1
|
Lu K, Baker J, Nicholas JM, Street RE, Keuss SE, Coath W, James SN, Keshavan A, Weston PSJ, Murray-Smith H, Cash DM, Malone IB, Wong A, Fox NC, Richards M, Crutch SJ, Schott JM. Associations between accelerated forgetting, amyloid deposition and brain atrophy in older adults. Brain 2025; 148:1302-1315. [PMID: 39423292 PMCID: PMC11969454 DOI: 10.1093/brain/awae316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 08/14/2024] [Accepted: 09/22/2024] [Indexed: 10/21/2024] Open
Abstract
Accelerated long-term forgetting (ALF) is the phenomenon whereby material is retained normally over short intervals (e.g. minutes) but forgotten abnormally rapidly over longer periods (days or weeks). ALF might be an early marker of cognitive decline, but little is known about its relationships with preclinical Alzheimer's disease pathology and how memory selectivity might influence which material is forgotten. We assessed ALF in 'Insight 46', a sub-study of the MRC National Survey of Health and Development (a population-based cohort born during the same week in 1946) (n = 429; 47% female; assessed at age ∼73 years). ALF assessment comprised visual and verbal memory tests: complex figure drawing and the face-name associative memory exam (FNAME). ALF scores were calculated as the percentage of material retained after 7 days, relative to 30 min. In 306 cognitively normal participants, we investigated effects on ALF of β-amyloid pathology (quantified using 18F-Florbetapir-PET, classified as positive/negative) and whole-brain and hippocampal atrophy rate (quantified from serial T1-MRI over ∼2.4 years preceding the ALF assessment), in addition to interactions between these pathologies. We categorized complex figure drawing items as 'outline' or 'detail', to test our hypothesis that forgetting the outline of the structure would be more sensitive to the effect of brain pathologies. We also investigated associations between ALF and subjective cognitive decline, measured with the MyCog questionnaire. Complex figure 'outline' items were better retained than 'detail' items (mean retention over 7 days = 94% versus 72%). Amyloid-positive participants showed greater forgetting of the complex figure outline compared with amyloid-negative participants (90% versus 95%; P < 0.01). There were interactions between amyloid pathology and cerebral atrophy, such that whole-brain and hippocampal atrophy predicted greater ALF on complex figure drawing among amyloid-positive participants only [e.g. 1.9 percentage-points lower retention per ml/year of whole-brain atrophy (95% confidence intervals 0.5, 3.7); P < 0.05]. Greater ALF on FNAME was associated with increased rate of hippocampal atrophy. ALF on complex figure drawing was also correlated with subjective cognitive decline [-0.45 percentage-points per MyCog point (-0.85, -0.05); P < 0.05]. These results provide evidence of associations between some measures of ALF and biomarkers of brain pathologies and subjective cognitive decline in cognitively normal older adults. On complex figure drawing, 'outline' items were better remembered than 'detail' items, illustrating the strategic role of memory selectivity, but 'outline' items were also relatively more vulnerable to ALF in individuals with amyloid pathology. Overall, our findings suggest that ALF might be a sensitive marker of cognitive changes in preclinical Alzheimer's disease.
Collapse
Affiliation(s)
- Kirsty Lu
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - John Baker
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Jennifer M Nicholas
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Rebecca E Street
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Sarah E Keuss
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - William Coath
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Sarah-Naomi James
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, WC1B 5JU, UK
| | - Ashvini Keshavan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Philip S J Weston
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
| | - Heidi Murray-Smith
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
| | - Ian B Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, WC1B 5JU, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
- UK Dementia Research Institute at UCL, University College London, London, WC1E 6BT, UK
| | - Marcus Richards
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, WC1B 5JU, UK
| | - Sebastian J Crutch
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| | - Jonathan M Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| |
Collapse
|
2
|
Giovane MD, Giunchiglia V, Cai Z, Leoni M, Street R, Lu K, Wong A, Popham M, Nicholas JM, Trender W, Hellyer PJ, Parker TD, Murray‐Smith H, Cash DM, Barnes J, Sudre CH, Malhotra PA, Crutch SJ, Richards M, Hampshire A, Schott JM. Remote cognitive tests predict neurodegenerative biomarkers in the Insight 46 cohort. Alzheimers Dement 2025; 21:e14572. [PMID: 39936232 PMCID: PMC11815243 DOI: 10.1002/alz.14572] [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: 09/27/2024] [Revised: 12/26/2024] [Accepted: 12/28/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND Alzheimer's disease-related biomarkers detect pathology years before symptoms emerge, when disease-modifying therapies might be most beneficial. Remote cognitive testing provides a means of assessing early cognitive changes. We explored the relationship between neurodegenerative biomarkers and cognition in cognitively normal individuals. METHODS We remotely deployed 13 computerized Cognitron tasks in 255 Insight 46 participants. We generated amyloid load and positivity, white matter hyperintensity volume (WMHV), whole brain and hippocampal volumes at age 73, plus rates of change over 2 years. We examined the relationship between Cognitron, biomarkers, and standard neuropsychological tests. RESULTS Slower response time on a delayed recognition task predicted amyloid positivity (odds ratio [OR] = 1.79, confidence interval [CI]: 1.15, 2.95), and WMHV (1.23, CI: 1.00, 1.56). Brain and hippocampal atrophy rates correlated with poorer visuospatial performance (b = -0.42, CI: -0.80, -0.05) and accuracy on immediate recognition (b = -0.01, CI: -0.012, -0.001), respectively. Standard tests correlated with Cognitron composites (rho = 0.50, p < 0.001). DISCUSSION Remote computerized testing correlates with standard supervised assessments and holds potential for studying early cognitive changes associated with neurodegeneration. HIGHLIGHTS 70% of the Online 46 cohort performed a set of remote online cognitive tasks. Response time and accuracy on a memory task predicted amyloid status and load (SUVR). Accuracy on memory and spatial span tasks correlated with longitudinal atrophy rate. The Cognitron tasks correlated with standard supervised cognitive tests. Online cognitive testing can help identify early AD-related memory deficits.
Collapse
Affiliation(s)
- Martina Del Giovane
- Imperial College LondonDepartment of Brain Sciences. Burlington DanesThe Hammersmith HospitalLondonUK
- Imperial College London and The University of SurreyUK Dementia Research Institute Care Research and Technology Centre, Sir Michael Uren HubLondonUK
| | - Valentina Giunchiglia
- Imperial College LondonDepartment of Brain Sciences. Burlington DanesThe Hammersmith HospitalLondonUK
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonDe Crespigny ParkLondonUK
- Department of Biomedical InformaticsHarvard Medical SchoolBostonMassachusettsUSA
| | - Ziyuan Cai
- Imperial College LondonDepartment of Brain Sciences. Burlington DanesThe Hammersmith HospitalLondonUK
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonDe Crespigny ParkLondonUK
| | - Marguerite Leoni
- Imperial College LondonDepartment of Brain Sciences. Burlington DanesThe Hammersmith HospitalLondonUK
| | - Rebecca Street
- Department of Neurodegenerative Disease, The Dementia Research CentreUniversity College London (UCL) Queen Square Institute of NeurologyLondonUK
| | - Kirsty Lu
- Department of Neurodegenerative Disease, The Dementia Research CentreUniversity College London (UCL) Queen Square Institute of NeurologyLondonUK
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | - Maria Popham
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | - Jennifer M. Nicholas
- Department of Neurodegenerative Disease, The Dementia Research CentreUniversity College London (UCL) Queen Square Institute of NeurologyLondonUK
- Department of Medical StatisticsLondon School of Hygiene and Tropical MedicineLondonUK
| | - William Trender
- Imperial College LondonDepartment of Brain Sciences. Burlington DanesThe Hammersmith HospitalLondonUK
| | - Peter J. Hellyer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonDe Crespigny ParkLondonUK
| | - Thomas D. Parker
- Imperial College LondonDepartment of Brain Sciences. Burlington DanesThe Hammersmith HospitalLondonUK
- Imperial College London and The University of SurreyUK Dementia Research Institute Care Research and Technology Centre, Sir Michael Uren HubLondonUK
- Department of Neurodegenerative Disease, The Dementia Research CentreUniversity College London (UCL) Queen Square Institute of NeurologyLondonUK
| | - Heidi Murray‐Smith
- Department of Neurodegenerative Disease, The Dementia Research CentreUniversity College London (UCL) Queen Square Institute of NeurologyLondonUK
| | - David M. Cash
- Department of Neurodegenerative Disease, The Dementia Research CentreUniversity College London (UCL) Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLUniversity College LondonLondonUK
| | - Josephine Barnes
- Department of Neurodegenerative Disease, The Dementia Research CentreUniversity College London (UCL) Queen Square Institute of NeurologyLondonUK
| | - Carole H. Sudre
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
- Hawkes InstituteDepartment of Computer ScienceUniversity College LondonLondonUK
- School of Biomedical Engineering & Imaging SciencesKing's College London StrandLondonUK
| | - Paresh A. Malhotra
- Imperial College LondonDepartment of Brain Sciences. Burlington DanesThe Hammersmith HospitalLondonUK
- Imperial College London and The University of SurreyUK Dementia Research Institute Care Research and Technology Centre, Sir Michael Uren HubLondonUK
| | - Sebastian J. Crutch
- Department of Neurodegenerative Disease, The Dementia Research CentreUniversity College London (UCL) Queen Square Institute of NeurologyLondonUK
| | - Marcus Richards
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | - Adam Hampshire
- Imperial College LondonDepartment of Brain Sciences. Burlington DanesThe Hammersmith HospitalLondonUK
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonDe Crespigny ParkLondonUK
| | - Jonathan M. Schott
- Department of Neurodegenerative Disease, The Dementia Research CentreUniversity College London (UCL) Queen Square Institute of NeurologyLondonUK
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| |
Collapse
|
3
|
Liu S, Liu J, Li X, Du X, Yin C, Luo Y, Li C. Fluorescent Particles Based on Aggregation-Induced Emission for Optical Diagnostics of the Central Nervous System. RESEARCH (WASHINGTON, D.C.) 2025; 8:0564. [PMID: 39866911 PMCID: PMC11757665 DOI: 10.34133/research.0564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/22/2024] [Accepted: 12/09/2024] [Indexed: 01/28/2025]
Abstract
In 2001, Tang's team discovered a unique type of luminogens with substantial enhanced fluorescence upon aggregation and introduced the concept of "aggregation-induced emission (AIE)". Unlike conventional fluorescent materials, AIE luminogens (AIEgens) emit weak or no fluorescence in solution but become highly fluorescent in aggregated or solid states, due to a mechanism known as restriction of intramolecular motions (RIM). Initially considered a purely inorganic chemical phenomenon, AIE was later applied in biomedicine to improve the sensitivity of immunoassays. Subsequently, AIE has been extensively explored in various biomedical applications, especially in cell imaging. Early studies achieved nonspecific cell imaging using nontargeted AIEgens, and later, specific cellular imaging was realized through the design of targeted AIEgens. These advancements have enabled the visualization of various biomacromolecules and intracellular organelles, providing valuable insights into cellular microenvironments and statuses. Neurological disorders affect over 3 billion people worldwide, highlighting the urgent need for advanced diagnostic and therapeutic tools. AIEgens offer promising opportunities for imaging the central nervous system (CNS), including nerve cells, neural tissues, and blood vessels. This review focuses on the application of AIEgens in CNS imaging, exploring their roles in the diagnosis of various neurological diseases. We will discuss the evolution and conclude with an outlook on the future challenges and opportunities for AIEgens in clinical diagnostics and therapeutics of CNS disorders.
Collapse
Affiliation(s)
- Shan Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital,
University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jinkuan Liu
- School of Medicine,
University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xue Li
- Juxintang (Chengdu) Biotechnology Co. Ltd., Chengdu 641400, China
| | - Xiaoxin Du
- Office of Scientific Research & Development,
University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Cheng Yin
- Department of Neurosurgery, Sichuan Provincial People’s Hospital,
University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yong Luo
- Department of Traditional Chinese Medicine, Sichuan Provincial People’s Hospital,
University of Electronic Science and Technology of China, Chengdu 610031, China
| | - Chenzhong Li
- Juxintang (Chengdu) Biotechnology Co. Ltd., Chengdu 641400, China
- Biomedical Engineering, School of Medicine,
The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
4
|
Alyami BA, Ahmad Z, Ghufran M, Mahnashi MH, Sadiq A, Ayaz M. Appraisal of the Neuroprotective Potentials of Isoeugenol Using In-vitro, In-vivo and In-silico Approaches. Curr Neuropharmacol 2025; 23:317-328. [PMID: 39989009 PMCID: PMC11808583 DOI: 10.2174/1570159x22666240329125626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative condition that affects the elder population and is linked to behavioral instability and cognitive decline. Only a few drugs are approved for clinical management of AD. Volatile oils and their components exhibit diverse pharmacological potentials, including neuroprotective properties. The current study aimed to evaluate isoeugenol's neuroprotective potentials against cognitive impairments caused by scopolamine. METHODS Standard protocols were followed in the In-vitro antioxidant, cholinesterase inhibitory and molecular docking assays. Isoeugenol was initially evaluated for antioxidant potential using DPPH and ABTS free radicals scavenging assays. Subsequently, AChE/BChE inhibition studies were performed following Ellman's assay. To assess the compound's binding effectiveness at the enzymes' target site, it was docked against the binding sites of cholinesterase. The effect of isoeugenol supplementation on scopolamine-induced amnesia was assessed using Shallow Water Maze (SWM), Y-Maze and Elevated Plus Maze (EPM) tests. RESULTS In DPPH and ABTS assays, isoeugenol exhibited considerable efficacy against free radicals with IC50 of 38.97 and 43.76 μg/mL, respectively. Isoeugenol revealed 78.39 ± 0.40% and 67.73 ± 0.03% inhibitions against AChE and BChE, respectively, at 1 mg/ml concentration. In docking studies, isoeugenol exhibited a docking score of -12.2390, forming two hydrogen bonds at the active site residues of AChE. Further, with a docking score of -10.1632, isoeugenol binds adequately to the BChE enzyme via two arene-hydrogen interactions and one hydrogen bond. CONCLUSION Isoeugenol offered considerable protection against scopolamine-induced memory deficits and improved the special memory of the rodents.
Collapse
Affiliation(s)
- Bandar A. Alyami
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Zeeshan Ahmad
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara, 18000 Dir (L), KP, Pakistan
| | - Mehreen Ghufran
- Department of Pathology, MTI Bacha Khan Medical College, Mardan, KP, Pakistan
| | - Mater H. Mahnashi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Najran University, Najran, Kingdom of Saudi Arabia
| | - Abdul Sadiq
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara, 18000 Dir (L), KP, Pakistan
| | - Muhammad Ayaz
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara, 18000 Dir (L), KP, Pakistan
| |
Collapse
|
5
|
Samal M, Srivastava V, Khan M, Insaf A, Penumallu NR, Alam A, Parveen B, Ansari SH, Ahmad S. Therapeutic Potential of Polyphenols in Cellular Reversal of Patho-Mechanisms of Alzheimer's Disease Using In Vitro and In Vivo Models: A Comprehensive Review. Phytother Res 2025; 39:25-50. [PMID: 39496498 DOI: 10.1002/ptr.8344] [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: 03/20/2024] [Revised: 07/28/2024] [Accepted: 08/31/2024] [Indexed: 11/06/2024]
Abstract
Alzheimer's disease (AD) is considered one of the most common neurological conditions associated with memory and cognitive impairment and mainly affects people aged 65 or above. Even with tremendous progress in modern neuroscience, a permanent remedy or cure for this crippling disease is still unattainable. Polyphenols are a group of naturally occurring potent compounds that can modulate the neurodegenerative processes typical of AD. The present comprehensive study has been conducted to find out the preclinical and clinical potential of polyphenols and elucidate their possible mechanisms in managing AD. Additionally, we have reviewed different clinical studies investigating polyphenols as single compounds or cotherapies, including those currently recruiting, completed, terminated, withdrawn, or suspended in AD treatment. Natural polyphenols were systematically screened and identified through electronic databases including Google Scholar, PubMed, and Scopus based on in vitro cell line studies and preclinical data demonstrating their potential for neuroprotection. A total of 63 significant polyphenols were identified. A multimechanistic pathway for polyphenol's mode of action has been proposed in the study. Out of 63, four potent polyphenols have been identified as promising potential candidates, based on their reported clinical efficacy. Polyphenols hold tremendous scope for the development of a future drug molecule as a phytopharmaceutical that may be incorporated as an adjuvant to the therapeutic regime. However, more high-quality studies with novel delivery methods and combinatorial approaches are required to overcome obstacles such as bioavailability and blood-brain barrier crossing to underscore the therapeutic potential of these compounds in AD management.
