisbane, QLD 4029, Australia;
| | - Brett Trounson
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Kelly Pertile
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Rebecca Rumble
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Vincent Doré
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC 3168, Australia; (Y.Y.L.); (V.D.)
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC 3084, Australia; (V.L.V.); (C.C.R.)
- Department of Medicine, The University of Melbourne, Melbourne, VIC 3084, Australia
| | - Victor L. Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC 3084, Australia; (V.L.V.); (C.C.R.)
- Department of Medicine, The University of Melbourne, Melbourne, VIC 3084, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Christopher C. Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, VIC 3084, Australia; (V.L.V.); (C.C.R.)
- Department of Medicine, The University of Melbourne, Melbourne, VIC 3084, Australia
| | - James S. Wiley
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Paul Maruff
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
- CogState Ltd., Melbourne, VIC 3001, Australia
| | - Colin L. Masters
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
| | - Ben J. Gu
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia; (Y.L.); (X.H.); (C.F.); (B.T.); (K.P.); (R.R.); (J.S.W.); (P.M.); (C.L.M.)
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Correspondence: ; Tel.: +61-3-9035-6317
| |
Collapse
|
27
|
Imabayashi E, Tamamura N, Yamaguchi Y, Kamitaka Y, Sakata M, Ishii K. Automated semi-quantitative amyloid PET analysis technique without MR images for Alzheimer's disease. Ann Nucl Med 2022; 36:865-875. [PMID: 35821311 PMCID: PMC9515054 DOI: 10.1007/s12149-022-01769-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/19/2022] [Indexed: 11/11/2022]
Abstract
Objective Although beta-amyloid (Aβ) positron emission tomography (PET) images are interpreted visually as positive or negative, approximately 10% are judged as equivocal in Alzheimer’s disease. Therefore, we aimed to develop an automated semi-quantitative analysis technique using 18F-flutemetamol PET images without anatomical images. Methods Overall, 136 cases of patients administered 18F-flutemetamol were enrolled. Of 136 cases, five PET images each with the highest and lowest values of standardized uptake value ratio (SUVr) of cerebral cortex-to-pons were used to create positive and negative templates. Using these templates, PET images of the remaining 126 cases were standardized, and SUVr images were produced with the pons as a reference region. The mean of SUVr values in the volume of interest delineated on the cerebral cortex was compared to those in the CortexID Suite (GE Healthcare). Furthermore, centiloid (CL) values were calculated for the 126 cases using data from the Centiloid Project (http://www.gaain.org/centiloid-project) and both templates. 18F-flutemetamol-PET was interpreted visually as positive/negative based on Aβ deposition in the cortex. However, the criterion "equivocal" was added for cases with focal or mild Aβ accumulation that were difficult to categorize. Optimal cutoff values of SUVr and CL maximizing sensitivity and specificity for Aβ detection were determined by receiver operating characteristic (ROC) analysis using the visual evaluation as a standard of truth. Results SUVr calculated by our method and CortexID were highly correlated (R2 = 0.9657). The 126 PET images comprised 84 negative and 42 positive cases of Aβ deposition by visual evaluation, of which 11 and 10 were classified as equivocal, respectively. ROC analyses determined the optimal cutoff values, sensitivity, and specificity for SUVr as 0.544, 89.3%, and 92.9%, respectively, and for CL as 12.400, 94.0%, and 92.9%, respectively. Both semi-quantitative analyses showed that 12 and 9 of the 21 equivocal cases were negative and positive, respectively, under the optimal cutoff values. Conclusions This semi-quantitative analysis technique using 18F-flutemetamol-PET calculated SUVr and CL automatically without anatomical images. Moreover, it objectively and homogeneously interpreted positive or negative Aβ burden in the brain as a supplemental tool for the visual reading of equivocal cases in routine clinical practice. Supplementary Information The online version contains supplementary material available at 10.1007/s12149-022-01769-x.
Collapse
Affiliation(s)
- Etsuko Imabayashi
- Research Team for Neuroimaging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan.,Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage, Chiba, 263-8555, Japan
| | - Naoyuki Tamamura
- Nihon Medi-Physics Co., Ltd., 3-4-10 Shinsuna, Koto-ku, Tokyo, 136-0075, Japan
| | - Yuzuho Yamaguchi
- Nihon Medi-Physics Co., Ltd., 3-4-10 Shinsuna, Koto-ku, Tokyo, 136-0075, Japan
| | - Yuto Kamitaka
- Research Team for Neuroimaging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Muneyuki Sakata
- Research Team for Neuroimaging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan.
| |
Collapse
|
28
|
Krishnadas N, Huang K, Schultz SA, Doré V, Bourgeat P, Goh AM, Lamb F, Bozinovski S, Burnham SC, Robertson JS, Laws SM, Maruff P, Masters CL, Villemagne VL, Rowe CC. Visually Identified Tau 18F-MK6240 PET Patterns in Symptomatic Alzheimer’s Disease. J Alzheimers Dis 2022; 88:1627-1637. [PMID: 35811517 PMCID: PMC9484111 DOI: 10.3233/jad-215558] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: In Alzheimer’s disease, heterogeneity has been observed in the postmortem distribution of tau neurofibrillary tangles. Visualizing the topography of tau in vivo may facilitate clinical trials and clinical practice. Objective: This study aimed to investigate whether tau distribution patterns that are limited to mesial temporal lobe (MTL)/limbic regions, and those that spare MTL regions, can be visually identified using 18F-MK6240, and whether these patterns are associated with different demographic and cognitive profiles. Methods: Tau 18F-MK6240 PET images of 151 amyloid-β positive participants with mild cognitive impairment (MCI) and dementia were visually rated as: tau negative, limbic predominant (LP), MTL-sparing, and Typical by two readers. Groups were evaluated for differences in age, APOE ɛ4 carriage, hippocampal volumes, and cognition (MMSE, composite memory and non-memory scores). Voxel-wise contrasts were also performed. Results: Visual rating resulted in 59.6% classified as Typical, 17.9% as MTL-sparing, 9.9% LP, and 12.6% as tau negative. Intra-rater and inter-rater reliability was strong (Cohen’s kappa values of 0.89 and 0.86 respectively). Tracer retention in a “hook”-like distribution on sagittal sequences was observed in the LP and Typical groups. The visually classified MTL-sparing group had lower APOE ɛ4 carriage and relatively preserved hippocampal volumes. Higher MTL tau was associated with greater amnestic cognitive impairment. High cortical tau was associated with greater impairments on non-memory domains of cognition, and individuals with high cortical tau were more likely to have dementia than MCI. Conclusion: Tau distribution patterns can be visually identified using 18F-MK6240 PET and are associated with differences in APOE ɛ4 carriage, hippocampal volumes, and cognition.
Collapse
Affiliation(s)
- Natasha Krishnadas
- Florey Department of Neurosciences & Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Kun Huang
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Stephanie A. Schultz
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Vincent Doré
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Melbourne, Victoria, Australia
| | - Pierrick Bourgeat
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Brisbane, QLD, Australia
| | - Anita M.Y. Goh
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
- National Ageing Research Institute, Parkville, VIC, Australia
| | - Fiona Lamb
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Svetlana Bozinovski
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Samantha C. Burnham
- Health and Biosecurity Flagship, The Australian eHealth Research Centre, Melbourne, Victoria, Australia
| | - Joanne S. Robertson
- Florey Institute of Neurosciences & Mental Health, Parkville, VIC, Australia
| | - Simon M. Laws
- Centre for Precision Health, Edith Cowan University, Perth, WA, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Paul Maruff
- Florey Institute of Neurosciences & Mental Health, Parkville, VIC, Australia
| | - Colin L. Masters
- Florey Institute of Neurosciences & Mental Health, Parkville, VIC, Australia
| | - Victor L. Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher C. Rowe
- Florey Department of Neurosciences & Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, VIC, Australia
- Florey Institute of Neurosciences & Mental Health, Parkville, VIC, Australia
| |
Collapse
|
29
|
Milicic L, Vacher M, Porter T, Doré V, Burnham SC, Bourgeat P, Shishegar R, Doecke J, Armstrong NJ, Tankard R, Maruff P, Masters CL, Rowe CC, Villemagne VL, Laws SM. Comprehensive analysis of epigenetic clocks reveals associations between disproportionate biological ageing and hippocampal volume. GeroScience 2022; 44:1807-1823. [PMID: 35445885 PMCID: PMC9213584 DOI: 10.1007/s11357-022-00558-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/30/2022] [Indexed: 12/21/2022] Open
Abstract
The concept of age acceleration, the difference between biological age and chronological age, is of growing interest, particularly with respect to age-related disorders, such as Alzheimer's Disease (AD). Whilst studies have reported associations with AD risk and related phenotypes, there remains a lack of consensus on these associations. Here we aimed to comprehensively investigate the relationship between five recognised measures of age acceleration, based on DNA methylation patterns (DNAm age), and cross-sectional and longitudinal cognition and AD-related neuroimaging phenotypes (volumetric MRI and Amyloid-β PET) in the Australian Imaging, Biomarkers and Lifestyle (AIBL) and the Alzheimer's Disease Neuroimaging Initiative (ADNI). Significant associations were observed between age acceleration using the Hannum epigenetic clock and cross-sectional hippocampal volume in AIBL and replicated in ADNI. In AIBL, several other findings were observed cross-sectionally, including a significant association between hippocampal volume and the Hannum and Phenoage epigenetic clocks. Further, significant associations were also observed between hippocampal volume and the Zhang and Phenoage epigenetic clocks within Amyloid-β positive individuals. However, these were not validated within the ADNI cohort. No associations between age acceleration and other Alzheimer's disease-related phenotypes, including measures of cognition or brain Amyloid-β burden, were observed, and there was no association with longitudinal change in any phenotype. This study presents a link between age acceleration, as determined using DNA methylation, and hippocampal volume that was statistically significant across two highly characterised cohorts. The results presented in this study contribute to a growing literature that supports the role of epigenetic modifications in ageing and AD-related phenotypes.
