1
|
Lu Y, Li D, Yu Y, Wang Q, Li A, Quan Y, Xing Y. Cerebrospinal fluid VGF is associated with the onset and progression of Alzheimer's disease. J Alzheimers Dis 2025; 104:1235-1242. [PMID: 40095667 DOI: 10.1177/13872877251323002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
BackgroundIt remains unclear whether cerebrospinal fluid (CSF) VGF (non-acronymic) is associated with the onset and progression of Alzheimer's disease (AD).ObjectiveTo assess the levels of CSF VGF throughout the AD continuum, and its association with primary AD pathology, cognition, brain atrophy, and brain metabolism.MethodsWe studied a total of 526 individuals including 377 amyloid-positive individuals (76 preclinical AD, 200 prodromal AD, and 101 AD dementia) and 149 amyloid-negative cognitively normal individuals. VGF peptide in CSF was analyzed using mass spectrometry.ResultsWe observed decreased CSF VGF in preclinical, prodromal, and AD dementia individuals than amyloid-negative cognitively normal individuals. Reduced CSF VGF was associated with cognitive decline, hippocampal atrophy, ventricle enlargement, and glucose hypometabolism at baseline, and it predicted a more marked deterioration over time.ConclusionsOur findings support the important contributions of VGF to disease pathogenesis and progression in the early stages of AD. Exploring the biologics modulating VGF might be a promising approach for AD prevention and early treatment.
Collapse
Affiliation(s)
- Yuanyuan Lu
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dan Li
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yueyi Yu
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qianqian Wang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Aonan Li
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yixin Quan
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yi Xing
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Pradeep S, Sai Chakith MR, Sindhushree SR, Reddy P, Sushmitha E, Purohit MN, Suresh D, Swamy Shivananju N, Silina E, Manturova N, Stupin V, Kollur SP, Shivamallu C, Achar RR. Exploring shared therapeutic targets for Alzheimer's disease and glioblastoma using network pharmacology and protein-protein interaction approach. Front Chem 2025; 13:1549186. [PMID: 40144222 PMCID: PMC11938128 DOI: 10.3389/fchem.2025.1549186] [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: 12/23/2024] [Accepted: 02/17/2025] [Indexed: 03/28/2025] Open
Abstract
Background Alzheimer's disease (AD) and glioblastoma (GBM) are complex neurological disorders with distinct pathologies but overlapping molecular mechanisms, including neuroinflammation, oxidative stress, and dysregulated signaling pathways. Despite significant advancements in research, effective therapies targeting both conditions remain elusive. Identifying shared molecular targets and potential therapeutic agents could offer novel treatment strategies for these disorders. Methodology The study employs an integrative network pharmacology approach to explore the therapeutic potential of bioactive compounds from Eclipta alba, a medicinal herb known for its neuroprotective and anti-inflammatory properties. A systematic methodology was adopted, starting with network pharmacology analysis using STRING and DisGeNET databases, which identified 617 common genes associated with AD and GBM. Among these, key hub genes-TP53, STAT3, AKT1, and IL6-were prioritized using Cytoscape for network visualization and analysis. Results Molecular docking studies were conducted using PyRx software to assess the binding interactions of 26 phytochemicals from Eclipta alba against the identified target genes. Luteolin exhibited the highest binding affinity to IL6 (-7.8 kcal/mol), forming stable hydrogen bonds and hydrophobic interactions. To further validate this interaction, molecular dynamics simulations (MDS) were performed using GROMACS, confirming the stability of the Luteolin-IL6 complex. Additionally, MM-PBSA binding energy calculations using AmberTools (-145.44 kJ/mol) provided further evidence of a strong and stable interaction. Pharmacokinetic and toxicity evaluations, conducted using SwissADME and pkCSM, highlighted luteolin's favorable drug-like properties, including good bioavailability and low toxicity. These findings suggest that luteolin may serve as a promising multi-target therapeutic agent for AD and GBM by modulating key pathological pathways. Conclusion The present study provides a strong computational foundation for further in vitro and in vivo validation. The results highlight the potential of luteolin in developing dual-target treatment strategies for neurodegenerative and oncological disorders, offering new avenues for therapeutic advancements.