Collapse
Affiliation(s)
- Monalisha Samal
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Varsha Srivastava
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Muzayyana Khan
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Areeba Insaf
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Naveen Reddy Penumallu
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Aftab Alam
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Bushra Parveen
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Shahid Hussain Ansari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sayeed Ahmad
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
6
|
Bergström S, Mravinacová S, Lindberg O, Zettergren A, Westman E, Wahlund LO, Blennow K, Zetterberg H, Kern S, Skoog I, Månberg A. CSF levels of brain-derived proteins correlate with brain ventricular volume in cognitively healthy 70-year-olds. Clin Proteomics 2024; 21:65. [PMID: 39668376 PMCID: PMC11636040 DOI: 10.1186/s12014-024-09517-1] [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: 09/20/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND The effect of varying brain ventricular volume on the cerebrospinal fluid (CSF) proteome has been discussed as possible confounding factors in comparative protein level analyses. However, the relationship between CSF volume and protein levels remains largely unexplored. Moreover, the few existing studies provide conflicting findings, indicating the need for further research. METHODS Here, we explored the association between levels of 88 pre-selected CSF proteins and ventricular volume derived from magnetic resonance imaging (MRI) measurements in 157 cognitively healthy 70-year-olds from the H70 Gothenburg Birth Cohort Studies, including individuals with and without pathological levels of Alzheimer's disease (AD) CSF markers (n = 123 and 34, respectively). Both left and right lateral, the inferior horn as well as the third and the fourth ventricular volumes were measured. Different antibody-based methods were employed for the protein measurements, with most being analyzed using a multiplex bead-based microarray technology. Furthermore, the associations between the protein levels and cortical thickness, fractional anisotropy, and mean diffusivity were assessed. RESULTS CSF levels of many brain-derived proteins correlated with ventricular volumes in A-T- individuals, with lower levels in individuals with larger ventricles. The strongest negative correlations with total ventricular volume were observed for neurocan (NCAN) and neurosecretory protein VGF (rho = -0.34 for both). Significant negative correlations were observed also for amyloid beta (Ab) 38, Ab40, total tau (t-tau), and phosphorylated tau (p-tau), with correlation ranging between - 0.34 and - 0.28, while no association was observed between ventricular volumes and Ab42 or neurofilament light chain (NfL). Proteins with negative correlations to ventricular volumes further demonstrated negative correlations to mean diffusivity and positive correlation to fractional anisotropy. However, only weak or no correlations were observed between the CSF protein levels and cortical thickness. A + T + individuals demonstrated higher CSF protein levels compared to A-T- individuals with the most significant differences observed for neurogranin (NRGN) and synuclein beta (SNCB). CONCLUSIONS Our findings suggest that the levels of many brain-derived proteins in CSF may be subjected to dilution effects depending on the size of the brain ventricles in healthy individuals without AD pathology. This phenomenon could potentially contribute to the inter-individual variations observed in CSF proteomic studies.
Collapse
Affiliation(s)
- Sofia Bergström
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Sára Mravinacová
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Olof Lindberg
- Division of Clinical Geriatrics, Department of Neurobiology, Center for Alzheimer Research, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Anna Zettergren
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Center for Alzheimer Research, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Lars-Olof Wahlund
- Division of Clinical Geriatrics, Department of Neurobiology, Center for Alzheimer Research, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Silke Kern
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Neuropsychiatry, Mölndal, Sweden
| | - Ingmar Skoog
- Neuropsychiatric Epidemiology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Neuropsychiatry, Mölndal, Sweden
| | - Anna Månberg
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden.
| |
Collapse
|
7
|
Althobaiti NA, Al-Abbas NS, Alsharif I, Albalawi AE, Almars AI, Basabrain AA, Jafer A, Ellatif SA, Bauthman NM, Almohaimeed HM, Soliman MH. Gadd45A-mediated autophagy regulation and its impact on Alzheimer's disease pathogenesis: Deciphering the molecular Nexus. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167353. [PMID: 39004381 DOI: 10.1016/j.bbadis.2024.167353] [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: 12/30/2023] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND The growth arrest and DNA damage-inducible 45 (Gadd45) gene has been implicated in various central nervous system (CNS) functions, both normal and pathological, including aging, memory, and neurodegenerative diseases. In this study, we examined whether Gadd45A deletion triggers pathways associated with neurodegenerative diseases including Alzheimer's disease (AD). METHODS Utilizing transcriptome data from AD-associated hippocampus samples, we identified Gadd45A as a pivotal regulator of autophagy. Comprehensive analyses, including Gene Ontology enrichment and protein-protein interaction network assessments, highlighted Cdkn1A as a significant downstream target of Gadd45A. Experimental validation confirmed Gadd45A's role in modulating Cdkn1A expression and autophagy levels in hippocampal cells. We also examined the effects of autophagy on hippocampal functions and proinflammatory cytokine secretion. Additionally, a murine model was employed to validate the importance of Gadd45A in neuroinflammation and AD pathology. RESULTS Our study identified 20 autophagy regulatory factors associated with AD, with Gadd45A emerging as a critical regulator. Experimental findings demonstrated that Gadd45A influences hippocampal cell fate by reducing Cdkn1A expression and suppressing autophagic activity. Comparisons between wild-type (WT) and Gadd45A knockout (Gadd45A-/-) mice revealed that Gadd45A-/- mice exhibited significant cognitive impairments, including deficits in working and spatial memory, increased Tau hyperphosphorylation, and elevated levels of kinases involved in Tau phosphorylation in the hippocampus. Additionally, Gadd45A-/- mice showed significant increases in pro-inflammatory cytokines and decreases autophagy markers in the brain. Neurotrophin levels and dendritic spine length were also reduced in Gadd45A-/- mice, likely contributing to the observed cognitive deficits. CONCLUSIONS These findings support the direct involvement of the Gadd45A gene in AD pathogenesis, and enhancing the expression of Gadd45A may represent a promising therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Norah A Althobaiti
- Biology Department, College of Science and Humanities, Shaqra University, Saudi Arabia
| | - Nouf S Al-Abbas
- Department of Biology, Jamoum University College, Umm Al-Qura University, 21955 Makkah, Saudi Arabia
| | - Ifat Alsharif
- Department of Biology, Jamoum University College, Umm Al-Qura University, 21955 Makkah, Saudi Arabia
| | - Aishah E Albalawi
- Faculty of Science, Department of Biology, University of Tabuk, Tabuk 47913, Saudi Arabia
| | - Amany I Almars
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ammar A Basabrain
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ayman Jafer
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sawsan Abd Ellatif
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), Alexandria 21934, Egypt
| | - Nuha M Bauthman
- Department of Obstetric & Gynecology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Hailah M Almohaimeed
- Department of Basic Science, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Mona H Soliman
- Botany and Microbiology Department, Faculty of Science, Cairo University, Giza 12613, Egypt; Biology Department, Faculty of Science, Taibah University, Al-Sharm, Yanbu El-Bahr, Yanbu 46429, Saudi Arabia.
| |
Collapse
|
8
|
Bieger A, Brum WS, Borelli WV, Therriault J, De Bastiani MA, Moreira AG, Benedet AL, Ferrari-Souza JP, Da Costa JC, Souza DO, Castilhos RM, Schumacher Schuh AF, Fagundes Chaves ML, Schöll M, Zetterberg H, Blennow K, Pascoal TA, Gauthier S, Rosa-Neto P, Schilling LP, Zimmer ER. Influence of Different Diagnostic Criteria on Alzheimer Disease Clinical Research. Neurology 2024; 103:e209753. [PMID: 39167736 PMCID: PMC11338500 DOI: 10.1212/wnl.0000000000209753] [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: 10/16/2023] [Accepted: 06/14/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Updates in Alzheimer disease (AD) diagnostic guidelines by the National Institute on Aging-Alzheimer's Association (NIA-AA) and the International Working Group (IWG) over the past 11 years may affect clinical diagnoses. We assessed how these guidelines affect clinical AD diagnosis in a cohort of cognitively unimpaired (CU) and cognitively impaired (CI) individuals. METHODS We applied clinical and biomarker data in algorithms to classify individuals from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort according to the following diagnostic guidelines for AD: 2011 NIA-AA, 2016 IWG-2, 2018 NIA-AA, and 2021 IWG-3, assigning the following generic diagnostic labels: (1) not AD (nAD), (2) increased risk of developing AD (irAD), and (3) AD. Diagnostic labels were compared according to their frequency, convergence across guidelines, biomarker profiles, and prognostic value. We also evaluated the diagnostic discordance among the criteria. RESULTS A total of 1,195 individuals (mean age 73.2 ± 7.2 years, mean education 16.1 ± 2.7, 44.0% female) presented different repartitions of diagnostic labels according to the 2011 NIA-AA (nAD = 37.8%, irAD = 23.0%, AD = 39.2%), 2016 IWG-2 (nAD = 37.7%, irAD = 28.7%, AD = 33.6%), 2018 NIA-AA (nAD = 40.7%, irAD = 9.3%, AD = 50.0%), and 2021 IWG-3 (nAD = 51.2%, irAD = 8.4%, AD = 48.3%) frameworks. Discordant diagnoses across all guidelines were found in 512 participants (42.8%) (138 [91.4%] occurring in only β-amyloid [CU 65.4%, CI 34.6%] and 191 [78.6%] in only tau-positive [CU 71.7%, CI 28.3%] individuals). Differences in predicting cognitive impairment between nAD and irAD groups were observed with the 2011 NIA-AA (hazard ratio [HR] 2.21, 95% CI 1.34-3.65, p = 0.002), 2016 IWG-2 (HR 2.81, 95% CI 1.59-4.96, p < 0.000), and 2021 IWG-3 (HR 3.61, 95% CI 2.09-6.23, p < 0.000), but not with 2018 NIA-AA (HR 1.69, 95% CI 0.87-3.28, p = 0.115). DISCUSSION Over 42% of the studied population presented discordant diagnoses when using the different examined AD criteria, mostly in individuals with a single positive biomarker. Except for 2018 NIA-AA, all guidelines identified asymptomatic individuals at risk of cognitive impairment. Our findings highlight important differences between the guidelines, emphasizing the necessity for updated criteria with enhanced staging metrics, considering clinical, research, therapeutic, and trial design aspects.
Collapse
Affiliation(s)
- Andrei Bieger
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Wagner S Brum
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Wyllians V Borelli
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Joseph Therriault
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Marco A De Bastiani
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Amanda G Moreira
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Andrea L Benedet
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - João Pedro Ferrari-Souza
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Jaderson C Da Costa
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Diogo O Souza
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Raphael M Castilhos
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Artur Francisco Schumacher Schuh
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Marcia L Fagundes Chaves
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Michael Schöll
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Henrik Zetterberg
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Kaj Blennow
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Tharick A Pascoal
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Serge Gauthier
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Pedro Rosa-Neto
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Lucas P Schilling
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Eduardo R Zimmer
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| |
Collapse
|
9
|
Qiang RR, Xiang Y, Zhang L, Bai XY, Zhang D, Li YJ, Yang YL, Liu XL. Ferroptosis: A new strategy for targeting Alzheimer's disease. Neurochem Int 2024; 178:105773. [PMID: 38789042 DOI: 10.1016/j.neuint.2024.105773] [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: 03/08/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a complex pathogenesis, which involves the formation of amyloid plaques and neurofibrillary tangles. Many recent studies have revealed a close association between ferroptosis and the pathogenesis of AD. Factors such as ferroptosis-associated iron overload, lipid peroxidation, disturbances in redox homeostasis, and accumulation of reactive oxygen species have been found to contribute to the pathological progression of AD. In this review, we explore the mechanisms underlying ferroptosis, describe the link between ferroptosis and AD, and examine the reported efficacy of ferroptosis inhibitors in treating AD. Finally, we discuss the potential challenges to ferroptosis inhibitors use in the clinic, enabling their faster use in clinical treatment.
Collapse
Affiliation(s)
| | - Yang Xiang
- College of Physical Education, Yan'an University, Shaanxi, 716000, China
| | - Lei Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Xin Yue Bai
- School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- School of Medicine, Yan'an University, Yan'an, China
| | - Yang Jing Li
- School of Medicine, Yan'an University, Yan'an, China
| | - Yan Ling Yang
- School of Medicine, Yan'an University, Yan'an, China
| | - Xiao Long Liu
- School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
10
|
Dittrich A, Westman E, Shams S, Skillbäck T, Zetterberg H, Blennow K, Zettergren A, Skoog I, Kern S. Proportion of Community-Dwelling Individuals Older Than 70 Years Eligible for Lecanemab Initiation: The Gothenburg H70 Birth Cohort Study. Neurology 2024; 102:e209402. [PMID: 38593394 PMCID: PMC11175644 DOI: 10.1212/wnl.0000000000209402] [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: 12/05/2023] [Accepted: 02/21/2024] [Indexed: 04/11/2024] Open
Abstract
OBJECTIVES To determine the prevalence of individuals with Alzheimer disease (AD) eligible for treatment with the recently FDA-approved lecanemab based on data from a population-based sample of 70-year-olds and extrapolate an estimation of individuals eligible in Europe and the United States. METHODS Participants from the Gothenburg H70 Birth Cohort Study with clinical data, CSF-amyloid beta 42, and brain MRI analysis were evaluated for eligibility to receive lecanemab treatment according to FDA-approved recommendations, noting factors requiring special consideration. Results were used to extrapolate the number of eligible individuals in Europe and the United States using public demographic data. RESULTS Thirty (10.3%) of 290 participants met the indication for treatment of whom 18 (6.2%) were eligible and did not present factors requiring special consideration. Our estimate that 6.2% of all 70-year-olds in the full cohort are eligible for treatment extrapolates to an approximation that around 5.9 million Europeans and 2.2 million US residents could be eligible. DISCUSSION Information on proportion of individuals eligible for AD treatment with lecanemab in the general public is limited. We provide information on 70-year-olds in Sweden and extrapolate these data to Europe and the United States. This study opens for larger studies on this proportion and implementation of lecanemab treatment.