Collapse
Affiliation(s)
- Lidija Milicic
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, 6027, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
| | - Michael Vacher
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, 6027, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- CSIRO Health and Biosecurity, Australian E-Health Research Centre, Floreat, Western Australia, 6014, Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, 6027, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, 6102, Australia
| | - Vincent Doré
- Australian E-Health Research Centre, CSIRO, Parkville, Victoria, 3052, Australia
- Department of Molecular Imaging and Therapy and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Samantha C Burnham
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, 6027, Australia
- Australian E-Health Research Centre, CSIRO, Parkville, Victoria, 3052, Australia
| | - Pierrick Bourgeat
- Australian E-Health Research Centre, CSIRO, Herston, Queensland, 4029, Australia
| | - Rosita Shishegar
- Australian E-Health Research Centre, CSIRO, Parkville, Victoria, 3052, Australia
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| | - James Doecke
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, 6027, Australia
- Australian E-Health Research Centre, CSIRO, Herston, Queensland, 4029, Australia
| | - Nicola J Armstrong
- Department of Mathematics and Statistics, Curtin University, Bentley, Western Australia, Australia
| | - Rick Tankard
- School of Mathematics and Statistics, Murdoch University, Perth, Western Australia, Australia
| | - Paul Maruff
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
- Cogstate Ltd, Melbourne, VIC, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Victor L Villemagne
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, 6027, Australia
- Department of Molecular Imaging and Therapy and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, 6027, Australia.
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, 6027, Australia.
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, 6102, Australia.
| |
Collapse
|
30
|
Spatial normalization and quantification approaches of PET imaging for neurological disorders. Eur J Nucl Med Mol Imaging 2022; 49:3809-3829. [PMID: 35624219 DOI: 10.1007/s00259-022-05809-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/19/2022] [Indexed: 12/17/2022]
Abstract
Quantification approaches of positron emission tomography (PET) imaging provide user-independent evaluation of pathophysiological processes in living brains, which have been strongly recommended in clinical diagnosis of neurological disorders. Most PET quantification approaches depend on spatial normalization of PET images to brain template; however, the spatial normalization and quantification approaches have not been comprehensively reviewed. In this review, we introduced and compared PET template-based and magnetic resonance imaging (MRI)-aided spatial normalization approaches. Tracer-specific and age-specific PET brain templates were surveyed between 1999 and 2021 for 18F-FDG, 11C-PIB, 18F-Florbetapir, 18F-THK5317, and etc., as well as adaptive PET template methods. Spatial normalization-based PET quantification approaches were reviewed, including region-of-interest (ROI)-based and voxel-wise quantitative methods. Spatial normalization-based ROI segmentation approaches were introduced, including manual delineation on template, atlas-based segmentation, and multi-atlas approach. Voxel-wise quantification approaches were reviewed, including voxel-wise statistics and principal component analysis. Certain concerns and representative examples of clinical applications were provided for both ROI-based and voxel-wise quantification approaches. At last, a recipe for PET spatial normalization and quantification approaches was concluded to improve diagnosis accuracy of neurological disorders in clinical practice.
Collapse
|
31
|
Xia Y, Eeles E, Fripp J, Pinsker D, Thomas P, Latter M, Doré V, Fazlollahi A, Bourgeat P, Villemagne VL, Coulson EJ, Rose S. Reduced cortical cholinergic innervation measured using [ 18F]-FEOBV PET imaging correlates with cognitive decline in mild cognitive impairment. Neuroimage Clin 2022; 34:102992. [PMID: 35344804 PMCID: PMC8958543 DOI: 10.1016/j.nicl.2022.102992] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/06/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022]
Abstract
Topographic FEOBV binding correlates with domain-specific cognitive performance. Global and regional reductions in cholinergic innervation are observed in MCI. Global FEOBV SUVR is associated with basal forebrain and hippocampal volumes. Our results provide proof of concept for FEOBV PET to assess cholinergic terminal integrity.
Dysfunction of the cholinergic basal forebrain (BF) neurotransmitter system, including cholinergic axon denervation of the cortex, plays an important role in cognitive decline and dementia. A validated method to directly quantify cortical cholinergic terminal integrity enables exploration of the involvement of this system in diverse cognitive profiles associated with dementia, particularly at a prodromal stage. In this study, we used the radiotracer [18F]-fluoroethoxybenzovesamicol (FEOBV) as a direct measure of cholinergic terminal integrity and investigated its value for the assessment of cholinergic denervation in the cortex and associated cognitive deficits. Eighteen participants (8 with mild cognitive impairment (MCI) and 10 cognitively unimpaired controls) underwent neuropsychological assessment and brain imaging using FEOBV and [18F]-florbetaben for amyloid-β imaging. The MCI group showed a significant global reduction of FEOBV retention in the cortex and in the parietal and occipital cortices specifically compared to the control group. The global cortical FEOBV retention of all participants positively correlated with the BF, hippocampus and grey matter volumes, but no association was found between the global FEOBV retention and amyloid-β status. Topographic profiles from voxel-wise analysis of FEOBV images revealed significant positive correlations with the cognitive domains associated with the underlying cortical areas. Overlapping profiles of decreased FEOBV were identified in correlation with impairment in executive function, attention and language, which covered the anterior cingulate gyrus, olfactory cortex, calcarine cortex, middle temporal gyrus and caudate nucleus. However, the absence of cortical atrophy in these areas suggested that reduced cholinergic terminal integrity in the cortex is an important factor underlying the observed cognitive decline in early dementia. Our results provide support for the utility and validity of FEOBV PET for quantitative assessment of region-specific cholinergic terminal integrity that could potentially be used for early detection of cholinergic dysfunction in dementia following further validation in larger cohorts.
Collapse
Affiliation(s)
- Ying Xia
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia.
| | - Eamonn Eeles
- Internal Medicine Service, The Prince Charles Hospital, Brisbane, QLD, Australia; School of Medicine, Northside Clinical School, The Prince Charles Hospital, Brisbane, QLD, Australia; Dementia & Neuro Mental Health Research Unit, UQCCR, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Jurgen Fripp
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| | - Donna Pinsker
- Internal Medicine Service, The Prince Charles Hospital, Brisbane, QLD, Australia; School of Psychology, The University of Queensland, Brisbane, QLD, Australia
| | - Paul Thomas
- Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Melissa Latter
- Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Vincent Doré
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia; Austin Health, Melbourne, VIC, Australia
| | - Amir Fazlollahi
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Pierrick Bourgeat
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| | - Victor L Villemagne
- Austin Health, Melbourne, VIC, Australia; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Elizabeth J Coulson
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Stephen Rose
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| |
Collapse
|
32
|
Unified spatial normalization method of brain PET images using adaptive probabilistic brain atlas. Eur J Nucl Med Mol Imaging 2022; 49:3073-3085. [PMID: 35258689 DOI: 10.1007/s00259-022-05752-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/01/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE A unique advantage of the brain positron emission tomography (PET) imaging is the ability to image different biological processes with different radiotracers. However, the diversity of the brain PET image patterns also makes their spatial normalization challenging. Since structural MR images are not always available in the clinical practice, this study proposed a PET-only spatial normalization method based on adaptive probabilistic brain atlas. METHODS The proposed method (atlas-based method) consists of two parts, an adaptive probabilistic brain atlas generation algorithm, and a probabilistic framework for registering PET image to the generated atlas. To validate this method, the results of MRI-based method and template-based method (a widely used PET-only method) were treated as the gold standard and control, respectively. A total of 286 brain PET images, including seven radiotracers (FDG, PIB, FBB, AV-45, AV-1451, AV-133, [18F]altanserin) and four groups of subjects (Alzheimer disease, Parkinson disease, frontotemporal dementia, and healthy control), were spatially normalized using the three methods. The results were then quantitatively compared by using correlation analysis, meta region of interest (meta-ROI) standardized uptake value ratio (SUVR) analysis, and statistical parametric mapping (SPM) analysis. RESULTS The Pearson correlation coefficient between the images computed by atlas-based method and the gold standard was 0.908 ± 0.005. The relative error of meta-ROI SUVR computed by atlas-based method was 2.12 ± 0.18%. Compared with template-based method, atlas-based method was also more consistent with the gold standard in SPM analysis. CONCLUSION The proposed method provides a unified approach to spatially normalize brain PET images of different radiotracers accurately without MR images. A free MATLAB toolbox for this method has been provided.