Collapse
Affiliation(s)
- Sushma Pradeep
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
- Centre for Digital Health and AI, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - M. R. Sai Chakith
- Department of Pharmacology, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - S. R. Sindhushree
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Pruthvish Reddy
- Department of Biotechnology, Acharya Institute of Technology, Bengaluru, Karnataka, India
| | - Esther Sushmitha
- Department of Pharmacology, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Madhusudan N. Purohit
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Divya Suresh
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Nanjunda Swamy Shivananju
- Department of Biotechnology, JSS Science and Technology University, Sri Jayachamarajendra College of Engineering, Mysuru, Karnataka, India
| | - Ekaterina Silina
- Institute of Digital Biodesign and Modeling of Living Systems, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Natalia Manturova
- Department of Surgery, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Victor Stupin
- Department of Surgery, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Shiva Prasad Kollur
- School of Physical Sciences, Amrita Vishwa Vidyapeetham, Mysuru Campus, Mysuru, Karnataka, India
| | - Chandan Shivamallu
- Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Raghu Ram Achar
- Division of Biochemistry, School of Life Sciences, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| |
Collapse
|
3
|
Wei P, Lin K, Chen X, Fang C, Qiu L, Hu J, Chang J. Sequential Proteomic Analysis Reveals the Key APOE4-Induced Pathological and Molecular Features at the Presymptomatic Stage in Alzheimer's Disease Mice. CNS Neurosci Ther 2025; 31:e70306. [PMID: 40075551 PMCID: PMC11903334 DOI: 10.1111/cns.70306] [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: 01/09/2025] [Revised: 02/06/2025] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
AIMS Alzheimer's disease (AD) involves a prolonged presymptomatic or preclinical stage with subtle pathological changes. Apolipoprotein E4 (APOE4) is a significant genetic risk factor for AD, yet its specific role at the presymptomatic stage is not fully understood. This study aimed to elucidate the cellular and molecular effects of APOE4 compared to APOE3 on AD progression during the presymptomatic stage. METHODS We generated 5xFAD AD mice carrying human APOE3 or APOE4 and their non-AD controls. Behavioral tests, immunostaining, quantitative proteomics and phosphoproteomics, Golgi staining, and Western blotting were conducted at 3 or 10 months of age, respectively. Cell culture experiments were performed to assess APOE4's direct impact on neuronal mitochondrial function. RESULTS APOE4 significantly increased β-amyloid (Aβ) deposition and microglial activation compared to APOE3 in 5xFAD mice at the presymptomatic stage, without aggravating the blood-brain barrier disruption. Proteomic and biochemical analysis revealed strong molecular features of synaptic degeneration and mitochondrial dysfunction associated with APOE4. Notably, APOE4 promoted mitochondrial fusion and mitophagy while inhibiting fission, leading to impaired neuronal energy supply and increased reactive oxygen species. CONCLUSION Our findings indicate that APOE4 accelerates AD pathologies at the presymptomatic stage by exacerbating Aβ deposition, neuroinflammation, and synaptic degeneration. The study highlights mitochondrial dysfunction as a critical mediator of APOE4-induced AD progression, providing potential targets for early intervention.