Collapse
Affiliation(s)
- Anna Dittrich
- From the Neuropsychiatric Epidemiology Unit (A.D., T.S., A.Z., I.S., S.K.), Sahlgrenska Academy, University of Gothenburg; Department of Psychiatry Cognition and Old Age Psychiatry (A.D., I.S., S.K.), Sahlgrenska University Hospital, Mölndal; Division of Clinical Geriatrics (E.W.), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska University Hospital, Stockholm, Sweden; Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Care Sciences and Society (S.S.), Karolinska Institutet, and Department of Radiology; Department of Clinical Neuroscience (S.S.), Karolinska University Hospital, Stockholm Sweden; Department of Radiology (S.S.), Stanford University, CA; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal Sweden; UK Dementia Research Institute at UCL (H.Z.); Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, London United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Hong Kong, China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison; Paris Brain Institute (K.B.), ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; and Neurodegenerative Disorder Research Center (K.B.), Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Eric Westman
- From the Neuropsychiatric Epidemiology Unit (A.D., T.S., A.Z., I.S., S.K.), Sahlgrenska Academy, University of Gothenburg; Department of Psychiatry Cognition and Old Age Psychiatry (A.D., I.S., S.K.), Sahlgrenska University Hospital, Mölndal; Division of Clinical Geriatrics (E.W.), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska University Hospital, Stockholm, Sweden; Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Care Sciences and Society (S.S.), Karolinska Institutet, and Department of Radiology; Department of Clinical Neuroscience (S.S.), Karolinska University Hospital, Stockholm Sweden; Department of Radiology (S.S.), Stanford University, CA; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal Sweden; UK Dementia Research Institute at UCL (H.Z.); Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, London United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Hong Kong, China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison; Paris Brain Institute (K.B.), ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; and Neurodegenerative Disorder Research Center (K.B.), Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Sara Shams
- From the Neuropsychiatric Epidemiology Unit (A.D., T.S., A.Z., I.S., S.K.), Sahlgrenska Academy, University of Gothenburg; Department of Psychiatry Cognition and Old Age Psychiatry (A.D., I.S., S.K.), Sahlgrenska University Hospital, Mölndal; Division of Clinical Geriatrics (E.W.), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska University Hospital, Stockholm, Sweden; Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Care Sciences and Society (S.S.), Karolinska Institutet, and Department of Radiology; Department of Clinical Neuroscience (S.S.), Karolinska University Hospital, Stockholm Sweden; Department of Radiology (S.S.), Stanford University, CA; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal Sweden; UK Dementia Research Institute at UCL (H.Z.); Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, London United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Hong Kong, China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison; Paris Brain Institute (K.B.), ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; and Neurodegenerative Disorder Research Center (K.B.), Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Tobias Skillbäck
- From the Neuropsychiatric Epidemiology Unit (A.D., T.S., A.Z., I.S., S.K.), Sahlgrenska Academy, University of Gothenburg; Department of Psychiatry Cognition and Old Age Psychiatry (A.D., I.S., S.K.), Sahlgrenska University Hospital, Mölndal; Division of Clinical Geriatrics (E.W.), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska University Hospital, Stockholm, Sweden; Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Care Sciences and Society (S.S.), Karolinska Institutet, and Department of Radiology; Department of Clinical Neuroscience (S.S.), Karolinska University Hospital, Stockholm Sweden; Department of Radiology (S.S.), Stanford University, CA; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal Sweden; UK Dementia Research Institute at UCL (H.Z.); Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, London United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Hong Kong, China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison; Paris Brain Institute (K.B.), ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; and Neurodegenerative Disorder Research Center (K.B.), Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Henrik Zetterberg
- From the Neuropsychiatric Epidemiology Unit (A.D., T.S., A.Z., I.S., S.K.), Sahlgrenska Academy, University of Gothenburg; Department of Psychiatry Cognition and Old Age Psychiatry (A.D., I.S., S.K.), Sahlgrenska University Hospital, Mölndal; Division of Clinical Geriatrics (E.W.), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska University Hospital, Stockholm, Sweden; Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Care Sciences and Society (S.S.), Karolinska Institutet, and Department of Radiology; Department of Clinical Neuroscience (S.S.), Karolinska University Hospital, Stockholm Sweden; Department of Radiology (S.S.), Stanford University, CA; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal Sweden; UK Dementia Research Institute at UCL (H.Z.); Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, London United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Hong Kong, China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison; Paris Brain Institute (K.B.), ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; and Neurodegenerative Disorder Research Center (K.B.), Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Kaj Blennow
- From the Neuropsychiatric Epidemiology Unit (A.D., T.S., A.Z., I.S., S.K.), Sahlgrenska Academy, University of Gothenburg; Department of Psychiatry Cognition and Old Age Psychiatry (A.D., I.S., S.K.), Sahlgrenska University Hospital, Mölndal; Division of Clinical Geriatrics (E.W.), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska University Hospital, Stockholm, Sweden; Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Care Sciences and Society (S.S.), Karolinska Institutet, and Department of Radiology; Department of Clinical Neuroscience (S.S.), Karolinska University Hospital, Stockholm Sweden; Department of Radiology (S.S.), Stanford University, CA; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal Sweden; UK Dementia Research Institute at UCL (H.Z.); Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, London United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Hong Kong, China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison; Paris Brain Institute (K.B.), ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; and Neurodegenerative Disorder Research Center (K.B.), Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Anna Zettergren
- From the Neuropsychiatric Epidemiology Unit (A.D., T.S., A.Z., I.S., S.K.), Sahlgrenska Academy, University of Gothenburg; Department of Psychiatry Cognition and Old Age Psychiatry (A.D., I.S., S.K.), Sahlgrenska University Hospital, Mölndal; Division of Clinical Geriatrics (E.W.), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska University Hospital, Stockholm, Sweden; Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Care Sciences and Society (S.S.), Karolinska Institutet, and Department of Radiology; Department of Clinical Neuroscience (S.S.), Karolinska University Hospital, Stockholm Sweden; Department of Radiology (S.S.), Stanford University, CA; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal Sweden; UK Dementia Research Institute at UCL (H.Z.); Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, London United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Hong Kong, China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison; Paris Brain Institute (K.B.), ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; and Neurodegenerative Disorder Research Center (K.B.), Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Ingmar Skoog
- From the Neuropsychiatric Epidemiology Unit (A.D., T.S., A.Z., I.S., S.K.), Sahlgrenska Academy, University of Gothenburg; Department of Psychiatry Cognition and Old Age Psychiatry (A.D., I.S., S.K.), Sahlgrenska University Hospital, Mölndal; Division of Clinical Geriatrics (E.W.), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska University Hospital, Stockholm, Sweden; Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Care Sciences and Society (S.S.), Karolinska Institutet, and Department of Radiology; Department of Clinical Neuroscience (S.S.), Karolinska University Hospital, Stockholm Sweden; Department of Radiology (S.S.), Stanford University, CA; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal Sweden; UK Dementia Research Institute at UCL (H.Z.); Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, London United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Hong Kong, China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison; Paris Brain Institute (K.B.), ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; and Neurodegenerative Disorder Research Center (K.B.), Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Silke Kern
- From the Neuropsychiatric Epidemiology Unit (A.D., T.S., A.Z., I.S., S.K.), Sahlgrenska Academy, University of Gothenburg; Department of Psychiatry Cognition and Old Age Psychiatry (A.D., I.S., S.K.), Sahlgrenska University Hospital, Mölndal; Division of Clinical Geriatrics (E.W.), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska University Hospital, Stockholm, Sweden; Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; Care Sciences and Society (S.S.), Karolinska Institutet, and Department of Radiology; Department of Clinical Neuroscience (S.S.), Karolinska University Hospital, Stockholm Sweden; Department of Radiology (S.S.), Stanford University, CA; Department of Psychiatry and Neurochemistry (H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Mölndal Sweden; UK Dementia Research Institute at UCL (H.Z.); Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, London United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.), Hong Kong, China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison; Paris Brain Institute (K.B.), ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; and Neurodegenerative Disorder Research Center (K.B.), Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| |
Collapse
|
11
|
He K, Li B, Huang L, Zhao J, Hua F, Wang T, Li J, Wang J, Huang Q, Chen K, Xu S, Ren S, Cai H, Jiang D, Hu J, Han X, Guan Y, Chen K, Guo Q, Xie F. Positive rate and quantification of amyloid pathology with [ 18F]florbetapir in the urban Chinese population. Eur Radiol 2024; 34:3331-3341. [PMID: 37889270 DOI: 10.1007/s00330-023-10366-z] [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: 11/26/2022] [Revised: 09/10/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023]
Abstract
OBJECTIVES Amyloid deposition is considered the initial pathology in Alzheimer's disease (AD). Personalized management requires investigation of amyloid pathology and the risk factors for both amyloid pathology and cognitive decline in the Chinese population. We aimed to investigate amyloid positivity and deposition in AD patients, as well as factors related to amyloid pathology in Chinese cities. METHODS This cross-sectional multicenter study was conducted in Shanghai and Zhengzhou, China. All participants were recruited from urban communities and memory clinics. Amyloid positivity and deposition were analyzed based on amyloid positron emission tomography (PET). We used partial least squares (PLS) models to investigate how related factors contributed to amyloid deposition and cognitive decline. RESULTS In total, 1026 participants were included: 768 participants from the community-based cohort (COMC) and 258 participants from the clinic-based cohort (CLIC). The overall amyloid-positive rates in individuals with clinically diagnosed AD, mild cognitive impairment (MCI), and normal cognition (NC) were 85.8%, 44.5%, and 26.9%, respectively. The global amyloid deposition standardized uptake value ratios (SUVr) (reference: cerebellar crus) were 1.44 ± 0.24, 1.30 ± 0.22, and 1.24 ± 0.14, respectively. CLIC status, apolipoprotein E (ApoE) ε4, and older age were strongly associated with amyloid pathology by PLS modeling. CONCLUSION The overall amyloid-positive rates accompanying AD, MCI, and NC in the Chinese population were similar to those in published cohorts of other populations. ApoE ε4 and CLIC status were risk factors for amyloid pathology across the AD continuum. Education was a risk factor for amyloid pathology in MCI. Female sex and age were risk factors for amyloid pathology in NC. CLINICAL RELEVANCE STATEMENT This study provides new details about amyloid pathology in the Chinese population. Factors related to amyloid deposition and cognitive decline can help to assess patients' AD risk. KEY POINTS • We studied amyloid pathology and related risk factors in the Chinese population. •·The overall amyloid-positive rates in individuals with clinically diagnosed AD, MCI, and NC were 85.8%, 44.5%, and 26.9%, respectively. • These overall amyloid-positive rates were in close agreement with the corresponding prevalence for other populations.
Collapse
Affiliation(s)
- Kun He
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Binyin Li
- Department of Neurology and Institute of Neurology, School of Medicine Affiliated Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200020, China
| | - Lin Huang
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Fengchun Hua
- Department of Nuclear Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Tao Wang
- Department of Geriatric Psychiatry, Shanghai Mental Health Centre, Shanghai Jiaotong University School of Medicine, Shanghai, 200122, China
| | - Junpeng Li
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jie Wang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qi Huang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Keliang Chen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shasha Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Shuhua Ren
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Huawei Cai
- Laboratory of Clinical Nuclear Medicine, Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Donglang Jiang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jingchao Hu
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
- School of Nursing, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Xingmin Han
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Kewei Chen
- Banner Alzheimer Institute, Arizona State University, University of Arizona and Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
12
|
Noda K, Lim Y, Goto R, Sengoku S, Kodama K. Cost-effectiveness comparison between blood biomarkers and conventional tests in Alzheimer's disease diagnosis. Drug Discov Today 2024; 29:103911. [PMID: 38311028 DOI: 10.1016/j.drudis.2024.103911] [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: 11/15/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/06/2024]
Abstract
Dementia management has evolved with drugs such as lecanemab, shifting management from palliative care to early diagnosis and intervention. However, the administration of these drugs presents challenges owing to the invasiveness, high cost and limited availability of amyloid-PET and cerebrospinal fluid tests for guiding drug administration. Our manuscript explores the potential of less invasive blood biomarkers as a diagnostic method, with a cost-effectiveness analysis and a comparison with traditional tests. Our findings suggest that blood biomarkers are a cost-effective alternative, but with lower accuracy, indicating the need for multiple specific biomarkers for precision. This underscores the importance of future research on new blood biomarkers and their clinical efficacy.
Collapse
Affiliation(s)
- Kenta Noda
- Graduate School of Design and Architecture, Nagoya City University, Nagoya 464-0083, Japan
| | | | - Rei Goto
- Graduate School of Health Management, Keio University, Fujisawa 252-0883, Kanagawa, Japan; Graduate School of Business Administration, Keio University, Yokohama 223-8526, Japan
| | - Shintaro Sengoku
- School of Environment and Society, Tokyo Institute of Technology, Tokyo 108-0023, Japan
| | - Kota Kodama
- Graduate School of Design and Architecture, Nagoya City University, Nagoya 464-0083, Japan; Ritsumeikan University, Osaka 567-8570, Japan; Faculty of Data Science, Nagoya City University, Nagoya 467-8501, Japan; Center for Research and Education on Drug Discovery, The Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan.
| |
Collapse
|
13
|
Peters‐Founshtein G, Gazit L, Naveh T, Domachevsky L, Korczyn AD, Bernstine H, Shaharabani‐Gargir L, Groshar D, Marshall GA, Arzy S. Lost in space(s): Multimodal neuroimaging of disorientation along the Alzheimer's disease continuum. Hum Brain Mapp 2024; 45:e26623. [PMID: 38488454 PMCID: PMC10941506 DOI: 10.1002/hbm.26623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 01/02/2024] [Accepted: 01/27/2024] [Indexed: 03/18/2024] Open
Abstract
Orientation is a fundamental cognitive faculty and the bedrock of the neurologic examination. Orientation is defined as the alignment between an individual's internal representation and the external world in the spatial, temporal, and social domains. While spatial disorientation is a recognized hallmark of Alzheimer's disease (AD), little is known about disorientation beyond space in AD. This study aimed to explore disorientation in spatial, temporal, and social domains along the AD continuum. Fifty-one participants along the AD continuum performed an ecological orientation task in the spatial, temporal, and social domains while undergoing functional MRI. Disorientation in AD followed a three-way association between orientation domain, brain region, and disease stage. Specifically, patients with early amnestic mild cognitive impairment exhibited spatio-temporal disorientation and reduced brain activity in temporoparietal regions, while patients with AD dementia showed additional social disorientation and reduced brain activity in frontoparietal regions. Furthermore, patterns of hypoactivation overlapped different subnetworks of the default mode network, patterns of fluorodeoxyglucose hypometabolism, and cortical atrophy characteristic of AD. Our results suggest that AD may encompass a disorder of orientation, characterized by a biphasic process manifesting as early spatio-temporal and late social disorientation. As such, disorientation may offer a unique window into the clinicopathological progression of AD. SIGNIFICANCE STATEMENT: Despite extensive research into Alzheimer's disease (AD), its core cognitive deficit remains a matter of debate. In this study, we investigated whether orientation, defined as the ability to align internal representations with the external world in spatial, temporal, and social domains, constitutes a core cognitive deficit in AD. To do so, we used PET-fMRI imaging to collect behavioral, functional, and metabolic data from 51 participants along the AD continuum. Our findings suggest that AD may constitute a disorder of orientation, characterized by an early spatio-temporal disorientation and followed by late social disorientation, manifesting in task-evoked and neurodegenerative changes. We propose that a profile of disorientation across multiple domains offers a unique window into the progression of AD and as such could greatly benefit disease diagnosis, monitoring, and evaluation of treatment response.
Collapse
Affiliation(s)
- Gregory Peters‐Founshtein
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
- Department of Nuclear MedicineSheba Medical CenterRamat‐GanIsrael
| | - Lidor Gazit
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
- Department of NeurologyHadassah Hebrew University Medical SchoolJerusalemIsrael
| | - Tahel Naveh
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
- Department of NeurologyHadassah Hebrew University Medical SchoolJerusalemIsrael
| | - Liran Domachevsky
- Department of Nuclear MedicineSheba Medical CenterRamat‐GanIsrael
- Department of Nuclear MedicineAssuta Medical CenterTel‐AvivIsrael
| | | | - Hanna Bernstine
- Department of Nuclear MedicineAssuta Medical CenterTel‐AvivIsrael
- Department of ImagingTel‐Aviv UniversityTel‐AvivIsrael
- Department of Nuclear MedicineRabin Medical CenterPetah TikvaIsrael
| | | | - David Groshar
- Department of Nuclear MedicineAssuta Medical CenterTel‐AvivIsrael
- Department of ImagingTel‐Aviv UniversityTel‐AvivIsrael
| | - Gad A. Marshall
- Department of Neurology, Center for Alzheimer Research and Treatment, Harvard Medical School, Brigham and Women's HospitalMassachusetts General HospitalBostonMassachusettsUSA
| | - Shahar Arzy
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of MedicineHebrew University of JerusalemJerusalemIsrael
- Department of NeurologyHadassah Hebrew University Medical SchoolJerusalemIsrael
| |
Collapse
|
14
|
Seidu NM, Kern S, Sacuiu S, Sterner TR, Blennow K, Zetterberg H, Lindberg O, Ferreira D, Westman E, Zettergren A, Skoog I. Association of CSF biomarkers with MRI brain changes in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12556. [PMID: 38406609 PMCID: PMC10884990 DOI: 10.1002/dad2.12556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/24/2024] [Indexed: 02/27/2024]
Abstract
The relation between cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) and magnetic resonance imaging (MRI) measures is poorly understood in cognitively healthy individuals from the general population. Participants' (n = 226) mean age was 70.9 years (SD = 0.4). CSF concentrations of amyloid beta (Aβ)1-42, total tau (t-tau), phosphorylated tau (p-tau), neurogranin, and neurofilament light, and volumes of hippocampus, amygdala, total basal forebrain (TBF), and cortical thickness were measured. Linear associations between CSF biomarkers and MRI measures were investigated. In Aβ1-42 positives, higher t-tau and p-tau were associated with smaller hippocampus (P = 0.001 and P = 0.003) and amygdala (P = 0.005 and P = 0.01). In Aβ1-42 negatives, higher t-tau, p-tau, and neurogranin were associated with larger TBF volume (P = 0.001, P = 0.001, and P = 0.01). No associations were observed between the CSF biomarkers and an AD signature score of cortical thickness. AD-specific biomarkers in cognitively healthy 70-year-olds may be related to TBF, hippocampus, and amygdala. Lack of association with cortical thickness might be due to early stage of disease.