Collapse
|
33
|
Lee J, Ha S, Kim REY, Lee M, Kim D, Lim HK. Development of Amyloid PET Analysis Pipeline Using Deep Learning-Based Brain MRI Segmentation—A Comparative Validation Study. Diagnostics (Basel) 2022; 12:diagnostics12030623. [PMID: 35328176 PMCID: PMC8947654 DOI: 10.3390/diagnostics12030623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 02/04/2023] Open
Abstract
Amyloid positron emission tomography (PET) scan is clinically essential for the non-invasive assessment of the presence and spatial distribution of amyloid-beta deposition in subjects with cognitive impairment suspected to have been a result of Alzheimer’s disease. Quantitative assessment can enhance the interpretation reliability of PET scan; however, its clinical application has been limited due to the complexity of preprocessing. This study introduces a novel deep-learning-based approach for SUVR quantification that simplifies the preprocessing step and significantly reduces the analysis time. Using two heterogeneous amyloid ligands, our proposed method successfully distinguished standardized uptake value ratio (SUVR) between amyloidosis-positive and negative groups. The proposed method’s intra-class correlation coefficients were 0.97 and 0.99 against PETSurfer and PMOD, respectively. The difference of global SUVRs between the proposed method and PETSurfer or PMOD were 0.04 and −0.02, which are clinically acceptable. The AUC-ROC exceeded 0.95 for three tools in the amyloid positive assessment. Moreover, the proposed method had the fastest processing time and had a low registration failure rate (1%). In conclusion, our proposed method calculates SUVR that is consistent with PETSurfer and PMOD, and has advantages of fast processing time and low registration failure rate. Therefore, PET quantification provided by our proposed method can be used in clinical practice.
Collapse
Affiliation(s)
- Jiyeon Lee
- Research Institute, Neurophet Inc., Seoul 06234, Korea; (J.L.); (R.E.Y.K.); (M.L.)
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Regina E. Y. Kim
- Research Institute, Neurophet Inc., Seoul 06234, Korea; (J.L.); (R.E.Y.K.); (M.L.)
| | - Minho Lee
- Research Institute, Neurophet Inc., Seoul 06234, Korea; (J.L.); (R.E.Y.K.); (M.L.)
| | - Donghyeon Kim
- Research Institute, Neurophet Inc., Seoul 06234, Korea; (J.L.); (R.E.Y.K.); (M.L.)
- Correspondence: (D.K.); (H.K.L.); Tel.: +82-10-9361-3781 (D.K.); +82-10-3797-6315 (H.K.L.)
| | - Hyun Kook Lim
- Department of Psychiatry, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea
- Correspondence: (D.K.); (H.K.L.); Tel.: +82-10-9361-3781 (D.K.); +82-10-3797-6315 (H.K.L.)
| |
Collapse
|
34
|
Koncz R, Thalamuthu A, Wen W, Catts VS, Dore V, Lee T, Mather KA, Slavin MJ, Wegner EA, Jiang J, Trollor JN, Ames D, Villemagne VL, Rowe CC, Sachdev PS. The heritability of amyloid burden in older adults: the Older Australian Twins Study. J Neurol Neurosurg Psychiatry 2022; 93:303-308. [PMID: 34921119 DOI: 10.1136/jnnp-2021-326677] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To determine the proportional genetic contribution to the variability of cerebral β-amyloid load in older adults using the classic twin design. METHODS Participants (n=206) comprising 61 monozygotic (MZ) twin pairs (68 (55.74%) females; mean age (SD): 71.98 (6.43) years), and 42 dizygotic (DZ) twin pairs (56 (66.67%) females; mean age: 71.14 (5.15) years) were drawn from the Older Australian Twins Study. Participants underwent detailed clinical and neuropsychological evaluations, as well as MRI, diffusion tensor imaging (DTI) and amyloid PET scans. Fifty-eight participants (17 MZ pairs, 12 DZ pairs) had PET scans with 11Carbon-Pittsburgh Compound B, and 148 participants (44 MZ pairs, 30 DZ pairs) with 18Fluorine-NAV4694. Cortical amyloid burden was quantified using the centiloid scale globally, as well as the standardised uptake value ratio (SUVR) globally and in specific brain regions. Small vessel disease (SVD) was quantified using total white matter hyperintensity volume on MRI, and peak width of skeletonised mean diffusivity on DTI. Heritability (h2) and genetic correlations were measured with structural equation modelling under the best fit model, controlling for age, sex, tracer and scanner. RESULTS The heritability of global amyloid burden was moderate (0.41 using SUVR; 0.52 using the centiloid scale) and ranged from 0.20 to 0.54 across different brain regions. There were no significant genetic or environmental correlations between global amyloid burden and markers of SVD. CONCLUSION Amyloid deposition, the hallmark early feature of Alzheimer's disease, is under moderate genetic influence, suggesting a major environmental contribution that may be amenable to intervention.
Collapse
Affiliation(s)
- Rebecca Koncz
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia .,Specialty of Psychiatry, Faculty of Medicine and Health, The University of Sydney, Concord, New South Wales, Australia
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia
| | - Wei Wen
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia
| | - Vibeke S Catts
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia
| | - Vincent Dore
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia.,The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Parkville, Victoria, Australia
| | - Teresa Lee
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia.,Neuropsychiatric Institute, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Randwick, New South Wales, Australia
| | - Melissa J Slavin
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia
| | - Eva A Wegner
- Department of Nuclear Medicine and PET, Prince of Wales Hospital, Randwick, New South Wales, Australia.,Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jiyang Jiang
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia
| | - Julian N Trollor
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia.,Department of Developmental Disability Neuropsychiatry, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia
| | - David Ames
- Academic Unit for Psychiatry of Old Age, University of Melbourne, Kew, Victoria, Australia.,National Ageing Research Institute, Parkville, Victoria, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia.,Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Sydney, Sydney, New South Wales, Australia.,Neuropsychiatric Institute, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | | |
Collapse
|
35
|
Woodfield A, Porter T, Gilani I, Noordin S, Li QX, Collins S, Martins RN, Maruff P, Masters CL, Rowe CC, Villemagne VL, Dore V, Newsholme P, Laws SM, Verdile G. Insulin resistance, cognition and Alzheimer's disease biomarkers: Evidence that CSF Aβ42 moderates the association between insulin resistance and increased CSF tau levels. Neurobiol Aging 2022; 114:38-48. [DOI: 10.1016/j.neurobiolaging.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/09/2022] [Accepted: 03/07/2022] [Indexed: 12/16/2022]
|
36
|
Vacher M, Porter T, Milicic L, Bourgeat P, Dore V, Villemagne VL, Laws SM, Doecke JD. A Targeted Association Study of Blood-Brain Barrier Gene SNPs and Brain Atrophy. J Alzheimers Dis 2022; 86:1817-1829. [DOI: 10.3233/jad-210644] [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: The blood-brain barrier (BBB) is formed by a high-density lining of endothelial cells, providing a border between circulating blood and the brain interstitial fluid. This structure plays a key role in protecting the brain microenvironment by restricting passage of certain molecules and circulating pathogens. Objective: To identify associations between brain volumetric changes and a set of 355 BBB-related single nucleotide polymorphisms (SNP). Method: In a population of 721 unrelated individuals, linear mixed effect models were used to assess if specific variants were linked to regional rates of atrophy over a 12-year time span. Four brain regions were investigated, including cortical grey matter, cortical white matter, ventricle, and hippocampus. Further, we also investigated the potential impact of history of hypertension, diabetes, and the incidence of stroke on regional brain volume change. Results: History of hypertension, diabetes, and stroke was not associated with longitudinal brain volume change. However, we identified a series of genetic variants associated with regional brain volume changes. The associations were independent of variation due to the APOEɛ4 allele and were significant post correction for multiple comparisons. Conclusion: This study suggests that key genes involved in the regulation of BBB integrity may be associated with longitudinal changes in specific brain regions. The derived polygenic risk scores indicate that these interactions are multigenic. Further research needs to be conducted to investigate how BBB functions maybe compromised by genetic variation.