Collapse
Affiliation(s)
- Pengju Wei
- State Key Laboratory of Biomedical Imaging Science and SystemInstitute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Kaihua Lin
- Shantou University Medical CollegeShantouGuangdongChina
- Department of NeurologyPeking University Shenzhen HospitalShenzhenGuangdongChina
| | - Xuhui Chen
- Department of NeurologyPeking University Shenzhen HospitalShenzhenGuangdongChina
| | - Cheng Fang
- State Key Laboratory of Biomedical Imaging Science and SystemInstitute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Linhui Qiu
- State Key Laboratory of Biomedical Imaging Science and SystemInstitute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Jun Hu
- Shantou University Medical CollegeShantouGuangdongChina
- Department of NeurologyPeking University Shenzhen HospitalShenzhenGuangdongChina
| | - Junlei Chang
- State Key Laboratory of Biomedical Imaging Science and SystemInstitute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| |
Collapse
|
4
|
Dhana A, DeCarli CS, Dhana K, Desai P, Krueger K, Evans DA, Rajan KB. Association of subjective memory complaints with serum biomarkers of neurodegeneration and cognition: A population-based study. J Am Geriatr Soc 2025; 73:859-866. [PMID: 39625703 PMCID: PMC11908938 DOI: 10.1111/jgs.19300] [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: 05/21/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 12/08/2024]
Abstract
BACKGROUND Subjective memory complaints (SMCs) refer to memory concerns reported by an individual, regardless of objective test impairment. We conducted a study to evaluate the association of SMCs with serum biomarkers of neurodegeneration, including neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), and total tau (t-tau). In addition, we evaluated the association of SMCs with the rate of cognitive decline. METHODS This study included 1096 individuals participating in the Chicago Health and Aging Project, with data on SMCs, neurodegenerative biomarkers (NfL, GFAP, and t-tau), and global cognitive scores. The SMC score ranges from 0 to 8 and higher scores reflect more memory complaints. Linear regression models were developed to investigate the association of SMCs with serum biomarkers, adjusted by age, sex, race, education, and APOE e4. Linear mixed-effects models were used to examine the independent association of SMCs with cognitive decline in a multivariable model that was additionally adjusted by biomarkers of neurodegeneration. RESULTS Of the 1096 individuals, 665 (60.7%) were female, 643 (58.7%) were African Americans, and the mean (SD) age at the baseline was 78.0 (5.8) years. Compared to individuals with fewer memory complaints (i.e., people in the first tertile of SMCs), those reporting more memory complaints (i.e., people in the third tertile of SMCs) had a 12.0% increase in serum concentrations of NfL and an 9.4% increase in GFAP. In addition, individuals reporting more memory complaints (i.e., those in the 3rd versus the 1st tertile of SMCs) had a faster rate of cognitive decline by 0.029 ( β -0.029, 95% CI -0.046 to -0.013) standardized units per year. CONCLUSIONS Individuals who reported more memory complaints had higher levels of biomarkers of neurodegeneration and a faster rate of cognitive decline, suggesting that SMCs may be valuable for identifying people at high risk of cognitive impairment.
Collapse
Affiliation(s)
- Anisa Dhana
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
| | - Charles S. DeCarli
- Department of Neurology, University of California at Davis, Sacramento, CA, USA
| | - Klodian Dhana
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
| | - Pankaja Desai
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
| | - Kristin Krueger
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
| | - Denis A. Evans
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
| | - Kumar B. Rajan
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL
- Department of Neurology, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
5
|
Bendl J, Fullard JF, Girdhar K, Dong P, Kosoy R, Zeng B, Hoffman GE, Roussos P. Chromatin accessibility provides a window into the genetic etiology of human brain disease. Trends Genet 2025:S0168-9525(25)00001-0. [PMID: 39855972 DOI: 10.1016/j.tig.2025.01.001] [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: 11/18/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/27/2025]
Abstract
Neuropsychiatric and neurodegenerative diseases have a significant genetic component. Risk variants often affect the noncoding genome, altering cis-regulatory elements (CREs) and chromatin structure, ultimately impacting gene expression. Chromatin accessibility profiling methods, especially assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq), have been used to pinpoint disease-associated SNPs and link them to affected genes and cell types in the brain. The integration of single-cell technologies with genome-wide association studies (GWAS) and transcriptomic data has further advanced our understanding of cell-specific chromatin dynamics. This review discusses recent findings regarding the role played by chromatin accessibility in brain disease, highlighting the need for high-quality data and rigorous computational tools. Future directions include spatial chromatin studies and CRISPR-based functional validation to bridge genetic discovery and clinical applications, paving the way for targeted gene-regulatory therapies.