Collapse
Affiliation(s)
- Nazib M Seidu
- Neuropsychiatric Epidemiology (EPINEP)Centre for Ageing and Health (AGECAP)Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Silke Kern
- Neuropsychiatric Epidemiology (EPINEP)Centre for Ageing and Health (AGECAP)Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University Hospital, Region Västra GötalandGothenburgSweden
| | - Simona Sacuiu
- Neuropsychiatric Epidemiology (EPINEP)Centre for Ageing and Health (AGECAP)Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University Hospital, Region Västra GötalandGothenburgSweden
- Division of Clinical GeriatricsDepartment of NeurobiologyCare Sciences and SocietyCenter for Alzheimer ResearchKarolinska InstitutetStockholmSweden
- Cognitive Disorders ClinicTheme Inflammation and AgingKarolinska University HospitalStockholmSweden
| | - Therese Rydberg Sterner
- Neuropsychiatric Epidemiology (EPINEP)Centre for Ageing and Health (AGECAP)Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Aging Research CenterDepartment of NeurobiologyCare Sciences and SocietyKarolinska Institutet and Stockholm UniversityStockholmSweden
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University Hospital, Region Västra GötalandGothenburgSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University Hospital, Region Västra GötalandGothenburgSweden
- UK Dementia Research Institute at UCLLondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- UW Department of MedicineSchool of Medicine and Public HealthMadisonWisconsinUSA
| | - Olof Lindberg
- Division of Clinical GeriatricsDepartment of NeurobiologyCare Sciences and SocietyCenter for Alzheimer ResearchKarolinska InstitutetStockholmSweden
| | - Daniel Ferreira
- Division of Clinical GeriatricsDepartment of NeurobiologyCare Sciences and SocietyCenter for Alzheimer ResearchKarolinska InstitutetStockholmSweden
- Facultad de Ciencias de la SaludUniversidad Fernando Pessoa CanariasLas PalmasSpain
| | - Eric Westman
- Division of Clinical GeriatricsDepartment of NeurobiologyCare Sciences and SocietyCenter for Alzheimer ResearchKarolinska InstitutetStockholmSweden
- Department of NeuroimagingCentre for Neuroimaging SciencesInstitute of PsychiatryPsychology and NeuroscienceKing's College LondonLondonUK
| | - Anna Zettergren
- Neuropsychiatric Epidemiology (EPINEP)Centre for Ageing and Health (AGECAP)Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Ingmar Skoog
- Neuropsychiatric Epidemiology (EPINEP)Centre for Ageing and Health (AGECAP)Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University Hospital, Region Västra GötalandGothenburgSweden
| |
Collapse
|
15
|
Marx GA, Kauffman J, McKenzie AT, Koenigsberg DG, McMillan CT, Morgello S, Karlovich E, Insausti R, Richardson TE, Walker JM, White CL, Babrowicz BM, Shen L, McKee AC, Stein TD, Farrell K, Crary JF. Histopathologic brain age estimation via multiple instance learning. Acta Neuropathol 2023; 146:785-802. [PMID: 37815677 PMCID: PMC10627911 DOI: 10.1007/s00401-023-02636-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023]
Abstract
Understanding age acceleration, the discordance between biological and chronological age, in the brain can reveal mechanistic insights into normal physiology as well as elucidate pathological determinants of age-related functional decline and identify early disease changes in the context of Alzheimer's and other disorders. Histopathological whole slide images provide a wealth of pathologic data on the cellular level that can be leveraged to build deep learning models to assess age acceleration. Here, we used a collection of digitized human post-mortem hippocampal sections to develop a histological brain age estimation model. Our model predicted brain age within a mean absolute error of 5.45 ± 0.22 years, with attention weights corresponding to neuroanatomical regions vulnerable to age-related changes. We found that histopathologic brain age acceleration had significant associations with clinical and pathologic outcomes that were not found with epigenetic based measures. Our results indicate that histopathologic brain age is a powerful, independent metric for understanding factors that contribute to brain aging.
Collapse
Affiliation(s)
- Gabriel A Marx
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Justin Kauffman
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Andrew T McKenzie
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel G Koenigsberg
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Cory T McMillan
- Frontotemporal Degeneration Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan Morgello
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, New York, NY, USA
| | - Esma Karlovich
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Ricardo Insausti
- Human Neuroanatomy Laboratory, School of Medicine, University of Castilla-La Mancha, Albacete, Spain
| | - Timothy E Richardson
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Jamie M Walker
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Charles L White
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bergan M Babrowicz
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, New York, NY, USA
| | - Ann C McKee
- Department of Pathology, Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Thor D Stein
- Department of Pathology, Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Kurt Farrell
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA.
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA.
| | - John F Crary
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA.
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA.
| |
Collapse
|
16
|
Xie F, Ni R. Collection on molecular imaging in neurodegeneration. Eur J Nucl Med Mol Imaging 2023; 50:3166-3167. [PMID: 37480370 DOI: 10.1007/s00259-023-06347-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Affiliation(s)
- Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Ruiqing Ni
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Erickson P, Simrén J, Brum WS, Ennis GE, Kollmorgen G, Suridjan I, Langhough R, Jonaitis EM, Van Hulle CA, Betthauser TJ, Carlsson CM, Asthana S, Ashton NJ, Johnson SC, Shaw LM, Blennow K, Andreasson U, Bendlin BB, Zetterberg H. Prevalence and Clinical Implications of a β-Amyloid-Negative, Tau-Positive Cerebrospinal Fluid Biomarker Profile in Alzheimer Disease. JAMA Neurol 2023; 80:2807607. [PMID: 37523162 PMCID: PMC10391361 DOI: 10.1001/jamaneurol.2023.2338] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/05/2023] [Indexed: 08/01/2023]
Abstract
Importance Knowledge is lacking on the prevalence and prognosis of individuals with a β-amyloid-negative, tau-positive (A-T+) cerebrospinal fluid (CSF) biomarker profile. Objective To estimate the prevalence of a CSF A-T+ biomarker profile and investigate its clinical implications. Design, Setting, and Participants This was a retrospective cohort study of the cross-sectional multicenter University of Gothenburg (UGOT) cohort (November 2019-January 2021), the longitudinal multicenter Alzheimer Disease Neuroimaging Initiative (ADNI) cohort (individuals with mild cognitive impairment [MCI] and no cognitive impairment; September 2005-May 2022), and 2 Wisconsin cohorts, Wisconsin Alzheimer Disease Research Center and Wisconsin Registry for Alzheimer Prevention (WISC; individuals without cognitive impairment; February 2007-November 2020). This was a multicenter study, with data collected from referral centers in clinical routine (UGOT) and research settings (ADNI and WISC). Eligible individuals had 1 lumbar puncture (all cohorts), 2 or more cognitive assessments (ADNI and WISC), and imaging (ADNI only) performed on 2 separate occasions. Data were analyzed on August 2022 to April 2023. Exposures Baseline CSF Aβ42/40 and phosphorylated tau (p-tau)181; cognitive tests (ADNI: modified preclinical Alzheimer cognitive composite [mPACC]; WISC: modified 3-test PACC [PACC-3]). Exposures in the ADNI cohort included [18F]-florbetapir amyloid positron emission tomography (PET), magnetic resonance imaging (MRI), [18F]-fluorodeoxyglucose PET (FDG-PET), and cross-sectional tau-PET (ADNI: [18F]-flortaucipir, WISC: [18F]-MK6240). Main Outcomes and Measures Primary outcomes were the prevalence of CSF AT biomarker profiles and continuous longitudinal global cognitive outcome and imaging biomarker trajectories in A-T+ vs A-T- groups. Secondary outcomes included cross-sectional tau-PET. Results A total of 7679 individuals (mean [SD] age, 71.0 [8.4] years; 4101 male [53%]) were included in the UGOT cohort, 970 individuals (mean [SD] age, 73 [7.0] years; 526 male [54%]) were included in the ADNI cohort, and 519 individuals (mean [SD] age, 60 [7.3] years; 346 female [67%]) were included in the WISC cohort. The prevalence of an A-T+ profile in the UGOT cohort was 4.1% (95% CI, 3.7%-4.6%), being less common than the other patterns. Longitudinally, no significant differences in rates of worsening were observed between A-T+ and A-T- profiles for cognition or imaging biomarkers. Cross-sectionally, A-T+ had similar tau-PET uptake to individuals with an A-T- biomarker profile. Conclusion and Relevance Results suggest that the CSF A-T+ biomarker profile was found in approximately 5% of lumbar punctures and was not associated with a higher rate of cognitive decline or biomarker signs of disease progression compared with biomarker-negative individuals.
Collapse
Affiliation(s)
- Pontus Erickson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joel Simrén
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Wagner S. Brum
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gilda E. Ennis
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | | | | | - Rebecca Langhough
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Erin M. Jonaitis
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Carol A. Van Hulle
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Tobey J. Betthauser
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Cynthia M. Carlsson
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Sanjay Asthana
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Nicholas J. Ashton
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, King’s College London, London, England
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, England
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Sterling C. Johnson
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ulf Andreasson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Barbara B. Bendlin
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Institute of Neurology, Department of Neurodegenerative Disease, University College London, London, England
- UK Dementia Research Institute, University College London, London, England
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| |
Collapse
|
18
|
Baicalein ameliorates Alzheimer's disease via orchestration of CX3CR1/NF-κB pathway in a triple transgenic mouse model. Int Immunopharmacol 2023; 118:109994. [PMID: 37098656 DOI: 10.1016/j.intimp.2023.109994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/21/2023] [Accepted: 03/04/2023] [Indexed: 03/15/2023]
Abstract
Alzheimer's disease (AD) is a common chronic neurodegenerative disease. Some studies have suggested that dysregulation of microglia activation and the resulting neuroinflammation play an important role in the development of AD pathology. Activated microglia have both M1 and M2 phenotypes and inhibition of M1 phenotype while stimulating M2 phenotype has been considered as a potential treatment for neuroinflammation-related diseases. Baicalein is a class of flavonoids with anti-inflammatory, antioxidant and other biological activities, but its role in AD and the regulation of microglia are limited. The purpose of this study was to investigate the effect of baicalein on the activation of microglia in AD model mice and the related molecular mechanism. Our results showed that baicalein significantly improved the learning and memory ability and AD-related pathology of 3 × Tg-AD mice, inhibited the level of pro-inflammatory factors TNF-α, IL-1β and IL-6, promoted the production of anti-inflammatory factors IL-4 and IL-10, and regulated the microglia phenotype through CX3CR1/NF-κB signaling pathway. In conclusion, baicalein can regulate the phenotypic transformation of activated microglia and reduce neuroinflammation through CX3CR1/NF-κB pathway, thereby improving the learning and memory ability of 3 × Tg-AD mice.
Collapse
|
19
|
Arvidsson Rådestig M, Skoog I, Skillbäck T, Zetterberg H, Kern J, Zettergren A, Andreasson U, Wetterberg H, Kern S, Blennow K. Cerebrospinal fluid biomarkers of axonal and synaptic degeneration in a population-based sample. Alzheimers Res Ther 2023; 15:44. [PMID: 36869347 PMCID: PMC9983206 DOI: 10.1186/s13195-023-01193-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/14/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND Neurofilament light (NfL) and neurogranin (Ng) are promising candidate AD biomarkers, reflecting axonal and synaptic damage, respectively. Since there is a need to understand the synaptic and axonal damage in preclinical Alzheimer's disease (AD), we aimed to determine the cerebrospinal fluid (CSF) levels of NfL and Ng in cognitively unimpaired elderly from the Gothenburg H70 Birth Cohort Studies classified according to the amyloid/tau/neurodegeneration (A/T/N) system. METHODS The sample consisted of 258 cognitively unimpaired older adults (age 70, 129 women and 129 men) from the Gothenburg Birth Cohort Studies. We compared CSF NfL and Ng concentrations in A/T/N groups using Student's T-test and ANCOVA. RESULTS CSF NfL concentration was higher in the A-T-N+ group (p=0.001) and the A-T+N+ group (p=0.006) compared with A-T-N-. CSF Ng concentration was higher in the A-T-N+, A-T+N+, A+T-N+, and A+T+N+ groups (p<0.0001) compared with A-T-N-. We found no difference in NfL or Ng concentration in A+ compared with A- (disregarding T- and N- status), whereas those with N+ had higher concentrations of NfL and Ng compared with N- (p<0.0001) (disregarding A- and T- status). CONCLUSIONS CSF NfL and Ng concentrations are increased in cognitively normal older adults with biomarker evidence of tau pathology and neurodegeneration.
Collapse
Affiliation(s)
- Maya Arvidsson Rådestig
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ingmar Skoog
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry, Cognition and Old Age Psychiatry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tobias Skillbäck
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden. .,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK.,UK Dementia Research Institute at UCL, London, WC1N 3BG, UK.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Jürgen Kern
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Anna Zettergren
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Hanna Wetterberg
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Silke Kern
- Department of Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry, Cognition and Old Age Psychiatry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
20
|
Kasuga K, Tsukie T, Kikuchi M, Tokutake T, Washiyama K, Simizu S, Yoshizawa H, Kuroha Y, Yajima R, Mori H, Arakawa Y, Onda K, Miyashita A, Onodera O, Iwatsubo T, Ikeuchi T. The Clinical Application of Optimized AT(N) Classification in Alzheimer’s Clinical Syndrome (ACS) and non-ACS Conditions. Neurobiol Aging 2023; 127:23-32. [PMID: 37030016 DOI: 10.1016/j.neurobiolaging.2023.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
We aimed to assess the utility of AT(N) classification in clinical practice. We measured the cerebrospinal fluid levels of amyloid-β (Aβ) 42, Aβ40, phosphorylated tau, total tau, and neurofilament light chain (NfL) in samples from 230 patients with Alzheimer's clinical syndrome (ACS) and 328 patients with non-ACS. The concordance of two A-markers (i.e., Aβ42 alone and the Aβ42/Aβ40 ratio) was not significantly different between the ACS (87.4%) and non-ACS (74.1%) groups. However, the frequency of discordant cases with AAβ42-alone+/AAβ-ratio- was significantly higher in the non-ACS (23.8%) than in the ACS group (7.4%). The concordance of two N-markers (i.e., total tau and NfL) was 40.4% in the ACS group and 24.4% in the non-ACS group. In the ACS samples, the frequency of biological Alzheimer's disease (i.e., A+T+) in Ntau+ cases was 95% while that in NNfL+ cases was 65%. Reflecting Aβ deposition and neurodegeneration more accurately, we recommend the use of AT(N) classification defined by cerebrospinal fluid AAβ-ratioTNNfL in clinical practice.
Collapse
|
21
|
Al-Kuraishy HM, Al-Gareeb AI, Alsayegh AA, Hakami ZH, Khamjan NA, Saad HM, Batiha GES, De Waard M. A Potential Link Between Visceral Obesity and Risk of Alzheimer's Disease. Neurochem Res 2023; 48:745-766. [PMID: 36409447 DOI: 10.1007/s11064-022-03817-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia characterized by the deposition of amyloid beta (Aβ) plaques and tau-neurofibrillary tangles in the brain. Visceral obesity (VO) is usually associated with low-grade inflammation due to higher expression of pro-inflammatory cytokines by adipose tissue. The objective of the present review was to evaluate the potential link between VO and the development of AD. Tissue hypoxia in obesity promotes tissue injury, production of adipocytokines, and release of pro-inflammatory cytokines leading to an oxidative-inflammatory loop with induction of insulin resistance. Importantly, brain insulin signaling is involved in the pathogenesis of AD and lower cognitive function. Obesity and enlargement of visceral adipose tissue are associated with the deposition of Aβ. All of this is consonant with VO increasing the risk of AD through the dysregulation of adipocytokines which affect the development of AD. The activated nuclear factor kappa B (NF-κB) pathway in VO might be a potential link in the development of AD. Likewise, the higher concentration of advanced glycation end-products in VO could be implicated in the pathogenesis of AD. Taken together, different inflammatory signaling pathways are activated in VO that all have a negative impact on the cognitive function and progression of AD except hypoxia-inducible factor 1 which has beneficial and neuroprotective effects in mitigating the progression of AD. In addition, VO-mediated hypoadiponectinemia and leptin resistance may promote the progression of Aβ formation and tau phosphorylation with the development of AD. In conclusion, VO-induced AD is mainly mediated through the induction of oxidative stress, inflammatory changes, leptin resistance, and hypoadiponectinemia that collectively trigger Aβ formation and neuroinflammation. Thus, early recognition of VO by visceral adiposity index with appropriate management could be a preventive measure against the development of AD in patients with VO.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Abdulrahman A Alsayegh
- Clinical Nutrition Department, Applied Medical Sciences College, Jazan University, Jazan, 82817, Saudi Arabia
| | - Zaki H Hakami
- Medical Laboratory Technology Department Applied Medical Sciences College, Jazan University, Jazan, 82817, Saudi Arabia
| | - Nizar A Khamjan
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt.
| | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, 38120, Saint-Egrève, France.,L'institut du thorax, INSERM, CNRS, UNIV NANTES, 44007, Nantes, France.,LabEx «Ion Channels, Science & Therapeutics», Université de Nice Sophia-Antipolis, 06560, Valbonne, France
| |
Collapse
|
22
|
Gustavsson A, Norton N, Fast T, Frölich L, Georges J, Holzapfel D, Kirabali T, Krolak-Salmon P, Rossini PM, Ferretti MT, Lanman L, Chadha AS, van der Flier WM. Global estimates on the number of persons across the Alzheimer's disease continuum. Alzheimers Dement 2023; 19:658-670. [PMID: 35652476 DOI: 10.1002/alz.12694] [Citation(s) in RCA: 327] [Impact Index Per Article: 163.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/15/2022] [Accepted: 04/04/2022] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Global estimates on numbers of persons in early stages of Alzheimer's disease (AD), including prodromal and preclinical, are lacking, yet are needed to inform policy decisions on preventive measures and planning for future therapies targeting AD pathology. METHODS We synthesized the literature on prevalence across the AD continuum and derived a model estimating the number of persons, stratified by 5-year age groups, sex, and disease stage (AD dementia, prodromal AD, and preclinical AD). RESULTS The global number of persons with AD dementia, prodromal AD, and preclinical AD were estimated at 32, 69, and 315 million, respectively. Together they constituted 416 million across the AD continuum, or 22% of all persons aged 50 and above. DISCUSSION Considering predementia stages, the number of persons with AD is much larger than conveyed in available literature. Our estimates are uncertain, especially for predementia stages in low- and middle-income regions where biomarker studies are missing.