Collapse
Affiliation(s)
- Michael Vacher
- CSIRO Health and Biosecurity, Australian e-Health Research Centre, Floreat, Western Australia, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia
| | - Lidija Milicic
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia
| | - Pierrick Bourgeat
- CSIRO Health and Biosecurity, Australian e-Health Research Centre, Herston, Queensland, Australia
| | - Vincent Dore
- CSIRO Health and Biosecurity, Australian e-Health Research Centre, Herston, Queensland, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simon M. Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia
| | - James D. Doecke
- CSIRO Health and Biosecurity, Australian e-Health Research Centre, Herston, Queensland, Australia
| |
Collapse
|
37
|
Chronic hypoperfusion due to intracranial large artery stenosis is not associated with cerebral β-amyloid deposition and brain atrophy. Chin Med J (Engl) 2022; 135:591-597. [PMID: 34985014 PMCID: PMC8920433 DOI: 10.1097/cm9.0000000000001918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: Insufficient cerebral perfusion is suggested to play a role in the development of Alzheimer disease (AD). However, there is a lack of direct evidence indicating whether hypoperfusion causes or aggravates AD pathology. We investigated the effect of chronic cerebral hypoperfusion on AD-related pathology in humans. Methods: We enrolled a group of cognitively normal patients (median age: 64 years) with unilateral chronic cerebral hypoperfusion. Regions of interest with the most pronounced hypoperfusion changes were chosen in the hypoperfused region and were then mirrored in the contralateral hemisphere to create a control region with normal perfusion. 11C-Pittsburgh compound-positron emission tomography standard uptake ratios and brain atrophy indices were calculated from the computed tomography images of each patient. Results: The median age of the 10 participants, consisting of 4 males and 6 females, was 64 years (47–76 years). We found that there were no differences in standard uptake ratios of the cortex (volume of interest [VOI]: P = 0.721, region of interest [ROI]: P = 0.241) and grey/white ratio (VOI: P = 0.333, ROI: P = 0.445) and brain atrophy indices (Bicaudate, Bifrontal, Evans, Cella, Cella media, and Ventricular index, P > 0.05) between the hypoperfused regions and contralateral normally perfused regions in patients with unilateral chronic cerebral hypoperfusion. Conclusion: Our findings suggest that chronic hypoperfusion due to large vessel stenosis may not directly induce cerebral β-amyloid deposition and neurodegeneration in humans.
Collapse
|
38
|
Sarant JZ, Harris DC, Busby PA, Fowler C, Fripp J, Masters CL, Maruff P. No Influence of Age-Related Hearing Loss on Brain Amyloid-β. J Alzheimers Dis 2022; 85:359-367. [PMID: 34806606 PMCID: PMC8842788 DOI: 10.3233/jad-215121] [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] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Hearing loss is independently associated with a faster rate of cognitive decline in older adults and has been identified as a modifiable risk factor for dementia. The mechanism for this association is unknown, and there has been limited exploration of potential casual pathology. OBJECTIVE Our objective was to investigate whether there was an association between degree of audiometrically measured hearing loss (HL) and brain amyloid-β (Aβ) in a pre-clinical sample. METHODS Participants of the Australian Imaging and Biomarker Longitudinal Study (AIBL; n = 143) underwent positron emission tomography (PET) imaging and objective measurement of hearing thresholds within 5 years of imaging, as well as cognitive assessment within 2 years of imaging in this observational cohort study. RESULTS With one exception, study participants who had cognitive assessments within 2 years of their PET imaging (n = 113) were classified as having normal cognition. There was no association between cognitive scores and degree of hearing loss, or between cognitive scores and Aβ load. No association between HL and Aβ load was found once age was controlled for. As previously reported, positive Apolipoprotein E4 (APOE4) carrier status increased the risk of being Aβ positive (p = 0.002). CONCLUSION Degree of HL was not associated with positive Aβ status.
Collapse
Affiliation(s)
| | | | | | | | - Jurgen Fripp
- Commonwealth Scientific and Industrial Research Organization, Queensland, Australia
| | | | | |
Collapse
|
39
|
Matsuda H, Yamao T, Shakado M, Shigemoto Y, Okita K, Sato N. Amyloid PET quantification using low-dose CT-guided anatomic standardization. EJNMMI Res 2021; 11:125. [PMID: 34905145 PMCID: PMC8671596 DOI: 10.1186/s13550-021-00867-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 11/26/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Centiloid (CL) scaling has become a standardized quantitative measure in amyloid PET because it facilitates the direct comparison of results across institutions, even when different analytical methods or tracers are used. Standard volumes of interest must be used to calculate the CL scale after the anatomic standardization of amyloid PET images using coregistered MRI; if the MRI is unavailable, the CL scale cannot be accurately calculated. This study sought to determine the substitutability of low-dose CT, which is used to correct PET attenuation in PET/CT equipment, by evaluating the measurement accuracy when low-dose CT is used as an alternative to MRI in the calculation of the CL scale. Amyloid PET images obtained using 18F-flutemetamol from 24 patients with possible or probable Alzheimer's disease were processed to calculate the CL scale using 3D T1-weighted MRI and low-dose CT of PET/CT. CLMRI and CLCT were, respectively, defined as the use of MRI and CT for anatomic standardization and compared. Regional differences in the CT-based and MRI-based standardized anatomic images were also investigated. TRIAL REGISTRATION Japan Registry of Clinical Trials, jRCTs031180321 (registered 18 March 2019, https://jrct.niph.go.jp/latest-detail/jRCTs031180321 ). RESULTS A Bland-Altman plot showed that CLCT was slightly but significantly underestimated (mean ± standard deviation, - 1.7 ± 2.4; p < 0.002) compared with CLMRI. The 95% limits of agreement ranged from - 2.8 to - 0.7. Pearson correlation analysis showed a highly significant correlation of r = 0.998 between CLCT and CLMRI (p < 0.001). The linear regression equation was CLMRI = 1.027 × CLCT + 0.762. In a Bland-Altman plot, Spearman correlation analysis did not identify a significant association between the difference in CLMRI versus CLCT and CL load (ρ = - 0.389, p = 0.060). This slight underestimation of CLCT may derive from slightly higher uptake when the cerebellum is used as a reference area in CT-based anatomically standardized PET images versus MRI-based images. CONCLUSIONS Low-dose CT of PET/CT can substitute for MRI in the anatomic standardization used to calculate the CL scale from amyloid PET, although a slight underestimation occurs.
Collapse
Affiliation(s)
- Hiroshi Matsuda
- Department of Biofunctional Imaging, Fukushima Medical University, 1 Hikariga-oka, Fukushima City, Fukushima, 960-1295, Japan. .,Drug Discovery and Cyclotron Research Center, Southern Tohoku Research Institute for Neuroscience, 7-61-2 Yatsuyamada, Koriyama, Fukushima, 963-8052, Japan. .,Department of Radiology, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8551, Japan. .,Department of Biofunctional Imaging, Fukushima Medical University, 6F(621), Shin-Otemachi Building, 2-2-1, Otemachi, Chiyoda-ku, Tokyo, 100-0004, Japan.