Collapse
Affiliation(s)
- Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kiran Girdhar
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pengfei Dong
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Roman Kosoy
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Biao Zeng
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY 10468, USA; Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY 10468, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY 10468, USA; Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY 10468, USA.
| |
Collapse
|
6
|
Milne R, Richard E, Brayne C. Challenges associated with the development of "trial ready cohorts" for dementia prevention trials. BMJ 2025; 388:e080275. [PMID: 39773881 DOI: 10.1136/bmj-2024-080275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Affiliation(s)
- Richard Milne
- Wellcome Connecting Science, Wellcome Sanger Institute, Hinxton, UK
- Kavli Centre for Ethics, Science, and the Public, University of Cambridge, UK
| | - Edo Richard
- Department of Neurology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, Nijmegen, Netherlands
- Department of Public and Occupational Health, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Carol Brayne
- Department of Psychiatry, Cambridge Public Health, University of Cambridge, UK
| |
Collapse
|
7
|
Wang X, Wang X, Edland SD, Broce IJ, Dale AM, Banks SJ, for the Alzheimer's Disease Neuroimaging Initiative. Enrichment for clinical trials of early AD: Combining genetic risk factors and plasma p-tau as screening instruments. Alzheimers Dement 2024; 20:8484-8502. [PMID: 39440707 PMCID: PMC11667492 DOI: 10.1002/alz.14284] [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: 05/22/2024] [Revised: 08/09/2024] [Accepted: 09/05/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Identifying low-cost, minimally-invasive screening instruments for Alzheimer's disease (AD) trial enrichment will improve the efficiency of AD trials. METHODS A total of 685 cognitively normal (CN) individuals and individuals with mild cognitive impairment (MCI) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were grouped according to cutoffs of genetic risk factor (G) polygenic hazard score (PHS) and tau pathology (T) plasma phosphorylated tau-181 (p-tau181) into four groups: G+T+, G-T-, G+T-, and G-T+. We assessed the associations between group level and longitudinal cognitive decline and AD conversion. Power analyses compared the estimated sample size required to detect differences in cognitive decline. RESULTS The G+T+ group was associated with faster cognitive decline and higher AD risk. Clinical trials enrolling G+T+ participants would benefit from significantly reduced sample sizes compared with similar trials using only single makers as an inclusion criterion. DISCUSSION The combination of two low-cost, minimally-invasive measures-genetics and plasma biomarkers-would be a promising screening procedure for clinical trial enrollment. HIGHLIGHTS Participants with unimpaired or mildly impaired cognition were grouped based on cutoffs on genetic risk factors (G: polygenic hazardous score [PHS]) and Alzheimer's pathology (T: baseline plasma phosphorylated tau-181 [p-tau181]). Participants with high PHSs and plasma p-tau181 levels (G+T+) were at risk of faster cognitive decline and AD progression. The combination of PHS and plasma p-tau181 could enhance clinical trial enrichment more effectively than using single biomarkers.
Collapse
Affiliation(s)
- Xin Wang
- Department of NeuroscienceUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Xinran Wang
- Division of Biostatistics, Herbert Wertheim School of Public Health and Human Longevity ScienceUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Steven D. Edland
- Department of NeuroscienceUniversity of California San DiegoLa JollaCaliforniaUSA
- Division of Biostatistics, Herbert Wertheim School of Public Health and Human Longevity ScienceUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Iris J. Broce
- Department of NeuroscienceUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Anders M. Dale
- Department of NeuroscienceUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Sarah J. Banks
- Department of NeuroscienceUniversity of California San DiegoLa JollaCaliforniaUSA
| | | |
Collapse
|
8
|
Liu H, Yu C, Qin C. The changes of peripheral blood hub genes in 24-week-old APP/PS1/Tau triple transgenic mouse model based on weighted gene co-expression network analysis. AN ACAD BRAS CIENC 2024; 96:e20240120. [PMID: 39383430 DOI: 10.1590/0001-3765202420240120] [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: 02/06/2024] [Accepted: 05/06/2024] [Indexed: 10/11/2024] Open
Abstract
Peripheral regulation emerges as a promising intervention in the early stages of Alzheimer's disease (AD). The hub genes in the peripheral blood of MCI patients from GEO database (GSE63060, GSE63061) were screened using weighted gene co-expression analysis (WGCNA). Meanwhile, behavioral tests, HE staining and Nissl staining were used to detect the memory impairment and histopathological changes in 24-week-old male 3×Tg-AD mice. Thioflavin-S and immunohistochemical staining were used to determine the Aβ deposition in both intracellular and extracellular neurons. Subsequently, the MCI-hub genes were verified by quantitative real-time PCR (qRT-PCR) in the peripheral blood of 3×Tg-AD mice. The research revealed ten hub genes associated with MCI were identified WGCNA. Short-term memory loss, intracellular Aβ deposition and limited of extracellular amyloid plaques in 3×Tg-AD mice. The qRT-PCR analysis of peripheral blood from these mice revealed significantly down-regulation in the expression levels of ATP5C1, ITGB2, EFTUD2 and RPS27A genes; whereas the expression level of VCP gene was significantly up-regulated. These findings confirmed that 24-week-old male 3×Tg-AD mice were a valuable animal model for simulating the early symptomatic stages of AD. Additionally, the peripheral blood MCI-hub genes related to immune response, energy metabolism and ribosomal coding efficiency provide potential biomarkers for this stage.