Collapse
Affiliation(s)
- Anders Gustavsson
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | | | | | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | | | - Drew Holzapfel
- CEO Initiative on Alzheimer's Disease, Philadelphia, USA
| | | | - Pierre Krolak-Salmon
- Lyon Institute for Aging, Clinical & Research Memory Center of Lyon, Hospices Civils de Lyon, University of Lyon, Lyon, France
| | - Paolo M Rossini
- Faculty of Medicine of the Catholic University of the Sacred Heart, Department of Neurosci & Neurorehab IRCCS San Raffaele-Rome, Rome, Italy
| | | | | | | | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Department of Epidemiology and Data Science, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
23
|
Gellersen HM, Trelle AN, Farrar BG, Coughlan G, Korkki SM, Henson RN, Simons JS. Medial temporal lobe structure, mnemonic and perceptual discrimination in healthy older adults and those at risk for mild cognitive impairment. Neurobiol Aging 2023; 122:88-106. [PMID: 36516558 DOI: 10.1016/j.neurobiolaging.2022.11.004] [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: 05/13/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/10/2022]
Abstract
Cognitive tests sensitive to the integrity of the medial temporal lobe (MTL), such as mnemonic discrimination of perceptually similar stimuli, may be useful early markers of risk for cognitive decline in older populations. Perceptual discrimination of stimuli with overlapping features also relies on MTL but remains relatively unexplored in this context. We assessed mnemonic discrimination in two test formats (Forced Choice, Yes/No) and perceptual discrimination of objects and scenes in 111 community-dwelling older adults at different risk status for cognitive impairment based on neuropsychological screening. We also investigated associations between performance and MTL sub-region volume and thickness. The at-risk group exhibited reduced entorhinal thickness and impaired perceptual and mnemonic discrimination. Perceptual discrimination impairment partially explained group differences in mnemonic discrimination and correlated with entorhinal thickness. Executive dysfunction accounted for Yes/No deficits in at-risk adults, demonstrating the importance of test format for the interpretation of memory decline. These results suggest that perceptual discrimination tasks may be useful tools for detecting incipient cognitive impairment related to reduced MTL integrity in nonclinical populations.
Collapse
Affiliation(s)
- Helena M Gellersen
- Department of Psychology, University of Cambridge, Cambridge, UK; German Center for Neurodegenerative Diseases, Magdeburg, Germany
| | | | | | - Gillian Coughlan
- Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Saana M Korkki
- Aging Research Center, Karolinska Institute and Stockholm University, Stockholm, Sweden
| | - Richard N Henson
- MRC Cognition and Brain Sciences Unit and Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Jon S Simons
- Department of Psychology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
24
|
Peters-Founshtein G, Gazit L, Naveh T, Domachevsky L, Korczyn A, Bernstine H, Groshar D, Marshall GA, Arzy S. Lost in space(s): multimodal neuroimaging of disorientation along the Alzheimer's disease continuum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525587. [PMID: 36747783 PMCID: PMC9900945 DOI: 10.1101/2023.01.25.525587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Orientation is a fundamental cognitive faculty, allowing the behaving self to link his/her current state to their internal representations of the external world. Once exclusively linked to knowledge of the current place and present time, in recent years, the concept of orientation has evolved to include processing of social, temporal, and abstract relations. Concordantly with the growing focus on orientation, spatial disorientation has been increasingly recognized as a hallmark symptom of Alzheimer's disease (AD). However, few studies have sought to explore disorientation along the AD continuum beyond the spatial domain. 51 participants along the AD continuum performed an orientation task in the spatial, temporal and social domains. Under functional magnetic resonance imaging (fMRI), participants determined which of two familiar places/events/people is geographically/ chronologically/ socially closer to them, respectively. A series of analyses revealed disorientation along the AD- continuum to follow a three-way association between (1) orientation domain, (2) brain region, and (3) disease stage. Specifically, participants with MCI exhibited impaired spatio-temporal orientation and reduced task-evoked activity in temporoparietal regions, while participants with AD dementia exhibited impaired social orientation and reduced task-evoked activity in frontoparietal regions. Furthermore, these patterns of hypoactivation coincided with Default Mode Network (DMN) sub-networks, with spatio-temporal orientation activation overlapping DMN-C and social orientation with DMN-A. Finally, these patterns of disorientation- associated hypoactivations coincided with patterns of fluorodeoxyglucose (FDG) hypometabolism and cortical atrophy characteristic to AD-dementia. Taken together, our results suggest that AD may constitute a disorder of orientation, characterized by a biphasic process as (1) early spatio-temporal and (2) late social disorientation, concurrently manifesting in task-evoked and neurodegenerative changes in temporoparietal and parieto-frontal brain networks, respectively. We propose that a profile of disorientation across multiple domains offers a unique window into the progression of AD.
Collapse
Affiliation(s)
- Gregory Peters-Founshtein
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Nuclear Medicine, Sheba Medical Center, Ramat-Gan, Israel
| | - Lidor Gazit
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| | - Tahel Naveh
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| | - Liran Domachevsky
- Department of Nuclear Medicine, Sheba Medical Center, Ramat-Gan, Israel
- Department of Nuclear Medicine, Assuta Medical Center, Tel-Aviv, Israel
| | - Amos Korczyn
- Department of Neurology, Tel-Aviv University, Tel-Aviv, Israel
| | - Hanna Bernstine
- Department of Imaging, Tel-Aviv University, Tel-Aviv, Israel
- Department of Nuclear Medicine, Assuta Medical Center, Tel-Aviv, Israel
- Department of Nuclear Medicine, Rabin Medical Center, Petah Tikva, Israel
| | - David Groshar
- Department of Imaging, Tel-Aviv University, Tel-Aviv, Israel
- Department of Nuclear Medicine, Assuta Medical Center, Tel-Aviv, Israel
| | - Gad A. Marshall
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Shahar Arzy
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| |
Collapse
|
25
|
Rådestig MA, Skoog J, Zetterberg H, Skillbäck T, Zettergren A, Sterner TR, Fässberg MM, Sacuiu S, Waern M, Wetterberg H, Blennow K, Skoog I, Kern S. Subtle Differences in Cognition in 70-Year-Olds with Elevated Cerebrospinal Fluid Neurofilament Light and Neurogranin: A H70 Cross-Sectional Study. J Alzheimers Dis 2023; 91:291-303. [PMID: 36617786 PMCID: PMC9881027 DOI: 10.3233/jad-220452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Most research on cerebrospinal fluid (CSF) neurofilament light protein (NfL) as a marker for neurodegeneration and neurogranin (Ng) for synaptic dysfunction has largely focused on clinical cohorts rather than population-based samples. OBJECTIVE We hypothesized that increased CSF levels of NfL and Ng are associated with subtle cognitive deficits in cognitively unimpaired (CU) older adults. METHODS The sample was derived from the Gothenburg H70 Birth Cohort Studies and comprised 258 CU 70-year-olds, with a Clinical Dementia Rating score of zero. All participants underwent extensive cognitive testing. CSF levels of NfL and Ng, as well as amyloid β1 - 42, total tau, and phosphorylated tau, were measured. RESULTS Participants with high CSF NfL performed worse in one memory-based test (Immediate recall, p = 0.013) and a language test (FAS, p = 0.016). Individuals with high CSF Ng performed worse on the memory-based test Supra Span (p = 0.035). When stratified according to CSF tau and Aβ42 concentrations, participants with high NfL and increased tau performed worse on a memory test than participants normal tau concentrations (Delayed recall, p = 0.003). In participants with high NfL, those with pathologic Aβ42 concentrations performed worse on the Delayed recall memory (p = 0.044). In the high Ng group, participants with pathological Aβ42 concentrations had lower MMSE scores (p = 0.027). However, in regression analysis we found no linear correlations between CSF NfL or CSF Ng in relation to cognitive tests when controlled for important co-variates. CONCLUSION Markers of neurodegeneration and synaptic pathology might be associated with subtle signs of cognitive decline in a population-based sample of 70-year-olds.
Collapse
Affiliation(s)
- Maya Arvidsson Rådestig
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Johan Skoog
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychology, University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UCL Institute of Neurology, Queen Square, London, UK
- The UK Dementia Research Institute, UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Tobias Skillbäck
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Anna Zettergren
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Therese Rydberg Sterner
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Madeleine Mellqvist Fässberg
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Simona Sacuiu
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Memory Disorders Clinic, Theme Inflammation and Aging, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society (NVS), Clinical Geriatric, Karolinska Institute, Stockholm, Sweden
| | - Margda Waern
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychosis Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Hanna Wetterberg
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ingmar Skoog
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Silke Kern
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| |
Collapse
|
26
|
van den Elzen EHT, Brehmer Y, Van Deun K, Mark RE. Stimulus material selection for the Dutch famous faces test for older adults. Front Med (Lausanne) 2023; 10:1124986. [PMID: 37122325 PMCID: PMC10140445 DOI: 10.3389/fmed.2023.1124986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
Worldwide, approximately 22% of all individuals aged 50 years and older are currently estimated to fall somewhere on the Alzheimer's disease (AD) continuum, which can be roughly divided into preclinical AD, mild cognitive impairment (MCI), and AD dementia. While episodic memory loss (among other aspects) is typically required for a diagnosis of AD dementia, MCI is said to have occurred when cognitive impairment (including memory loss) is worse than expected for the person's age but not enough to be classified as dementia. On the other hand, preclinical AD can currently only be detected using biomarkers; clinical symptoms are not apparent using traditional neuropsychological tests. The main aim of the current paper was to explore the possibility of a test which could distinguish preclinical AD from normal aging. Recent scientific evidence suggests that the Famous Faces Test (FFT) could differentiate preclinical AD from normal aging up to 5 years before a clinical AD diagnosis. Problematic with existing FFTs is the selection of stimulus material. Faces famous in a specific country and a specific decade might not be equally famous for individuals in another country or indeed for people of different ages. The current article describes how famous faces were systematically selected and chosen for the Dutch older (60+) population using five steps. The goal was to design and develop short versions of the FFT for Dutch older adults of equivalent mean difficulty. In future work, these nine parallel versions will be necessary for (a) cross-sectional comparison as well as subsequent longitudinal assessment of cognitively normal and clinical groups and (b) creating personalized norms for the normal aged controls that could be used to compare performance within individuals with clinical diagnoses. The field needs a simple, cognitive test which can distinguish the earliest stages of the dementia continuum from normal aging.
Collapse
Affiliation(s)
- Evi H. T. van den Elzen
- Department of Developmental Psychology, Tilburg University, Tilburg, Netherlands
- Department of Cognitive Neuropsychology, Tilburg University, Tilburg, Netherlands
| | - Yvonne Brehmer
- Department of Developmental Psychology, Tilburg University, Tilburg, Netherlands
- Aging Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Katrijn Van Deun
- Department of Methodology and Statistics, Tilburg University, Tilburg, Netherlands
| | - Ruth E. Mark
- Department of Cognitive Neuropsychology, Tilburg University, Tilburg, Netherlands
- *Correspondence: Ruth E. Mark,
| |
Collapse
|
27
|
Pascoal TA, Leuzy A, Therriault J, Chamoun M, Lussier F, Tissot C, Strandberg O, Palmqvist S, Stomrud E, Ferreira PCL, Ferrari‐Souza JP, Smith R, Benedet AL, Gauthier S, Hansson O, Rosa‐Neto P. Discriminative accuracy of the A/T/N scheme to identify cognitive impairment due to Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12390. [PMID: 36733847 PMCID: PMC9886860 DOI: 10.1002/dad2.12390] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/09/2022] [Accepted: 11/29/2022] [Indexed: 02/03/2023]
Abstract
Introduction The optimal combination of amyloid-β/tau/neurodegeneration (A/T/N) biomarker profiles for the diagnosis of Alzheimer's disease (AD) dementia is unclear. Methods We examined the discriminative accuracy of A/T/N combinations assessed with neuroimaging biomarkers for the differentiation of AD from cognitively unimpaired (CU) elderly and non-AD neurodegenerative diseases in the TRIAD, BioFINDER-1 and BioFINDER-2 cohorts (total n = 832) using area under the receiver operating characteristic curves (AUC). Results For the diagnosis of AD dementia (vs. CU elderly), T biomarkers performed as well as the complete A/T/N system (AUC range: 0.90-0.99). A and T biomarkers in isolation performed as well as the complete A/T/N system in differentiating AD dementia from non-AD neurodegenerative diseases (AUC range; A biomarker: 0.84-1; T biomarker: 0.83-1). Discussion In diagnostic settings, the use of A or T neuroimaging biomarkers alone can reduce patient burden and medical costs compared with using their combination, without significantly compromising accuracy.
Collapse
Affiliation(s)
- Tharick A. Pascoal
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurologySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Translational Neuroimaging LaboratoryThe McGill University Research Centre for Studies in AgingDepartment of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuébecCanada
| | - Antoine Leuzy
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
| | - Joseph Therriault
- Translational Neuroimaging LaboratoryThe McGill University Research Centre for Studies in AgingDepartment of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuébecCanada
- Montreal Neurological InstituteMcGill UniversityMontrealQuébecCanada
| | - Mira Chamoun
- Translational Neuroimaging LaboratoryThe McGill University Research Centre for Studies in AgingDepartment of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuébecCanada
- Montreal Neurological InstituteMcGill UniversityMontrealQuébecCanada
| | - Firoza Lussier
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Translational Neuroimaging LaboratoryThe McGill University Research Centre for Studies in AgingDepartment of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuébecCanada
| | - Cecile Tissot
- Department of NeurologySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
| | - Olof Strandberg
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Sebastian Palmqvist
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Erik Stomrud
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Pamela C. L. Ferreira
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - João Pedro Ferrari‐Souza
- Department of PsychiatrySchool of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Graduate Program in Biological Sciences: BiochemistryUniversidade Federal do Rio Grande do SulPorto AlegreRSBrazil
| | - Ruben Smith
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Andrea Lessa Benedet
- Translational Neuroimaging LaboratoryThe McGill University Research Centre for Studies in AgingDepartment of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuébecCanada
- Montreal Neurological InstituteMcGill UniversityMontrealQuébecCanada
| | - Serge Gauthier
- Translational Neuroimaging LaboratoryThe McGill University Research Centre for Studies in AgingDepartment of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuébecCanada
| | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityLundSweden
- Memory ClinicSkåne University HospitalLundSweden
| | - Pedro Rosa‐Neto
- Translational Neuroimaging LaboratoryThe McGill University Research Centre for Studies in AgingDepartment of Neurology and NeurosurgeryFaculty of MedicineMcGill UniversityMontrealQuébecCanada
- Montreal Neurological InstituteMcGill UniversityMontrealQuébecCanada
| |
Collapse
|
28
|
Preclinical Alzheimer's dementia: a useful concept or another dead end? Eur J Ageing 2022; 19:997-1004. [PMID: 36692779 PMCID: PMC9729660 DOI: 10.1007/s10433-022-00735-w] [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] [Accepted: 10/10/2022] [Indexed: 02/01/2023] Open
Abstract
The term, preclinical dementia, was introduced in 2011 when new guidelines for the diagnosis of Alzheimer's dementia (AD) were published. In the intervening 11 years, many studies have appeared in the literature focusing on this early stage. A search conducted in English on Google Scholar on 06.23.2022 using the term "preclinical (Alzheimer's) dementia" produced 121, 000 results. However, the label is arguably more relevant for research purposes, and it is possible that the knowledge gained may lead to a cure for AD. The term has not been widely adopted by clinical practitioners. Furthermore, it is still not possible to predict who, after a diagnosis of preclinical dementia, will go on to develop AD, and if so, what the risk factors (modifiable and non-modifiable) might be. This Review/Theoretical article will focus on preclinical Alzheimer's dementia (hereafter called preclinical AD). We outline how preclinical AD is currently defined, explain how it is diagnosed and explore why this is problematic at a number of different levels. We also ask the question: Is the concept 'preclinical AD' useful in clinical practice or is it just another dead end in the Holy Grail to find a treatment for AD? Specific recommendations for research and clinical practice are provided.
Collapse
|
29
|
Statins Use in Alzheimer Disease: Bane or Boon from Frantic Search and Narrative Review. Brain Sci 2022; 12:brainsci12101290. [PMID: 36291224 PMCID: PMC9599431 DOI: 10.3390/brainsci12101290] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
Abstract
Alzheimer’s disease (AD) was used to describe pre-senile dementia to differentiate it from senile dementia, which develops in the adult age group of more than 65 years. AD is characterized by the deposition of amyloid beta (Aβ) plaque and tau-neurofibrillary tangles (TNTs) in the brain. The neuropathological changes in AD are related to the deposition of amyloid plaques, neurofibrillary tangles, and progression of neuroinflammation, neuronal mitochondrial dysfunction, autophagy dysfunction, and cholinergic synaptic dysfunction. Statins are one of the main cornerstone drugs for the management of cardiovascular disorders regardless of dyslipidemia status. Increasing the use of statins, mainly in the elderly groups for primary and secondary prevention of cardiovascular diseases, may affect their cognitive functions. Extensive and prolonged use of statins may affect cognitive functions in healthy subjects and dementia patients. Statins-induced cognitive impairments in both patients and health providers had been reported according to the post-marketing survey. This survey depends mainly on sporadic cases, and no cognitive measures were used. Evidence from prospective and observational studies gives no robust conclusion regarding the beneficial or detrimental effects of statins on cognitive functions in AD patients. Therefore, this study is a narrative review aimed with evidences to the beneficial, detrimental, and neutral effects of statins on AD.