| | - Tensho Yamao
- Drug Discovery and Cyclotron Research Center, Southern Tohoku Research Institute for Neuroscience, 7-61-2 Yatsuyamada, Koriyama, Fukushima, 963-8052, Japan.,Department of Radiological Sciences, School of Health Sciences, Fukushima Medical University, 10-6, Sakae, Fukushima, 960-8516, Japan
| | - Mitsuru Shakado
- Department of Radiology, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8551, Japan
| | - Yoko Shigemoto
- Drug Discovery and Cyclotron Research Center, Southern Tohoku Research Institute for Neuroscience, 7-61-2 Yatsuyamada, Koriyama, Fukushima, 963-8052, Japan.,Department of Radiology, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8551, Japan
| | - Kyoji Okita
- Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8551, Japan
| | - Noriko Sato
- Department of Radiology, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8551, Japan
| |
Collapse
|
40
|
Iaccarino L, La Joie R, Koeppe R, Siegel BA, Hillner BE, Gatsonis C, Whitmer RA, Carrillo MC, Apgar C, Camacho MR, Nosheny R, Rabinovici GD. rPOP: Robust PET-only processing of community acquired heterogeneous amyloid-PET data. Neuroimage 2021; 246:118775. [PMID: 34890793 DOI: 10.1016/j.neuroimage.2021.118775] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/12/2021] [Accepted: 11/30/2021] [Indexed: 11/17/2022] Open
Abstract
The reference standard for amyloid-PET quantification requires structural MRI (sMRI) for preprocessing in both multi-site research studies and clinical trials. Here we describe rPOP (robust PET-Only Processing), a MATLAB-based MRI-free pipeline implementing non-linear warping and differential smoothing of amyloid-PET scans performed with any of the FDA-approved radiotracers (18F-florbetapir/FBP, 18F-florbetaben/FBB or 18F-flutemetamol/FLUTE). Each image undergoes spatial normalization based on weighted PET templates and data-driven differential smoothing, then allowing users to perform their quantification of choice. Prior to normalization, users can choose whether to automatically reset the origin of the image to the center of mass or proceed with the pipeline with the image as it is. We validate rPOP with n = 740 (514 FBP, 182 FBB, 44 FLUTE) amyloid-PET scans from the Imaging Dementia-Evidence for Amyloid Scanning - Brain Health Registry sub-study (IDEAS-BHR) and n = 1,518 scans from the Alzheimer's Disease Neuroimaging Initiative (n = 1,249 FBP, n = 269 FBB), including heterogeneous acquisition and reconstruction protocols. After running rPOP, a standard quantification to extract Standardized Uptake Value ratios and the respective Centiloids conversion was performed. rPOP-based amyloid status (using an independent pathology-based threshold of ≥24.4 Centiloid units) was compared with either local visual reads (IDEAS-BHR, n = 663 with complete valid data and reads available) or with amyloid status derived from an MRI-based PET processing pipeline (ADNI, thresholds of >20/>18 Centiloids for FBP/FBB). Finally, within the ADNI dataset, we tested the linear associations between rPOP- and MRI-based Centiloid values. rPOP achieved accurate warping for N = 2,233/2,258 (98.9%) in the first pass. Of the N = 25 warping failures, 24 were rescued with manual reorientation and origin reset prior to warping. We observed high concordance between rPOP-based amyloid status and both visual reads (IDEAS-BHR, Cohen's k = 0.72 [0.7-0.74], ∼86% concordance) or MRI-pipeline based amyloid status (ADNI, k = 0.88 [0.87-0.89], ∼94% concordance). rPOP- and MRI-pipeline based Centiloids were strongly linearly related (R2:0.95, p<0.001), with this association being significantly modulated by estimated PET resolution (β= -0.016, p<0.001). rPOP provides reliable MRI-free amyloid-PET warping and quantification, leveraging widely available software and only requiring an attenuation-corrected amyloid-PET image as input. The rPOP pipeline enables the comparison and merging of heterogeneous datasets and is publicly available at https://github.com/leoiacca/rPOP.
Collapse
Affiliation(s)
- Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Robert Koeppe
- Department of Radiology, University of Michigan, Ann Arbor, MI, United States
| | - Barry A Siegel
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, MO, United States
| | - Bruce E Hillner
- Department of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Constantine Gatsonis
- Center for Statistical Sciences, Brown University School of Public Health, Providence, RI, United States; Department of Biostatistics, Brown University School of Public Health, Providence, RI, United States
| | - Rachel A Whitmer
- Division of Research, Kaiser Permanente, Oakland, CA, United States; Department of Public Health Sciences, University of California Davis, Davis, CA, United States
| | - Maria C Carrillo
- Medical and Scientific Relations Division, Alzheimer's Association, Chicago, IL, United States
| | - Charles Apgar
- American College of Radiology, Reston, VA, United States
| | - Monica R Camacho
- San Francisco VA Medical Center, San Francisco, CA, United States; Northern California Institute for Research and Education (NCIRE), San Francisco, CA, United States
| | - Rachel Nosheny
- San Francisco VA Medical Center, San Francisco, CA, United States; Department of Psychiatry, University of California San Francisco, San Francisco, CA, United States
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States; Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States.
| | | |
Collapse
|
41
|
Xia Y, Yassi N, Raniga P, Bourgeat P, Desmond P, Doecke J, Ames D, Laws SM, Fowler C, Rainey-Smith SR, Martins R, Maruff P, Villemagne VL, Masters CL, Rowe CC, Fripp J, Salvado O. Comorbidity of Cerebrovascular and Alzheimer's Disease in Aging. J Alzheimers Dis 2021; 78:321-334. [PMID: 32986666 DOI: 10.3233/jad-200419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Cerebrovascular disease often coexists with Alzheimer's disease (AD). While both diseases share common risk factors, their interrelationship remains unclear. Increasing the understanding of how cerebrovascular changes interact with AD is essential to develop therapeutic strategies and refine biomarkers for early diagnosis. OBJECTIVE We investigate the prevalence and risk factors for the comorbidity of amyloid-β (Aβ) and cerebrovascular disease in the Australian Imaging, Biomarkers and Lifestyle Study of Ageing, and further examine their cross-sectional association. METHODS A total of 598 participants (422 cognitively normal, 89 with mild cognitive impairment, 87 with AD) underwent positron emission tomography and structural magnetic resonance imaging for assessment of Aβ deposition and cerebrovascular disease. Individuals were categorized based on the comorbidity status of Aβ and cerebrovascular disease (V) as Aβ-V-, Aβ-V+, Aβ+V-, or Aβ+V+. RESULTS Advancing age was associated with greater likelihood of cerebrovascular disease, high Aβ load and their comorbidity. Apolipoprotein E ɛ4 carriage was only associated with Aβ positivity. Greater total and regional WMH burden were observed in participants with AD. However, no association were observed between Aβ and WMH measures after stratification by clinical classification, suggesting that the observed association between AD and cerebrovascular disease was driven by the common risk factor of age. CONCLUSION Our observations demonstrate common comorbid condition of Aβ and cerebrovascular disease in later life. While our study did not demonstrate a convincing cross-sectional association between Aβ and WMH burden, future longitudinal studies are required to further confirm this.
Collapse
Affiliation(s)
- Ying Xia
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| | - Nawaf Yassi
- Department of Medicine and Neurology, Melbourne Brain Centre at The Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.,Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Parnesh Raniga
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| | - Pierrick Bourgeat
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| | - Patricia Desmond
- Department of Radiology, The Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - James Doecke
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| | - David Ames
- National Ageing Research Institute, Parkville, VIC, Australia.,Academic Unit for Psychiatry of Old Age, University of Melbourne, Parkville, VIC, Australia
| | - Simon M Laws
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Perth, WA, Australia
| | - Christopher Fowler
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Stephanie R Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Perth, WA, Australia
| | - Ralph Martins
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Perth, WA, Australia
| | - Paul Maruff
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Cog State Ltd, Melbourne, VIC, Australia
| | - Victor L Villemagne
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia.,Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Christopher C Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia.,Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia
| | - Jurgen Fripp
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| | - Olivier Salvado
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia.,CSIRO Data61, Brisbane, QLD, Australia
| | | |
Collapse
|
42
|
Chatterjee P, Pedrini S, Ashton NJ, Tegg M, Goozee K, Singh AK, Karikari TK, Simrén J, Vanmechelen E, Armstrong NJ, Hone E, Asih PR, Taddei K, Doré V, Villemagne VL, Sohrabi HR, Zetterberg H, Masters CL, Blennow K, Martins RN. Diagnostic and prognostic plasma biomarkers for preclinical Alzheimer's disease. Alzheimers Dement 2021; 18:1141-1154. [PMID: 34494715 DOI: 10.1002/alz.12447] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/03/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION This study involved a parallel comparison of the diagnostic and longitudinal monitoring potential of plasma glial fibrillary acidic protein (GFAP), total tau (t-tau), phosphorylated tau (p-tau181 and p-tau231), and neurofilament light (NFL) in preclinical Alzheimer's disease (AD). METHODS Plasma proteins were measured using Simoa assays in cognitively unimpaired older adults (CU), with either absence (Aβ-) or presence (Aβ+) of brain amyloidosis. RESULTS Plasma GFAP, t-tau, p-tau181, and p-tau231 concentrations were higher in Aβ+ CU compared with Aβ- CU cross-sectionally. GFAP had the highest effect size and area under the curve (AUC) in differentiating between Aβ+ and Aβ- CU; however, no statistically significant differences were observed between the AUCs of GFAP, p-tau181, and p-tau231, but all were significantly higher than the AUC of NFL, and the AUC of GFAP was higher than the AUC of t-tau. The combination of a base model (BM), comprising the AD risk factors, age, sex, and apolipoprotein E gene (APOE) ε4 status with GFAP was observed to have a higher AUC (>90%) compared with the combination of BM with any of the other proteins investigated in the current study. Longitudinal analyses showed increased GFAP and p-tau181 in Aβ+ CU and increased NFL in Aβ- CU, over a 12-month duration. GFAP, p-tau181, p-tau231, and NFL showed significant correlations with cognition, whereas no significant correlations were observed with hippocampal volume. DISCUSSION These findings highlight the diagnostic and longitudinal monitoring potential of GFAP and p-tau for preclinical AD.
Collapse
Affiliation(s)
- Pratishtha Chatterjee
- Department of Biomedical Sciences, Macquarie University, North Ryde, New South Wales, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Steve Pedrini
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Michelle Tegg
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Kathryn Goozee
- Department of Biomedical Sciences, Macquarie University, North Ryde, New South Wales, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia.,The Cooperative Research Centre for Mental Health, Carlton South, Australia.,KaRa Institute of Neurological Disease, Macquarie Park, Australia
| | - Abhay K Singh
- Macquarie Business School, Macquarie University, North Ryde, New South Wales, Australia
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Joel Simrén
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Nicola J Armstrong
- Department of Mathematics & Statistics, Curtin University, Bentley, Western Australia, Australia
| | - Eugene Hone
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Prita R Asih
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Kevin Taddei
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Nedlands, Western Australia, Australia
| | - Vincent Doré
- eHealth, CSIRO Health and Biosecurity, Herston, Queensland, Australia.,Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Victor L Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia.,Department of Psychiatry, University of Pittsburgh, Pennsylvania, USA
| | - Hamid R Sohrabi
- Department of Biomedical Sciences, Macquarie University, North Ryde, New South Wales, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Nedlands, Western Australia, Australia.,Centre for Healthy Ageing, Health Future Institute, Murdoch University, Murdoch, Western Australia, Australia
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ralph N Martins
- Department of Biomedical Sciences, Macquarie University, North Ryde, New South Wales, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia.,The Cooperative Research Centre for Mental Health, Carlton South, Australia.,KaRa Institute of Neurological Disease, Macquarie Park, Australia.,Australian Alzheimer's Research Foundation, Nedlands, Western Australia, Australia
| |
Collapse
|
43
|
Pegueroles J, Montal V, Bejanin A, Vilaplana E, Aranha M, Santos‐Santos MA, Alcolea D, Carrió I, Camacho V, Blesa R, Lleó A, Fortea J. AMYQ: An index to standardize quantitative amyloid load across PET tracers. Alzheimers Dement 2021; 17:1499-1508. [PMID: 33797846 PMCID: PMC8519100 DOI: 10.1002/alz.12317] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/21/2021] [Accepted: 01/31/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Positron emission tomography (PET) amyloid quantification methods require magnetic resonance imaging (MRI) for spatial registration and a priori reference region to scale the images. Furthermore, different tracers have distinct thresholds for positivity. We propose the AMYQ index, a new measure of amyloid burden, to overcome these limitations. METHODS We selected 18F-amyloid scans from ADNI and Australian Imaging, Biomarker & Lifestyle Flagship Study of Ageing (AIBL) with the corresponding T1-MRI. A subset also had neuropathological data. PET images were normalized, and the AMYQ was calculated based on an adaptive template. We compared AMYQ with the Centiloid scale on clinical and neuropathological diagnostic performance. RESULTS AMYQ was related with amyloid neuropathological burden and had excellent diagnostic performance to discriminate controls from patients with Alzheimer's disease (AD) (area under the curve [AUC] = 0.86). AMYQ had a high agreement with the Centiloid scale (intraclass correlation coefficient [ICC] = 0.88) and AUC between 0.94 and 0.99 to discriminate PET positivity when using different Centiloid cutoffs. DISCUSSION AMYQ is a new MRI-independent index for standardizing and quantifying amyloid load across tracers.
Collapse
Affiliation(s)
- Jordi Pegueroles
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Victor Montal
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Eduard Vilaplana
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Mateus Aranha
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Miguel Angel Santos‐Santos
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Ignasi Carrió
- Department of Nuclear MedicineHospital de la Santa Creu i Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Valle Camacho
- Department of Nuclear MedicineHospital de la Santa Creu i Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | | | | |
Collapse
|
44
|
Bischof GN, Bartenstein P, Barthel H, van Berckel B, Doré V, van Eimeren T, Foster N, Hammes J, Lammertsma AA, Minoshima S, Rowe C, Sabri O, Seibyl J, Van Laere K, Vandenberghe R, Villemagne V, Yakushev I, Drzezga A. Toward a Universal Readout for 18F-Labeled Amyloid Tracers: The CAPTAINs Study. J Nucl Med 2021; 62:999-1005. [PMID: 33712532 DOI: 10.2967/jnumed.120.250290] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/09/2020] [Accepted: 10/21/2020] [Indexed: 11/16/2022] Open
Abstract
To date, 3 18F-labeled PET tracers have been approved for assessing cerebral amyloid plaque pathology in the diagnostic workup of suspected Alzheimer disease (AD). Although scanning protocols are relatively similar across tracers, U.S. Food and Drug Administration- and the European Medicines Agency-approved visual rating protocols differ among the 3 tracers. This proof-of-concept study assessed the comparability of the 3 approved visual rating protocols to classify a scan as amyloid-positive or -negative, when applied by groups of experts and nonexperts to all 3 amyloid tracers. Methods: In an international multicenter approach, both expert (n = 4) and nonexpert raters (n = 3) rated scans acquired with 18F-florbetaben, 18F-florbetapir and 18F-flutemetamol. Scans obtained with each tracer were presented for reading according to all 3 approved visual rating protocols. In a randomized order, every single scan was rated by each reader according to all 3 protocols. Raters were blinded for the amyloid tracer used and asked to rate each scan as positive or negative, giving a confidence judgment after each response. Percentage of visual reader agreement, interrater reliability, and agreement of each visual read with binary quantitative measures (fixed SUV ratio threshold for positive or negative scans) were computed. These metrics were analyzed separately for expert and nonexpert groups. Results: No significant differences in using the different approved visual rating protocols were observed across the different metrics of agreement in the group of experts. Nominal differences suggested that the 18F-florbetaben visual rating protocol achieved the highest interrater reliability and accuracy especially under low confidence conditions. For the group of nonexpert raters, significant differences between the different visual rating protocols were observed with overall moderate-to-fair accuracy and with the highest reliability for the 18F-florbetapir visual rating protocol. Conclusion: We observed high interrater agreement despite applying different visual rating protocols for all 18F-labeled amyloid tracers. This implies that the results of the visual interpretation of amyloid imaging can be well standardized and do not depend on the rating protocol in experts. Consequently, the creation of a universal visual assessment protocol for all amyloid imaging tracers appears feasible, which could benefit especially the less-experienced readers.