Collapse
|
30
|
Kasuga K, Kikuchi M, Tsukie T, Suzuki K, Ihara R, Iwata A, Hara N, Miyashita A, Kuwano R, Iwatsubo T, Ikeuchi T. Different AT(N) profiles and clinical progression classified by two different N markers using total tau and neurofilament light chain in cerebrospinal fluid. BMJ Neurol Open 2022; 4:e000321. [PMID: 36046332 PMCID: PMC9379489 DOI: 10.1136/bmjno-2022-000321] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
Background The AT(N) classification was proposed for categorising individuals according to biomarkers. However, AT(N) profiles may vary depending on the markers chosen and the target population. Methods We stratified 177 individuals who participated in the Japanese Alzheimer's Disease Neuroimaging Initiative by AT(N) classification according to cerebrospinal fluid (CSF) biomarkers. We compared the frequency of AT(N) profiles between the classification using total tau and neurofilament light chain (NfL) as N markers (AT(N)tau and AT(N)NfL). Baseline characteristics, and longitudinal biological and clinical changes were examined between AT(N) profiles. Results We found that 9% of cognitively unimpaired subjects, 49% of subjects with mild cognitive impairment, and 61% of patients with Alzheimer's disease (AD) dementia had the biological AD profile (ie, A+T+) in the cohort. The frequency of AT(N) profiles substantially differed between the AT(N)tau and AT(N)NfL classifications. When we used t-tau as the N marker (AT(N)tau), those who had T- were more frequently assigned to (N)-, whereas those who had T+were more frequently assigned to (N)+ than when we used NfL as the N marker (AT(N)NfL). During a follow-up, the AD continuum group progressed clinically and biologically compared with the normal biomarker group in both the AT(N)tau and AT(N)NfL classifications. More frequent conversion to dementia was observed in the non-AD pathological change group in the AT(N)tau classification, but not in the AT(N)NfL classification. Conclusions AT(N)tau and AT(N)NfL in CSF may capture different aspects of neurodegeneration and provide a different prognostic value. The AT(N) classification aids in understanding the AD continuum biology in various populations.
Collapse
Affiliation(s)
- Kensaku Kasuga
- Molecular Genetics, Niigata University Brain Research Institute, Niigata, Japan
| | - Masataka Kikuchi
- Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Tamao Tsukie
- Molecular Genetics, Niigata University Brain Research Institute, Niigata, Japan
| | - Kazushi Suzuki
- Neurology, National Defense Medical College, Tokorozawa, Japan
| | - Ryoko Ihara
- Neurology, Tokyo Metropolitan Geriatric Medical Center Hospital, Tokyo, Japan
| | - Atsushi Iwata
- Neurology, Tokyo Metropolitan Geriatric Medical Center Hospital, Tokyo, Japan
| | - Norikazu Hara
- Molecular Genetics, Niigata University Brain Research Institute, Niigata, Japan
| | - Akinori Miyashita
- Molecular Genetics, Niigata University Brain Research Institute, Niigata, Japan
| | | | - Takeshi Iwatsubo
- Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Ikeuchi
- Molecular Genetics, Niigata University Brain Research Institute, Niigata, Japan
| | | |
Collapse
|
31
|
Skillbäck T, Blennow K, Zetterberg H, Skoog J, Rydén L, Wetterberg H, Guo X, Sacuiu S, Mielke MM, Zettergren A, Skoog I, Kern S. Slowing gait speed precedes cognitive decline by several years. Alzheimers Dement 2022; 18:1667-1676. [PMID: 35142034 PMCID: PMC9514316 DOI: 10.1002/alz.12537] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 08/26/2021] [Accepted: 10/25/2021] [Indexed: 01/24/2023]
Abstract
INTRODUCTION In this longitudinal study, we aimed to examine if slowing gait speed preceded cognitive decline and correlated with brain amyloidosis. METHODS The sample (n = 287) was derived from the Gothenburg H70 Birth Cohort Studies, with follow-ups between 2000 and 2015. Gait speed was measured by indoor walk, and cognition using the Clinical Dementia Rating (CDR) score. All participants had CDR = 0 at baseline. Some participants had data on cerebrospinal fluid (CSF) amyloid beta (Aβ)1-42 concentrations at the 2009 examination. RESULTS Gait speed for participants who worsened in CDR score during follow-up was slower at most examinations. Baseline gait speed could significantly predict CDR change from baseline to follow-up. Subjects with pathological CSF Aβ1- 42 concentrations at the 2009 visit had lost more gait speed compared to previous examinations. DISCUSSION Our results indicate that gait speed decline precedes cognitive decline, is linked to Alzheimer's pathology, and might be used for early detection of increased risk for dementia development.
Collapse
Affiliation(s)
- Tobias Skillbäck
- Institute of Neuroscience and Physiology, Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Department of Neuropsychiatric epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgGothenburgSweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- Dementia Research Institute at UCLLondonUK
| | - Johan Skoog
- Department of Neuropsychiatric epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University HospitalRegion Västra GötalandMölndalSweden
| | - Lina Rydén
- Department of Neuropsychiatric epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University HospitalRegion Västra GötalandMölndalSweden
| | - Hanna Wetterberg
- Department of Neuropsychiatric epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgGothenburgSweden
| | - Xinxin Guo
- Department of Neuropsychiatric epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgGothenburgSweden
| | - Simona Sacuiu
- Department of Neuropsychiatric epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University HospitalRegion Västra GötalandMölndalSweden
| | - Michelle M. Mielke
- Department of Health Sciences Research, Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Anna Zettergren
- Institute of Neuroscience and Physiology, Department of Psychiatry and NeurochemistryThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Department of Neuropsychiatric epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgGothenburgSweden
| | - Ingmar Skoog
- Department of Neuropsychiatric epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University HospitalRegion Västra GötalandMölndalSweden
| | - Silke Kern
- Department of Neuropsychiatric epidemiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyCentre for Ageing and Health (AgeCap) at the University of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University HospitalRegion Västra GötalandMölndalSweden
| |
Collapse
|
32
|
Li R, Wang L, Zhang Q, Duan H, Qian D, Yang F, Xia J. Alpiniae oxyphyllae fructus possesses neuroprotective effects on H 2O 2 stimulated PC12 cells via regulation of the PI3K/Akt signaling Pathway. Front Pharmacol 2022; 13:966348. [PMID: 36091821 PMCID: PMC9454318 DOI: 10.3389/fphar.2022.966348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/25/2022] [Indexed: 12/19/2022] Open
Abstract
Backgroud: Alzheimer's disease (AD) is a typical neurodegenerative disease, which occurs in the elderly population. Alpiniae oxyphyllae Fructus (AOF) is a traditional Chinese medicine that has potential therapeutic effect on AD, but the mechanism behind it is unclear. Methods: Firstly, the main chemical components of AOF were identified by LC-MS, while the main active ingredients and targets were screened by TCMSP database. At the same time, AD-related target proteins were obtained using Genecards and OMIM databases. PPI was constructed by cross-linking AOF and AD targets, and GO enrichment analysis and KEGG pathway enrichment analysis were performed to identify the relevant biological processes and signaling pathways. Finally, based on the H2O2-stimulated PC12 cell, flow cytometry, WB and immunofluorescence experiments were performed to verify the protective effect of AOF on AD. Results: We identified 38 active ingredients with 662 non-repetitive targets in AOF, of which 49 were potential therapeutic AD targets of AOF. According to the GO and KEGG analysis, these potential targets are mainly related to oxidative stress and apoptosis. The role of AOF in the treatment of AD is mainly related to the PI3K/AKT signaling pathway. Protocatechuic acid and nootkatone might be the main active ingredients of AOF. In subsequent experiments, the results of CCK-8 showed that AOF mitigated PC12 cell damage induced by H2O2. Kits, flow cytometry, and laser confocal microscopy indicated that AOF could decrease ROS and increase the activity of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-Px), while AOF could also increase mitochondrial membrane potential (MMP), thereby inhibiting apoptosis. Finally, immunofluorescence and WB results showed that AOF inhibited the expression of BAX and caspase-3 in PC12 cells, and promoted the expression of Bcl-2. At the same time, the phosphorylation levels of PI3K and Akt proteins were also significantly increased. Conclusion: This study suggests that AOF had the potential to treat AD by suppressing apoptosis induced by oxidative stress via the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Ruolan Li
- School of Pharmacy, School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lingyu Wang
- School of Pharmacy, School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | - Huxinyue Duan
- School of Pharmacy, School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Die Qian
- School of Pharmacy, School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Xia
- School of Pharmacy, School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
33
|
Weber GE, Khrestian M, Tuason ED, Shao Y, Pillai J, Rao S, Feng H, Zhou Y, Cheng F, DeSilva TM, Stauffer S, Leverenz JB, Bekris LM. Peripheral sTREM2-Related Inflammatory Activity Alterations in Early-Stage Alzheimer's Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2283-2299. [PMID: 35523454 PMCID: PMC9117433 DOI: 10.4049/jimmunol.2100771] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 03/07/2022] [Indexed: 05/17/2023]
Abstract
Alzheimer's disease (AD) has been linked to multiple immune system-related genetic variants. Triggering receptor expressed on myeloid cells 2 (TREM2) genetic variants are risk factors for AD and other neurodegenerative diseases. In addition, soluble TREM2 (sTREM2) isoform is elevated in cerebrospinal fluid in the early stages of AD and is associated with slower cognitive decline in a disease stage-dependent manner. Multiple studies have reported an altered peripheral immune response in AD. However, less is known about the relationship between peripheral sTREM2 and an altered peripheral immune response in AD. The objective of this study was to explore the relationship between human plasma sTREM2 and inflammatory activity in AD. The hypothesis of this exploratory study was that sTREM2-related inflammatory activity differs by AD stage. We observed different patterns of inflammatory activity across AD stages that implicate early-stage alterations in peripheral sTREM2-related inflammatory activity in AD. Notably, fractalkine showed a significant relationship with sTREM2 across different analyses in the control groups that was lost in later AD-related stages with high levels in mild cognitive impairment. Although multiple other inflammatory factors either differed significantly between groups or were significantly correlated with sTREM2 within specific groups, three inflammatory factors (fibroblast growth factor-2, GM-CSF, and IL-1β) are notable because they exhibited both lower levels in AD, compared with mild cognitive impairment, and a change in the relationship with sTREM2. This evidence provides important support to the hypothesis that sTREM2-related inflammatory activity alterations are AD stage specific and provides critical information for therapeutic strategies focused on the immune response.
Collapse
Affiliation(s)
- Grace E Weber
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH
| | | | | | - Yvonne Shao
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Jagan Pillai
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Stephen Rao
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Hao Feng
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Yadi Zhou
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Feixiong Cheng
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Tara M DeSilva
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH; and
| | - Shaun Stauffer
- Center for Therapeutics Discovery, Cleveland Clinic, Cleveland, OH
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Lynn M Bekris
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH;
| |
Collapse
|
34
|
Ivanova O, Meilán JJG, Martínez-Sánchez F, Martínez-Nicolás I, Llorente TE, González NC. Discriminating speech traits of Alzheimer's disease assessed through a corpus of reading task for Spanish language. COMPUT SPEECH LANG 2022. [DOI: 10.1016/j.csl.2021.101341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
Abstract
Alzheimer’s disease (AD) is prevalent throughout the world and is the leading cause of dementia in older individuals (aged ≥ 65 years). To gain a deeper understanding of the recent literature on the epidemiology of AD and its progression, we conducted a review of the PubMed-indexed literature (2014–2021) in North America, Europe, and Asia. The worldwide toll of AD is evidenced by rising prevalence, incidence, and mortality due to AD—estimates which are low because of underdiagnosis of AD. Mild cognitive impairment (MCI) due to AD can ultimately progress to AD dementia; estimates of AD dementia etiology among patients with MCI range from 40% to 75% depending on the populations studied and whether the MCI diagnosis was made clinically or in combination with biomarkers. The risk of AD dementia increases with progression from normal cognition with no amyloid-beta (Aβ) accumulation to early neurodegeneration and subsequently to MCI. For patients with Aβ accumulation and neurodegeneration, lifetime risk of AD dementia has been estimated to be 41.9% among women and 33.6% among men. Data on progression from preclinical AD to MCI are sparse, but an analysis of progression across the three preclinical National Institute on Aging and Alzheimer’s Association (NIA-AA) stages suggests that NIA-AA stage 3 (subtle cognitive decline with AD biomarker positivity) could be useful in combination with other tools for treatment decision-making. Factors shown to increase risk include lower Mini-Mental State Examination (MMSE) score, higher Alzheimer’s Disease Assessment Scale (ADAS-cog) score, positive APOE4 status, white matter hyperintensities volume, entorhinal cortex atrophy, cerebrospinal fluid (CSF) total tau, CSF neurogranin levels, dependency in instrumental activities of daily living (IADL), and being female. Results suggest that use of biomarkers alongside neurocognitive tests will become an important part of clinical practice as new disease-modifying therapies are introduced.
Collapse
|
36
|
Dittrich A, Ashton NJ, Zetterberg H, Blennow K, Simrén J, Geiger F, Zettergren A, Shams S, Machado A, Westman E, Schöll M, Skoog I, Kern S. Plasma and CSF NfL are differentially associated with biomarker evidence of neurodegeneration in a community-based sample of 70-year-olds. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12295. [PMID: 35280965 PMCID: PMC8897823 DOI: 10.1002/dad2.12295] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 12/13/2022]
Abstract
Neurofilament light protein (NfL) in cerebrospinal fluid (CSF) and plasma (P) are suggested to be interchangeable markers of neurodegeneration. However, evidence is scarce from community-based samples. NfL was examined in a small-scale sample of 287 individuals from the Gothenburg H70 Birth cohort 1944 study, using linear models in relation to CSF and magnetic resonance imaging (MRI) biomarker evidence of neurodegeneration. CSF-NfL and P-NfL present distinct associations with biomarker evidence of Alzheimer's disease (AD) pathology and neurodegeneration. P-NfL was associated with several markers that are characteristic of AD, including smaller hippocampal volumes, amyloid beta (Aβ)42, Aβ42/40, and Aβ42/t-tau (total tau). CSF-NfL demonstrated associations with measures of synaptic and neurodegeneration, including t-tau, phosphorylated tau (p-tau), and neurogranin. Our findings suggest that P-NfL and CSF-NfL may exert different effects on markers of neurodegeneration in a small-scale community-based sample of 70-year-olds.
Collapse
Affiliation(s)
- Anna Dittrich
- Department of Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University HospitalMölndalSweden
| | - Nicholas J. Ashton
- Department of Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Center of Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- King's College London, Institute of Psychiatry, Psychology and NeuroscienceMaurice Wohl Institute Clinical Neuroscience InstituteLondonUK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS FoundationLondonUK
| | - Henrik Zetterberg
- Department of Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- UK Dementia Research Institute at UCLUCL Institute of NeurologyLondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | - Kaj Blennow
- Department of Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Joel Simrén
- Department of Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Fiona Geiger
- Department of Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Anna Zettergren
- Department of Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Sara Shams
- Department of Clinical NeuroscienceKarolinska University HospitalStockholmSweden
- Care Sciences and Society, Karolinska Institutet, and Department of RadiologyKarolinska University HospitalStockholmSweden
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| | - Alejandra Machado
- Division of Clinical GeriatricsDepartment of NeurobiologyKarolinska University HospitalStockholmSweden
| | - Eric Westman
- Division of Clinical GeriatricsDepartment of NeurobiologyKarolinska University HospitalStockholmSweden
| | - Michael Schöll
- Department of Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Center of Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | - Ingmar Skoog
- Department of Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University HospitalMölndalSweden
| | - Silke Kern
- Department of Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Psychiatry Cognition and Old Age PsychiatrySahlgrenska University HospitalMölndalSweden
| |
Collapse
|
37
|
Impairment of the autophagy-lysosomal pathway in Alzheimer's diseases: Pathogenic mechanisms and therapeutic potential. Acta Pharm Sin B 2022; 12:1019-1040. [PMID: 35530153 PMCID: PMC9069408 DOI: 10.1016/j.apsb.2022.01.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/09/2021] [Accepted: 12/16/2021] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disorder, is characterized by memory loss and cognitive dysfunction. The accumulation of misfolded protein aggregates including amyloid beta (Aβ) peptides and microtubule associated protein tau (MAPT/tau) in neuronal cells are hallmarks of AD. So far, the exact underlying mechanisms for the aetiologies of AD have not been fully understood and the effective treatment for AD is limited. Autophagy is an evolutionarily conserved cellular catabolic process by which damaged cellular organelles and protein aggregates are degraded via lysosomes. Recently, there is accumulating evidence linking the impairment of the autophagy–lysosomal pathway with AD pathogenesis. Interestingly, the enhancement of autophagy to remove protein aggregates has been proposed as a promising therapeutic strategy for AD. Here, we first summarize the recent genetic, pathological and experimental studies regarding the impairment of the autophagy–lysosomal pathway in AD. We then describe the interplay between the autophagy–lysosomal pathway and two pathological proteins, Aβ and MAPT/tau, in AD. Finally, we discuss potential therapeutic strategies and small molecules that target the autophagy–lysosomal pathway for AD treatment both in animal models and in clinical trials. Overall, this article highlights the pivotal functions of the autophagy–lysosomal pathway in AD pathogenesis and potential druggable targets in the autophagy–lysosomal pathway for AD treatment.