Collapse
Affiliation(s)
- Gérard N Bischof
- University Hospital Cologne, Multimodal Neuroimaging Group, Department of Nuclear Medicine, Cologne, Germany;
| | | | - Henryk Barthel
- University Hospital of Leipzig, Department of Nuclear Medicine, Leipzig, Germany
| | - Bart van Berckel
- Amsterdam University Medical Centers, Location VUmc Radiology and Nuclear Medicine, Amsterdam, The Netherlands
| | - Vincent Doré
- CSIRO Health and Biosecurity, Parkville 3052, Victoria, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia
| | - Thilo van Eimeren
- University Hospital Cologne, Multimodal Neuroimaging Group, Department of Nuclear Medicine, Cologne, Germany
- Department of Neurology, University Hospital Cologne, Cologne, Germany
- German Center of Neurodegenerative Disease (DZNE), Bonn, Germany
| | - Norman Foster
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| | - Jochen Hammes
- University Hospital Cologne, Multimodal Neuroimaging Group, Department of Nuclear Medicine, Cologne, Germany
| | - Adriaan A Lammertsma
- Amsterdam University Medical Centers, Location VUmc Radiology and Nuclear Medicine, Amsterdam, The Netherlands
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| | - Chris Rowe
- CSIRO Health and Biosecurity, Parkville 3052, Victoria, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia
| | - Osama Sabri
- University Hospital of Leipzig, Department of Nuclear Medicine, Leipzig, Germany
| | - John Seibyl
- Institute for Neurodegenerative Disorders, New Haven, Connecticut
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, University Hospital Leuven and Department of Imaging and Pathology KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Memory Clinic, University Hospital Leuven and Department of Neurosciences, KU Leuven, Belgium
| | - Victor Villemagne
- CSIRO Health and Biosecurity, Parkville 3052, Victoria, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Melbourne, Australia
| | - Igor Yakushev
- Department of Nuclear Medicine, Technical University of Munich, Germany; and
| | - Alexander Drzezga
- University Hospital Cologne, Multimodal Neuroimaging Group, Department of Nuclear Medicine, Cologne, Germany
- German Center of Neurodegenerative Disease (DZNE), Bonn, Germany
- Institute of Neuroscience and Medicine (INM-2), Molecular Organization of the Brain, Forschungszentrum Jülich, Germany
| |
Collapse
|
45
|
Gardener SL, Weinborn M, Sohrabi HR, Doecke JD, Bourgeat P, Rainey-Smith SR, Shen KK, Fripp J, Taddei K, Maruff P, Salvado O, Savage G, Ames D, Masters CL, Rowe CC, Martins RN. Longitudinal Trajectories in Cortical Thickness and Volume Atrophy: Superior Cognitive Performance Does Not Protect Against Brain Atrophy in Older Adults. J Alzheimers Dis 2021; 81:1039-1052. [PMID: 33935071 PMCID: PMC8293653 DOI: 10.3233/jad-201243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Previous research has identified a small subgroup of older adults that maintain a high level of cognitive functioning well into advanced age. Investigation of those with superior cognitive performance (SCP) for their age is important, as age-related decline has previously been thought to be inevitable. Objective: Preservation of cortical thickness and volume was evaluated in 76 older adults with SCP and 100 typical older adults (TOAs) assessed up to five times over six years. Methods: Regions of interest (ROIs) found to have been associated with super-aging status (a construct similar to SCP status) in previous literature were investigated, followed by a discovery phase analyses of additional regions. SCPs were aged 70 + at baseline, scoring at/above normative memory (CVLT-II) levels for demographically similar individuals aged 30–44 years old, and in the unimpaired range for all other cognitive domains over the course of the study. Results: In linear mixed models, following adjustment for multiple comparisons, there were no significant differences between rates of thinning or volume atrophy between SCPs and TOAs in previously identified ROIs, or the discovery phase analyses. With only amyloid-β negative individuals in the analyses, again there were no significant differences between SCPs and TOAs. Conclusion: The increased methodological rigor in classifying groups, together with the influence of cognitive reserve, are discussed as potential factors accounting for our findings as compared to the extant literature on those with superior cognitive performance for their age.
Collapse
Affiliation(s)
- Samantha L Gardener
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia
| | - Michael Weinborn
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia.,School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia
| | - Hamid R Sohrabi
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia.,College of Science, Health, Engineering and Education, Murdoch University, Murdoch, Western Australia, Australia.,Department of Biomedical Sciences, Macquarie University, New South Wales, Australia
| | - James D Doecke
- CSIRO Health and Biosecurity/Australian eHealth Research Centre, Herston, Queensland, Australia
| | - Pierrick Bourgeat
- CSIRO Health and Biosecurity/Australian eHealth Research Centre, Herston, Queensland, Australia
| | - Stephanie R Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia.,School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia.,Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia
| | - Kai-Kai Shen
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia.,CSIRO Health and Biosecurity/Australian eHealth Research Centre, Herston, Queensland, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity/Australian eHealth Research Centre, Herston, Queensland, Australia
| | - Kevin Taddei
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia
| | - Paul Maruff
- CogState, Ltd., Melbourne, Victoria, Australia
| | - Olivier Salvado
- CSIRO Health and Biosecurity/Australian eHealth Research Centre, Herston, Queensland, Australia.,CSIRO Data61, Sydney, Australia
| | - Greg Savage
- ARC Centre of Excellence in Cognition and its Disorders and Department of Psychology, Macquarie University, New South Wales, Australia
| | - David Ames
- National Ageing Research Institute, Royal Melbourne Hospital, Melbourne, Australia.,Academic Unit for Psychiatry of Old Age, University of Melbourne, Melbourne, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Centre for PET, Austin Health, Heidelberg, Victoria, Australia.,Florey Department of the University of Melbourne
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia.,Department of Biomedical Sciences, Macquarie University, New South Wales, Australia
| | | |
Collapse
|
46
|
Moussavi Nik SH, Porter T, Newman M, Bartlett B, Khan I, Sabale M, Eccles M, Woodfield A, Groth D, Dore V, Villemagne VL, Masters CL, Martins RN, Laws SM, Lardelli M, Verdile G. Relevance of a Truncated PRESENILIN 2 Transcript to Alzheimer's Disease and Neurodegeneration. J Alzheimers Dis 2021; 80:1479-1489. [PMID: 33720885 DOI: 10.3233/jad-201133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The PRESENILIN genes (PSEN1, PSEN2) encoding for their respective proteins have critical roles in many aspects of Alzheimer's disease (AD) pathogenesis. The PS2V transcript of PSEN2 encodes a truncated protein and is upregulated in AD brains; however, its relevance to AD and disease progression remains to be determined. OBJECTIVE Assess transcript levels in postmortem AD and non-AD brain tissue and in lymphocytes collected under the Australian Imaging Biomarker and Lifestyle (AIBL) study. METHODS Full length PSEN2 and PS2V transcript levels were assessed by quantitative digital PCR in postmortem brain tissue (frontal cortex and hippocampus) from control, AD, frontotemporal dementia (FTD), and Lewy body dementia (LBD). Transcript levels were also assessed in lymphocytes obtained from the Perth subset of the AIBL study (n = 160). Linear regression analysis was used to assess correlations between transcript copy number and brain volume and neocortical amyloid load. RESULTS PS2V levels increased in AD postmortem brain but PS2V was also present at significant levels in FTD and LBD brains. PS2V transcript was detected in lymphocytes and PS2V/PSEN2 ratios were increased in mild cognitive impairment (p = 0.024) and AD (p = 0.019) groups compared to control group. Increased ratios were significantly correlated with hippocampal volumes only (n = 62, β= -0.269, p = 0.03). CONCLUSION Taken together, these results suggest that PS2V may be a marker of overall neurodegeneration.
Collapse
Affiliation(s)
- Seyyed Hani Moussavi Nik
- University of Adelaide, School of Biological Sciences, Centre for Molecular Pathology, Adelaide, SA, Australia
| | - Tenielle Porter
- Collaborative Genomics and Translation Group, Strategic Research Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Morgan Newman
- University of Adelaide, School of Biological Sciences, Centre for Molecular Pathology, Adelaide, SA, Australia
| | - Benjamin Bartlett
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia.,Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia.,School of Medicine, University of Western Australia, Crawley, Western Australia, Australia
| | - Imran Khan
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Miheer Sabale
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia.,Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, New South Wales, Australia
| | - Melissa Eccles
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Amy Woodfield
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - David Groth
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Vincent Dore
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia
| | - Victor L Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ralph N Martins
- Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, New South Wales, Australia.,School of Medical and Health Sciences, Edith Cowan University, Western Australia, Australia
| | - Simon M Laws
- Collaborative Genomics and Translation Group, Strategic Research Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Michael Lardelli
- University of Adelaide, School of Biological Sciences, Centre for Molecular Pathology, Adelaide, SA, Australia
| | - Giuseppe Verdile
- Collaborative Genomics and Translation Group, Strategic Research Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| |
Collapse
|
47
|
Bucci M, Savitcheva I, Farrar G, Salvadó G, Collij L, Doré V, Gispert JD, Gunn R, Hanseeuw B, Hansson O, Shekari M, Lhommel R, Molinuevo JL, Rowe C, Sur C, Whittington A, Buckley C, Nordberg A. A multisite analysis of the concordance between visual image interpretation and quantitative analysis of [ 18F]flutemetamol amyloid PET images. Eur J Nucl Med Mol Imaging 2021; 48:2183-2199. [PMID: 33844055 PMCID: PMC8175298 DOI: 10.1007/s00259-021-05311-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 03/09/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND [18F]flutemetamol PET scanning provides information on brain amyloid load and has been approved for routine clinical use based upon visual interpretation as either negative (equating to none or sparse amyloid plaques) or amyloid positive (equating to moderate or frequent plaques). Quantitation is however fundamental to the practice of nuclear medicine and hence can be used to supplement amyloid reading methodology especially in unclear cases. METHODS A total of 2770 [18F]flutemetamol images were collected from 3 clinical studies and 6 research cohorts with available visual reading of [18F]flutemetamol and quantitative analysis of images. These were assessed further to examine both the discordance and concordance between visual and quantitative imaging primarily using thresholds robustly established using pathology as the standard of truth. Scans covered a wide range of cases (i.e. from cognitively unimpaired subjects to patients attending the memory clinics). Methods of quantifying amyloid ranged from using CE/510K cleared marked software (e.g. CortexID, Brass), to other research-based methods (e.g. PMOD, CapAIBL). Additionally, the clinical follow-up of two types of discordance between visual and quantitation (V+Q- and V-Q+) was examined with competing risk regression analysis to assess possible differences in prediction for progression to Alzheimer's disease (AD) and other diagnoses (OD). RESULTS Weighted mean concordance between visual and quantitation using the autopsy-derived threshold was 94% using pons as the reference region. Concordance from a sensitivity analysis which assessed the maximum agreement for each cohort using a range of cut-off values was also estimated at approximately 96% (weighted mean). Agreement was generally higher in clinical cases compared to research cases. V-Q+ discordant cases were 11% more likely to progress to AD than V+Q- for the SUVr with pons as reference region. CONCLUSIONS Quantitation of amyloid PET shows a high agreement vs binary visual reading and also allows for a continuous measure that, in conjunction with possible discordant analysis, could be used in the future to identify possible earlier pathological deposition as well as monitor disease progression and treatment effectiveness.