Collapse
|
38
|
Fatima T, Jacobsson LT, Kern S, Zettergren A, Blennow K, Zetterberg H, Johansson L, Dehlin M, Skoog I. Association between serum urate and CSF markers of Alzheimer's disease pathology in a population-based sample of 70-year-olds. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12241. [PMID: 34934798 PMCID: PMC8652407 DOI: 10.1002/dad2.12241] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/09/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The relationship between urate and biomarkers for Alzheimer's disease (AD) pathophysiology has not been investigated. METHODS We examined whether serum concentration of urate was associated with cerebrospinal fluid biomarkers, amyloid beta (Aβ)42, Aβ40, phosphorylated tau (p-tau), total tau (t-tau), neurofilament light (NfL), and Aβ42/Aβ40 ratio, in cognitively unimpaired 70-year-old individuals from Gothenburg, Sweden. We also evaluated whether possible associations were modulated by the apolipoprotein E (APOE) ε4 allele. RESULTS Serum urate was positively associated with Aβ42 in males (β = 0.55 pg/mL, P = .04). There was a positive urate-APOE ε4 interaction (1.24 pg/mL, P interaction = .02) in relation to Aβ42 association. The positive urate and Aβ42 association strengthened in male APOE ε4 carriers (β = 1.28 pg/mL, P = .01). DISCUSSION The positive association between urate and Aβ42 in cognitively healthy men may suggest a protective effect of urate against deposition of amyloid protein in the brain parenchyma, and in the longer term, maybe against AD dementia.
Collapse
Affiliation(s)
- Tahzeeb Fatima
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Clinical SciencesLundSection of RheumatologyLund UniversityLundSweden
| | - Lennart T.H. Jacobsson
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Silke Kern
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Anna Zettergren
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- UK Dementia Research Institute at UCLLondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | - Lena Johansson
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Mats Dehlin
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Ingmar Skoog
- Department of Psychiatry and Neurochemistry at Institute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
39
|
Lu K, Nicholas JM, Pertzov Y, Grogan J, Husain M, Pavisic IM, James SN, Parker TD, Lane CA, Keshavan A, Keuss SE, Buchanan SM, Murray-Smith H, Cash DM, Malone IB, Sudre CH, Coath W, Wong A, Henley SM, Fox NC, Richards M, Schott JM, Crutch SJ. Dissociable effects of APOE-ε4 and β-amyloid pathology on visual working memory. NATURE AGING 2021; 1:1002-1009. [PMID: 34806027 PMCID: PMC7612005 DOI: 10.1038/s43587-021-00117-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 08/17/2021] [Indexed: 01/21/2023]
Abstract
Although APOE-ε4 carriers are at significantly higher risk of developing Alzheimer's disease than non-carriers1, controversial evidence suggests that APOE-ε4 might confer some advantages, explaining the survival of this gene (antagonistic pleiotropy)2,3. In a population-based cohort born in one week in 1946 (assessed aged 69-71), we assessed differential effects of APOE-ε4 and β-amyloid pathology (quantified using 18F-Florbetapir-PET) on visual working memory (object-location binding). In 398 cognitively normal participants, APOE-ε4 and β-amyloid had opposing effects on object identification, predicting better and poorer recall respectively. ε4-carriers also recalled locations more precisely, with a greater advantage at higher β-amyloid burden. These results provide evidence of superior visual working memory in ε4-carriers, showing that some benefits of this genotype are demonstrable in older age, even in the preclinical stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Kirsty Lu
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Jennifer M. Nicholas
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Yoni Pertzov
- Department of Psychology, The Hebrew University of Jerusalem, Israel
| | - John Grogan
- Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Masud Husain
- Nuffield Department of Clinical Neurosciences, University of Oxford, UK
- Department of Experimental Psychology, University of Oxford, UK
| | - Ivanna M. Pavisic
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - Sarah-Naomi James
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - Thomas D. Parker
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Christopher A. Lane
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Ashvini Keshavan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sarah E. Keuss
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sarah M. Buchanan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Heidi Murray-Smith
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - David M. Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Ian B. Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Carole H. Sudre
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - William Coath
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - Susie M.D. Henley
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Nick C. Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Marcus Richards
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - Jonathan M. Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sebastian J. Crutch
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
40
|
Jayatunga DPW, Hone E, Khaira H, Lunelli T, Singh H, Guillemin GJ, Fernando B, Garg ML, Verdile G, Martins RN. Therapeutic Potential of Mitophagy-Inducing Microflora Metabolite, Urolithin A for Alzheimer's Disease. Nutrients 2021; 13:nu13113744. [PMID: 34836000 PMCID: PMC8617978 DOI: 10.3390/nu13113744] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/28/2021] [Accepted: 10/12/2021] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial dysfunction including deficits of mitophagy is seen in aging and neurodegenerative disorders including Alzheimer’s disease (AD). Apart from traditionally targeting amyloid beta (Aβ), the main culprit in AD brains, other approaches include investigating impaired mitochondrial pathways for potential therapeutic benefits against AD. Thus, a future therapy for AD may focus on novel candidates that enhance optimal mitochondrial integrity and turnover. Bioactive food components, known as nutraceuticals, may serve as such agents to combat AD. Urolithin A is an intestinal microbe-derived metabolite of a class of polyphenols, ellagitannins (ETs). Urolithin A is known to exert many health benefits. Its antioxidant, anti-inflammatory, anti-atherogenic, anti-Aβ, and pro-mitophagy properties are increasingly recognized. However, the underlying mechanisms of urolithin A in inducing mitophagy is poorly understood. This review discusses the mitophagy deficits in AD and examines potential molecular mechanisms of its activation. Moreover, the current knowledge of urolithin A is discussed, focusing on its neuroprotective properties and its potential to induce mitophagy. Specifically, this review proposes potential mechanisms by which urolithin A may activate and promote mitophagy.
Collapse
Affiliation(s)
- Dona Pamoda W. Jayatunga
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA 6027, Australia; (D.P.W.J.); (E.H.); (B.F.); (G.V.)
| | - Eugene Hone
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA 6027, Australia; (D.P.W.J.); (E.H.); (B.F.); (G.V.)
- Cooperative Research Centre for Mental Health, Carlton, VIC 3053, Australia
| | - Harjot Khaira
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand; (H.K.); (T.L.); (H.S.); (M.L.G.)
| | - Taciana Lunelli
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand; (H.K.); (T.L.); (H.S.); (M.L.G.)
| | - Harjinder Singh
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand; (H.K.); (T.L.); (H.S.); (M.L.G.)
| | - Gilles J. Guillemin
- Department of Pharmacology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia;
- St. Vincent’s Centre for Applied Medical Research, Sydney, NSW 2011, Australia
| | - Binosha Fernando
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA 6027, Australia; (D.P.W.J.); (E.H.); (B.F.); (G.V.)
| | - Manohar L. Garg
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand; (H.K.); (T.L.); (H.S.); (M.L.G.)
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Giuseppe Verdile
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA 6027, Australia; (D.P.W.J.); (E.H.); (B.F.); (G.V.)
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Ralph N. Martins
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA 6027, Australia; (D.P.W.J.); (E.H.); (B.F.); (G.V.)
- Australian Alzheimer’s Research Foundation, Ralph and Patricia Sarich Neuroscience Research Institute, 8 Verdun Street., Nedlands, WA 6009, Australia
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Correspondence: ; Tel.: +61-8-9347-4200
| |
Collapse
|
41
|
Diaz-Galvan P, Cedres N, Figueroa N, Barroso J, Westman E, Ferreira D. Cerebrovascular Disease and Depressive Symptomatology in Individuals With Subjective Cognitive Decline: A Community-Based Study. Front Aging Neurosci 2021; 13:656990. [PMID: 34385912 PMCID: PMC8353130 DOI: 10.3389/fnagi.2021.656990] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/28/2021] [Indexed: 12/31/2022] Open
Abstract
Subjective cognitive decline (SCD) may be the first sign of Alzheimer's disease (AD), but it can also reflect other pathologies such as cerebrovascular disease or conditions like depressive symptomatology. The role of depressive symptomatology in SCD is controversial. We investigated the association between depressive symptomatology, cerebrovascular disease, and SCD. We recruited 225 cognitively unimpaired individuals from a prospective community-based study [mean age (SD) = 54.64 (10.18); age range 35-77 years; 55% women; 123 individuals with one or more subjective cognitive complaints, 102 individuals with zero complaints]. SCD was assessed with a scale of 9 memory and non-memory subjective complaints. Depressive symptomatology was assessed with established questionnaires. Cerebrovascular disease was assessed with magnetic resonance imaging markers of white matter signal abnormalities (WMSA) and mean diffusivity (MD). We combined correlation, multiple regression, and mediation analyses to investigate the association between depressive symptomatology, cerebrovascular disease, and SCD. We found that SCD was associated with more cerebrovascular disease, older age, and increased depressive symptomatology. In turn, depressive symptomatology was not associated with cerebrovascular disease. Variability in MD was mediated by WMSA burden, presumably reflecting cerebrovascular disease. We conclude that, in our community-based cohort, depressive symptomatology is associated with SCD but not with cerebrovascular disease. In addition, depressive symptomatology did not influence the association between cerebrovascular disease and SCD. We suggest that therapeutic interventions for depressive symptomatology could alleviate the psychological burden of negative emotions in people with SCD, and intervening on vascular risk factors to reduce cerebrovascular disease should be tested as an opportunity to minimize neurodegeneration in SCD individuals from the community.
Collapse
Affiliation(s)
- Patricia Diaz-Galvan
- Department of Neurobiology, Care Sciences, and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Nira Cedres
- Department of Neurobiology, Care Sciences, and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
| | - Nerea Figueroa
- Department of Clinical Psychology, Psychobiology, and Methodology, Faculty of Psychology, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| | - Jose Barroso
- Department of Clinical Psychology, Psychobiology, and Methodology, Faculty of Psychology, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| | - Eric Westman
- Department of Neurobiology, Care Sciences, and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
- Department of Neuroimaging, Center for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Daniel Ferreira
- Department of Neurobiology, Care Sciences, and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet (KI), Stockholm, Sweden
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
- Department of Clinical Psychology, Psychobiology, and Methodology, Faculty of Psychology, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain
| |
Collapse
|
42
|
Libowitz MR, Wei K, Tran T, Chu K, Moncrieffe K, Harrington MG, King K. Regional brain volumes relate to Alzheimer's disease cerebrospinal fluid biomarkers and neuropsychometry: A cross-sectional, observational study. PLoS One 2021; 16:e0254332. [PMID: 34292973 PMCID: PMC8297871 DOI: 10.1371/journal.pone.0254332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/27/2021] [Indexed: 11/18/2022] Open
Abstract
We hypothesized that automated assessment of brain volumes on MRI can predict presence of cerebrospinal fluid abnormal ß-amyloid42 and Tau protein levels and thus serve as a useful screening test for possible Alzheimer's disease. 113 participants ranging from cognitively healthy to Alzheimer's disease underwent MRI exams to obtain measurements of hippocampus, prefrontal cortex, precuneus, parietal cortex, and occipital lobe volumes. A non-exclusive subset (n = 107) consented to lumbar punctures to obtain cerebrospinal fluid for ß-amyloid42 and Tau protein assessment including cognitively health (n = 75), mild cognitively impaired (n = 22), and Alzheimer's disease (n = 10). After adjustment for false discovery rate, ß-amyloid42 was significantly associated with volumes in the hippocampus (p = 0.043), prefrontal cortex (p = 0.010), precuneus (p = 0.024), and the posterior cingulate (p = 0.002). No association between Tau levels and regional brain volume survived multiple test correction. Secondary analysis was performed to determine associations between MRI brain volumes and CSF protein levels to neuropsychological impairment. A non-exclusive subset (n = 96) including cognitively healthy (n = 72), mild cognitively impaired (n = 21), and Alzheimer's disease (n = 3) participants underwent Stroop Interference and Boston Naming neuropsychological testing. A higher score on the Boston Naming Test was optimally predicted in a selective regression model by greater hippocampus volume (p = 0.002), a higher ratio of ß-amyloid42 to Tau protein levels (p < 0.001), greater posterior cingulate volume (p = 0.0193), age (p = 0.0271), and a higher education level (p = 0.002). A better performance on the Stroop Interference Test was optimally predicted by greater hippocampus volume (p = 0.0003) and a higher education level (p < 0.001). Lastly, impaired cognitive status (mild cognitive impairment and Alzheimer's Disease) was optimally predicted in a selective regression model by a worse performance on the Stroop Interference Test (p < 0.001), a worse performance on the Boston Naming Test (p < 0.001), along with lower prefrontal cortex volume (p = 0.002) and lower hippocampus volume (p = 0.007).
Collapse
Affiliation(s)
- Mark R. Libowitz
- Magnetic Resonance Program, Huntington Medical Research Institutes, Pasadena, California, United States of America
- * E-mail:
| | - Ke Wei
- Magnetic Resonance Program, Huntington Medical Research Institutes, Pasadena, California, United States of America
| | - Thao Tran
- Magnetic Resonance Program, Huntington Medical Research Institutes, Pasadena, California, United States of America
| | - Karen Chu
- Magnetic Resonance Program, Huntington Medical Research Institutes, Pasadena, California, United States of America
| | - Kristina Moncrieffe
- Fuller Graduate School of Psychology, Pasadena, California, United States of America
| | - Michael G. Harrington
- Molecular Neurology Program, Huntington Medical Research Institutes, Pasadena, California, United States of America
| | - Kevin King
- Barrow Neurological Institute, Phoenix, Arizona, United States of America
| |
Collapse
|
43
|
Mikuła E. Recent Advancements in Electrochemical Biosensors for Alzheimer's Disease Biomarkers Detection. Curr Med Chem 2021; 28:4049-4073. [PMID: 33176635 PMCID: PMC8287894 DOI: 10.2174/0929867327666201111141341] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/28/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023]
Abstract
Background It is estimated that the average time between the diagnosis of Alzheimer’s disease (AD) and the patient’s death is 5-9 years. Therefore, both the initial phase of the disease and the preclinical state can be included in the critical period in disease diagnosis. Accordingly, huge progress has recently been observed in biomarker research to identify risk factors for dementia in older people with normal cognitive functions and mild cognitive impairments. Methods Electrochemical biosensors are excellent analytical tools that are used in the detection of AD biomarkers as they are easy to use, portable, and can do analysis in real time. Results This review presents the analytical techniques currently used to determine AD biomarkers in terms of their advantages and disadvantages; the most important clinical biomarkers of AD and their role in the disease. All recently used biorecognition molecules in electrochemical biosensor development, i.e., receptor protein, antibodies, aptamers and nucleic acids, are summarized for the first time. Novel electrochemical biosensors for AD biomarker detection, as ideal analytical platforms for point-of-care diagnostics, are also reviewed. Conclusion The article focuses on various strategies of biosensor chemical surface modifications to immobilize biorecognition molecules, enabling specific, quantitative AD biomarker detection in synthetic and clinical samples. In addition, this is the first review that presents innovative single-platform systems for simultaneous detection of multiple biomarkers and other important AD-associated biological species based on electrochemical techniques. The importance of these platforms in disease diagnosis is discussed.
Collapse
Affiliation(s)
- Edyta Mikuła
- Department of Biosensors, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland
| |
Collapse
|
44
|
Serdyuk OV, Sidenkova AP, Khiliuk DA. Clinical and Dynamic Features and Prognostic Value of Non-Cognitive Psychopathological Symptoms in Mild Cognitive Impairment (MCI). PSIKHIATRIYA 2021; 19:17-28. [DOI: 10.30629/2618-6667-2021-19-2-17-28] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2024]
Abstract
Background: high prevalence of pre-dementia cognitive disorders (in particular mild cognitive impairment, MCI) in older people, unfavorable predictive value of MCI with a high risk of conversion to dementia, lack of a unified view of facultative, noncognitive psychopathological symptoms (NPS) in its syndromic structure, their clinical heterogeneity, close relationships with mild cognitive impairment syndrome is actualized by the analysis of the mutual influence of the components of MCI syndromes.The aim: to investigate clinical and dynamic features and determine the prognostic value of non-cognitive psychopathological symptoms in MCI, including various psychopathological symptoms (affective, psychotic, behavioral disorders).Patients and methods: a comparative dynamic prospective selective observational study of 264 older patients with MCI was carried out. The main study group included 189 patients with NPS; compared group made up 75 cases with MCI represented only by cognitive impairments. Repeated clinical, psychopathological and psychometric assessments (MMSE, MoCa, NPI) were performed at 12 and 24 months. Results: differences were revealed in the structure and dynamics of cognitive indicators in patients with dominance of affective, psychotic or behavioral symptoms. Patients with affective symptoms showed low scores on subtests of line drawing, phonemic fluency, and ability to verbal categorization (p < 0.05). In these individuals, with repeated measurements, the indices of delayed reproduction, retention, abstract thinking were significantly reduced in comparison with the results of the rest of the study participants (p < 0.05). Patients with psychotic symptoms showed the lowest indices of delayed reproduction, spatial-visual, speech tests, false reproductions both at the initial assessment and in the dynamics of observation (p < 0.05). But their parameters of counting, attention, working memory, “recognition” are higher in them than in representatives of other psychopathological subgroups (p < 0.05). It was found that mild cognitive impairment without psychopathological inclusions has a lower tendency to transform into dementia over a two-year follow-up period than MCI, in the structure of which non-cognitive psychopathological symptoms are present.Conclusions: non-cognitive psychopathological symptoms of MCI are heterogeneous in terms of clinical and dynamic characteristics. The structure and dynamics of cognitive indicators is different in the affective, psychotic, behavioral subgroups of patients with MCI. Individuals with affective pathology had the highest rate of MCI conversion to dementia. In patients with MCI without neuropsychiatric symptoms, a favorable course of cognitive impairment was more common than in patients with MCI with non-cognitive psychopathological symptoms.
Collapse
Affiliation(s)
- O. V. Serdyuk
- Sverdlovsk Region “Sverdlovsk Regional Clinical Psychiatric Hospital”
| | - A. P. Sidenkova
- Federal State Budgetary Educational Institution of Higher Education “Ural State Medical University of the Ministry of Health of Russia”
| | - D. A. Khiliuk
- Sverdlovsk Region “Sverdlovsk Regional Clinical Psychiatric Hospital”
| |
Collapse
|
45
|
Bellaver B, Ferrari-Souza JP, Uglione da Ros L, Carter SF, Rodriguez-Vieitez E, Nordberg A, Pellerin L, Rosa-Neto P, Leffa DT, Zimmer ER. Astrocyte Biomarkers in Alzheimer Disease: A Systematic Review and Meta-analysis. Neurology 2021; 96:e2944-e2955. [PMID: 33952650 DOI: 10.1212/wnl.0000000000012109] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/19/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To perform a systematic review and meta-analysis to determine whether fluid and imaging astrocyte biomarkers are altered in Alzheimer disease (AD). METHODS PubMed and Web of Science databases were searched for articles reporting fluid or imaging astrocyte biomarkers in AD. Pooled effect sizes were determined with standardized mean differences (SMDs) using the Hedge G method with random effects to determine biomarker performance. Adapted questions from the Quality Assessment of Diagnostic Accuracy Studies were applied for quality assessment. A protocol for this study has been previously registered in PROSPERO (registration number: CRD42020192304). RESULTS The initial search identified 1,425 articles. After exclusion criteria were applied, 33 articles (a total of 3,204 individuals) measuring levels of glial fibrillary acidic protein (GFAP), S100B, chitinase-3-like protein 1 (YKL-40), and aquaporin 4 in the blood and CSF, as well as monoamine oxidase-B indexed by PET 11C-deuterium-l-deprenyl, were included. GFAP (SMD 0.94, 95% confidence interval [CI] 0.71-1.18) and YKL-40 (SMD 0.76, 95% CI 0.63-0.89) levels in the CSF and S100B levels in the blood (SMD 2.91, 95% CI 1.01-4.8) were found to be significantly increased in patients with AD. CONCLUSIONS Despite significant progress, applications of astrocyte biomarkers in AD remain in their early days. This meta-analysis demonstrated that astrocyte biomarkers are consistently altered in AD and supports further investigation for their inclusion in the AD clinical research framework for observational and interventional studies.
Collapse
Affiliation(s)
- Bruna Bellaver
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - João Pedro Ferrari-Souza
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Lucas Uglione da Ros
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Stephen F Carter
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Elena Rodriguez-Vieitez
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Agneta Nordberg
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Luc Pellerin
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Pedro Rosa-Neto
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Douglas Teixeira Leffa
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil
| | - Eduardo R Zimmer
- From the Graduate Program in Biological Sciences: Biochemistry (B.B., J.P.F.-S., L.U.d.R., E.R.Z.), Department of Pharmacology (E.R.Z.), and Graduate Program in Biological Sciences: Pharmacology and Therapeutics (E.R.Z.), Universidade Federal do Rio Grande do Sul; Department of Psychiatry (S.F.C.), University of Cambridge; Wolfson Molecular Imaging Centre (S.F.C.), University of Manchester, UK; Department of Neurobiology (E.R.-V, A.N.), Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet (E.R.-V, A.N.); Theme Aging (A.N.), Karolinska University Hospital Stockholm, Stockholm, Sweden; Inserm U1082 (L.P.), Université de Poitiers, France; Translational Neuroimaging Laboratory (P.R.-N.), McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; McGill University (P.R.-N.), Montreal, Quebec, Canada; and ADHD Outpatient Program & Development Psychiatry Program (D.T.L.), Hospital de Clínicas de Porto Alegre, Brazil.
| |
Collapse
|
46
|
Samuelsson J, Kern S, Zetterberg H, Blennow K, Rothenberg E, Wallengren O, Skoog I, Zettergren A. A Western-style dietary pattern is associated with cerebrospinal fluid biomarker levels for preclinical Alzheimer's disease-A population-based cross-sectional study among 70-year-olds. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12183. [PMID: 34027029 PMCID: PMC8129853 DOI: 10.1002/trc2.12183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 03/17/2021] [Accepted: 04/20/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Diet may be a modifiable factor for reducing the risk of Alzheimer's disease (AD). Western-style dietary patterns are considered to increase the risk, whereas Mediterranean-style dietary patterns are considered to reduce the risk. An association between diet and AD-related biomarkers have been suggested, but studies are limited. AIM To investigate potential relations between dietary patterns and cerebrospinal fluid (CSF) biomarkers for AD among dementia-free older adults. METHODS Data were derived from the population-based Gothenburg H70 Birth Cohort Studies, Sweden. A total of 269 dementia-free 70-year-olds with dietary and cerebrospinal fluid (CSF) amyloid beta (Aβ42 and Aβ40), total tau (t-tau), and phosphorylated tau (p-tau) data were investigated. Dietary intake was determined by the diet history method, and four dietary patterns were derived by principal component analysis. A Western dietary pattern, a Mediterranean/prudent dietary pattern, a high-protein and alcohol pattern, and a high-total and saturated fat pattern. Logistic regression models, with CSF biomarker pathology (yes/no) as dependent variables, and linear regression models with continuous CSF biomarker levels as dependent variables were performed. The analyses were adjusted for sex, energy intake, body mass index (BMI), educational level, and physical activity level. RESULTS The odds ratio for having total tau pathology (odds ratio [OR] 1.43; 95% confidence interval [CI] 1.02 to 2.01) and preclinical AD (Aβ42 and tau pathology; OR 1.79; 95% CI 1.03 to 3.10) was higher among those with a higher adherence to a Western dietary pattern. There were no other associations between the dietary patterns and CSF biomarkers that remained significant in both unadjusted and adjusted models. DISCUSSION Our findings suggest that higher adherence to a Western dietary pattern may be associated with pathological levels of AD biomarkers in the preclinical phase of AD. These findings can be added to the increasing amount of evidence linking diet with AD and may be useful for future intervention studies investigating dietary intake in relation to AD.
Collapse
Affiliation(s)
- Jessica Samuelsson
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of GothenburgGothenburgSweden
| | - Silke Kern
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of GothenburgGothenburgSweden
| | - Henrik Zetterberg
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgGothenburgSweden
- UK Dementia Research Institute at UCLLondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | - Kaj Blennow
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgGothenburgSweden
| | | | - Ola Wallengren
- Clinical Nutrition UnitSahlgrenska University HospitalGothenburgSweden
| | - Ingmar Skoog
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of GothenburgGothenburgSweden
| | - Anna Zettergren
- Neuropsychiatric Epidemiology UnitDepartment of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiology, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP) at the University of GothenburgGothenburgSweden
| |
Collapse
|
47
|
Babulal GM, Roe CM, Stout SH, Rajasekar G, Wisch JK, Benzinger TLS, Morris JC, Ances BM. Depression is Associated with Tau and Not Amyloid Positron Emission Tomography in Cognitively Normal Adults. J Alzheimers Dis 2021; 74:1045-1055. [PMID: 32144985 DOI: 10.3233/jad-191078] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Depression is also common with older age. Alzheimer's disease (AD) studies suggest that both cerebrospinal fluid and positron emission tomography (PET) amyloid biomarkers are associated with more depressive symptoms in cognitively normal older adults. The recent availability of tau radiotracers offers the ability to examine in vivo tauopathy. It is unclear if the tau biomarker is associated with depression diagnosis. OBJECTIVE We examined if tau and amyloid imaging were associated with a depression diagnosis among cognitively normal adults (Clinical Dementia Rating = 0) and whether antidepressants modified this relationship. METHODS Among 301 participants, logistic regression models evaluated whether in vivo PET tau was associated with depression, while another model tested the interaction between PET tau and antidepressant use. A second set of models substituted PET amyloid for PET tau. A diagnosis of depression (yes/no) was made during an annual clinical assessment by a clinician. Antidepressant use (yes/no) was determined by comparing medications the participants used to a list of 30 commonly used antidepressants. All models adjusted for age, sex, education, race, and apolipoprotein ɛ4. Similar models explored the association between the biomarkers and depressive symptoms. RESULTS Participants with elevated tau were twice as likely to be depressed. Antidepressant use modified this relationship where participants with elevated tau who were taking antidepressants had greater odds of being depressed. Relatedly, elevated amyloid was not associated with depression. CONCLUSIONS Our results demonstrate that tau, not amyloid, was associated with a depression diagnosis. Additionally, antidepressant use interacts with tau to increase the odds of depression among cognitively normal adults.
Collapse
Affiliation(s)
- Ganesh M Babulal
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Catherine M Roe
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah H Stout
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ganesh Rajasekar
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Julie K Wisch
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA
| | - John C Morris
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Physical Therapy, Washington University School of Medicine, St. Louis, MO, USA.,Department of Occupational Therapy, Washington University School of Medicine, St. Louis, MO, USA
| | - Beau M Ances
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
48
|
Burke BT, Latimer C, Keene CD, Sonnen JA, McCormick W, Bowen JD, McCurry SM, Larson EB, Crane PK. Theoretical impact of the AT(N) framework on dementia using a community autopsy sample. Alzheimers Dement 2021; 17:1879-1891. [PMID: 33900044 DOI: 10.1002/alz.12348] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/02/2021] [Accepted: 03/11/2021] [Indexed: 11/07/2022]
Abstract
The AT(N) research framework categorizes eight biomarker profiles using amyloid (A), tauopathy (T), and neurodegeneration (N), regardless of dementia status. We evaluated associations with dementia risk in a community-based cohort by approximating AT(N) profiles using autopsy-based neuropathology correlates, and considered cost implications for clinical trials for secondary prevention of dementia based on AT(N) profiles. We used Consortium to Establish a Registry for Alzheimer's Disease (moderate/frequent) to approximate A+, Braak stage (IV-VI) for T+, and temporal pole lateral ventricular dilation for (N)+. Outcomes included dementia prevalence at death and incidence in the last 5 years of life. A+T+(N)+ was the most common profile (31%). Dementia prevalence ranged from 14% (A-T-[N]-) to 79% (A+T+[N]+). Between 8% (A+T-[N]-) and 68% (A+T+[N]-) of decedents developed incident dementia in the last 5 years of life. Clinical trials would incur substantial expense to characterize AT(N). Many people with biomarker-defined preclinical Alzheimer's disease will never develop clinical dementia during life, highlighting resilience to clinical expression of AD neuropathologic changes and the need for improved tools for prediction beyond current AT(N) biomarkers.
Collapse
Affiliation(s)
- Bridget Teevan Burke
- Kaiser Permanente, Washington Health Research Institute, Seattle, Washington, USA
| | - Caitlin Latimer
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Joshua A Sonnen
- Department of Pathology, McGill University, Montreal, Quebec, Canada
| | - Wayne McCormick
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - James D Bowen
- Department of Neurology, Swedish Hospital Medical Center, Seattle, Washington, USA
| | - Susan M McCurry
- Department of Community Health and Nursing, University of Washington, Seattle, Washington, USA
| | - Eric B Larson
- Kaiser Permanente, Washington Health Research Institute, Seattle, Washington, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
49
|
Gopalan D, Pandey A, Alex AT, Kalthur G, Pandey S, Udupa N, Mutalik S. Nanoconstructs as a versatile tool for detection and diagnosis of Alzheimer biomarkers. NANOTECHNOLOGY 2021; 32:142002. [PMID: 33238254 DOI: 10.1088/1361-6528/abcdcb] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The current review focuses towards the advancements made in the past decade in the field of nanotechnology for the early Alzheimer's disease (AD) diagnosis. This review includes the application of nanomaterials and nanosensors for the early detection of the main AD biomarkers (amyloid beta, phosphorylated tau, apolipoprotein E4 allele or APOE4, microRNAs, cholesterol, hydrogen peroxide etc) in biological fluids, to detect the biomarkers at a very low concentration ranging in pico, femto and even atto molar concentrations. The field of drug development has always aimed and is constantly working on developing disease modifying drugs, but these drugs will only succeed when given in the early disease stages. Thus, developing efficient diagnostic tools is of vital importance. Various nanomaterials such as liposomes; dendrimers; polymeric nanoparticles; coordination polymers; inorganic nanoparticles such as silica, manganese oxide, zinc oxide, iron oxide, super paramagnetic iron oxides; quantum dots, silver nanoparticles, gold nanoparticles, and carbon based nanostructures (carbon nanotubes, graphene oxide, nanofibres, nanodiamonds, carbon dots); Up-conversion nanoparticles; 2D nanomaterials; and radioactive nanoprobes have been used in constructing and improving efficiency of nano-sensors for AD biosensing at an early stage of diagnosis.
Collapse
Affiliation(s)
- Divya Gopalan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Abhijeet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Angel Treasa Alex
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Guruprasad Kalthur
- Department of Clinical Embryology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Sureshwar Pandey
- School of Pharmacy, Faculty of Medical Sciences, The university of West Indies, St. Augustine, Trinidad and Tobago, Jamaica
| | - Nayanabhirama Udupa
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| |
Collapse
|
50
|
Rabipour S, Rajagopal S, Pasvanis S, Rajah MN. Generalization of memory-related brain function in asymptomatic older women with a family history of late onset Alzheimer's Disease: Results from the PREVENT-AD Cohort. Neurobiol Aging 2021; 104:42-56. [PMID: 33964608 DOI: 10.1016/j.neurobiolaging.2021.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/04/2021] [Accepted: 03/18/2021] [Indexed: 11/18/2022]
Abstract
Late-onset Alzheimer's disease (AD) disproportionately affects women compared to men. Episodic memory decline is one of the earliest and most pronounced deficits observed in AD. However, it remains unclear whether sex influences episodic memory-related brain function in cognitively intact older adults at risk of developing AD. Here we used task-based multivariate partial least squares analysis to examine sex differences in episodic memory-related brain activity and brain activity-behavior correlations in a matched sample of cognitively intact older women and men with a family history of AD from the PREVENT-AD cohort study in Montreal, Canada (Mage=63.03±3.78; Meducation=15.41±3.40). We observed sex differences in task-related brain activity and brain activity-behavior correlations during the encoding of object-location associative memories and object-only item memory, and the retrieval of object only item memories. Our findings suggest a generalization of episodic memory-related brain activation and performance in women compared to men. Follow up analyses should test for sex differences in the relationship between brain activity patterns and performance longitudinally, in association with risk factors for AD development. This article is part of the Virtual Special Issue titled COGNITIVE NEUROSCIENCE OF HEALTHY AND PATHOLOGICAL AGING. The full issue can be found on ScienceDirect at https://www.sciencedirect.com/journal/neurobiology-of-aging/special-issue/105379XPWJP.
Collapse
Affiliation(s)
- Sheida Rabipour
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Canada; Department of Psychiatry, McGill University, Montreal, Canada
| | | | - Stamatoula Pasvanis
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Canada
| | - M Natasha Rajah
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Canada; Department of Psychiatry, McGill University, Montreal, Canada.
| |
Collapse
|