Collapse
Affiliation(s)
- Marco Bucci
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Irina Savitcheva
- Medical Radiation Physics and Nuclear Medicine, Section for Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Gill Farrar
- Pharmaceutical Diagnostics, GE Healthcare, Amersham, UK
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Lyduine Collij
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, 1117, Amsterdam, Netherlands
| | - Vincent Doré
- Austin Health, University of Melbourne, Melbourne, Australia.,Health and Biosecurity, CSIRO, Parkville, Australia
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain.,Centro de Investigación Biomédica en Red Bioingenieriá, Biomateriales y Nanomedicina, (CIBER-BBN), Barcelona, Spain
| | - Roger Gunn
- Invicro, London, UK.,Division of Brain Sciences, Department of Medicine, Imperial College, London, UK
| | - Bernard Hanseeuw
- Neurology and Nuclear Medicine Departments, Saint-Luc University Hospital, Av. Hippocrate, 10, 1200, Brussels, Belgium.,Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmo, Lund University, Lund, Sweden
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Renaud Lhommel
- Neurology and Nuclear Medicine Departments, Saint-Luc University Hospital, Av. Hippocrate, 10, 1200, Brussels, Belgium
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Christopher Rowe
- Austin Health, University of Melbourne, Melbourne, Australia.,Department of Medicine, The University of Melbourne, Melbourne, Australia
| | | | | | | | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden. .,Department of Aging, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
48
|
Tsubaki Y, Kitamura T, Shimokawa N, Akamatsu G, Sasaki M. Improved Accuracy of Amyloid PET Quantification with Adaptive Template-Based Anatomic Standardization. J Nucl Med Technol 2021; 49:256-261. [PMID: 33820861 DOI: 10.2967/jnmt.120.261701] [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: 12/12/2020] [Accepted: 03/01/2021] [Indexed: 11/16/2022] Open
Abstract
Amyloid PET noninvasively visualizes amyloid-β accumulation in the brain. Visual binary reading is the standard method for interpreting amyloid PET, whereas objective quantitative evaluation is required in research and clinical trials. Anatomic standardization is important for quantitative analysis, and various standard templates are used for this purpose. To address the large differences in radioactivity distribution between amyloid-positive and amyloid-negative participants, an adaptive-template method has been proposed for the anatomic standardization of amyloid PET. In this study, we investigated the difference between the adaptive-template method and the single-template methods (use of a positive or a negative template) in amyloid PET quantitative evaluation, focusing on the accuracy in diagnosing Alzheimer's disease (AD). Methods: In total, 166 participants (58 healthy controls [HCs], 62 patients with mild cognitive impairment [MCI], and 46 patients with AD) who underwent 11C-Pittsburgh compound B (11C-PiB) PET through the Japanese Alzheimer's Disease Neuroimaging Initiative study were examined. For the anatomic standardization of 11C-PiB PET images, we applied 3 methods: a positive-template-based method, a negative-template-based method, and an adaptive-template-based method. The positive template was created by averaging the PET images for 4 patients with AD and 7 patients with MCI. Conversely, the negative template was created by averaging the PET images for 8 HCs. In the adaptive-template-based method, either of the templates was used on the basis of the similarity (normalized cross-correlation [NCC]) between the individual standardized image and the corresponding template. An empiric PiB-prone region of interest was used to evaluate specific regions where amyloid-β accumulates. The reference region was the cerebellar cortex, and the evaluated regions were the posterior cingulate gyrus and precuneus and the frontal, lateral temporal, lateral parietal, and occipital lobes. The mean cortical SUV ratio (mcSUVR) was calculated for quantitative evaluation. Results: The NCCs of single-template-based methods (the positive template or negative template) showed a significant difference among the HC, MCI, and AD groups (P < 0.05), whereas the NCC of the adaptive-template-based method did not (P > 0.05). The mcSUVR exhibited significant differences among the HC, MCI, and AD groups with all methods (P < 0.05). The mcSUVR area under the curve by receiver operating characteristic analysis between the positive group (MCI and AD) and the HC group did not significantly differ among templates. With regard to diagnostic accuracy based on mcSUVR, the sensitivity of the negative-template-based and adaptive-template-based methods was superior to that of the positive-template-based method (P < 0.05); however, there was no significant difference in specificity between them. Conclusion: In quantitative evaluation of AD by amyloid PET, the adaptive-template-based anatomic standardization method had greater diagnostic accuracy than the single-template-based methods.
Collapse
Affiliation(s)
- Yuma Tsubaki
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takayoshi Kitamura
- Department of Health Sciences, School of Medicine, Kyushu University, Fukuoka, Japan; and
| | - Natsumi Shimokawa
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Go Akamatsu
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masayuki Sasaki
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan;
| | | |
Collapse
|
49
|
Schwarz CG. Uses of Human MR and PET Imaging in Research of Neurodegenerative Brain Diseases. Neurotherapeutics 2021; 18:661-672. [PMID: 33723751 PMCID: PMC8423895 DOI: 10.1007/s13311-021-01030-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 01/18/2023] Open
Abstract
In the past decades, many neuroimaging studies have aimed to improve the scientific understanding of human neurodegenerative diseases using MRI and PET. This article is designed to provide an overview of the major classes of brain imaging and how/why they are used in this line of research. It is intended as a primer for individuals who are relatively unfamiliar with the methods of neuroimaging research to gain a better understanding of the vocabulary and overall methodologies. It is not intended to describe or review any research findings for any disease or biology, but rather to broadly describe the imaging methodologies that are used in conducting this neurodegeneration research. We will also review challenges and strategies for analyzing neuroimaging data across multiple sites and studies, i.e., harmonization and standardization of imaging data for multi-site and meta-analyses.
Collapse
|
50
|
Fernandez S, Burnham SC, Milicic L, Savage G, Maruff P, Peretti M, Sohrabi HR, Lim YY, Weinborn M, Ames D, Masters CL, Martins RN, Rainey-Smith S, Rowe CC, Salvado O, Groth D, Verdile G, Villemagne VL, Porter T, Laws SM. SPON1 Is Associated with Amyloid-β and APOE ε4-Related Cognitive Decline in Cognitively Normal Adults. J Alzheimers Dis Rep 2021; 5:111-120. [PMID: 33782664 PMCID: PMC7990462 DOI: 10.3233/adr-200246] [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] [Indexed: 11/21/2022] Open
Abstract
Abstract.
Collapse
Affiliation(s)
- Shane Fernandez
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Samantha C Burnham
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,CSIRO Health and Biosecurity, Parkville, Victoria, Australia
| | - Lidija Milicic
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Greg Savage
- ARC Centre of Excellence in Cognition and its Disorders, Department of Psychology, Macquarie University, North Ryde, NSW, Australia
| | - Paul Maruff
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.,CogState Ltd., Melbourne, Victoria, Australia
| | - Madeline Peretti
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Hamid R Sohrabi
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Centre for Healthy Ageing, Murdoch University, Murdoch, Western Australia, Australia.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Yen Ying Lim
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Michael Weinborn
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia
| | - David Ames
- Academic Unit for Psychiatry of Old Age, St. Vincent's Health, The University of Melbourne, Kew, Victoria, Australia.,National Ageing Research Institute, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ralph N Martins
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Stephanie Rainey-Smith
- Australian Alzheimer's Research Foundation, Nedlands, Western Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Christopher C Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia.,Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, Australia
| | - Olivier Salvado
- CSIRO Health and Biosecurity/Australian e-Health Research Centre, Herston, Queensland, Australia
| | - David Groth
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Giuseppe Verdile
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Victor L Villemagne
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Department of Nuclear Medicine and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
| | - Tenielle Porter
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Simon M Laws
- Collaborative Genomics and Translation Group, Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| |
Collapse
|