Collapse
Affiliation(s)
- Hexu Liu
- First Affiliated Hospital of Guangxi Medical University, Department of Neurology, No. 6 ShuangYong Road, Nanning 530021, Guangxi Autonomous Region, China
| | - Changyin Yu
- Affiliated Hospital of Zunyi Medical University, Department of Neurology, No. 149 Dalian Road, Zunyi 563000, Guizhou Province, China
| | - Chao Qin
- First Affiliated Hospital of Guangxi Medical University, Department of Neurology, No. 6 ShuangYong Road, Nanning 530021, Guangxi Autonomous Region, China
| |
Collapse
|
9
|
Miller MJ, Diaz A, Conti C, Albala B, Flenniken D, Fockler J, Kwang W, Sacrey DT, Ashford MT, Skirrow C, Weston J, Fristed E, Farias ST, Korecka M, Wan Y, Aisen PS, Beckett L, Harvey D, Lee EB, Petersen RC, Shaw LM, Okonkwo OC, Mindt MR, Weiner MW, Nosheny RL, Alzheimer's Disease Neuroimaging Initiative. The ADNI4 Digital Study: A novel approach to recruitment, screening, and assessment of participants for AD clinical research. Alzheimers Dement 2024; 20:7232-7247. [PMID: 39219153 PMCID: PMC11485063 DOI: 10.1002/alz.14234] [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: 05/01/2024] [Revised: 07/18/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION We evaluated preliminary feasibility of a digital, culturally-informed approach to recruit and screen participants for the Alzheimer's Disease Neuroimaging Initiative (ADNI4). METHODS Participants were recruited using digital advertising and completed digital surveys (e.g., demographics, medical exclusion criteria, 12-item Everyday Cognition Scale [ECog-12]), Novoic Storyteller speech-based cognitive test). Completion rates and assessment performance were compared between underrepresented populations (URPs: individuals from ethnoculturally minoritized or low education backgrounds) and non-URPs. RESULTS Of 3099 participants who provided contact information, 654 enrolled in the cohort, and 595 completed at least one assessment. Two hundred forty-seven participants were from URPs. Of those enrolled, 465 met ADNI4 inclusion criteria and 237 evidenced possible cognitive impairment from ECog-12 or Storyteller performance. URPs had lower ECog and Storyteller completion rates. Scores varied by ethnocultural group and educational level. DISCUSSION Preliminary results demonstrate digital recruitment and screening assessment of an older diverse cohort, including those with possible cognitive impairment, are feasible. Improving engagement and achieving educational diversity are key challenges. HIGHLIGHTS A total of 654 participants enrolled in a digital cohort to facilitate ADNI4 recruitment. Culturally-informed digital ads aided enrollment of underrepresented populations. From those enrolled, 42% were from underrepresented ethnocultural and educational groups. Digital screening tools indicate > 50% of participants likely cognitively impaired. Completion rates and assessment performance vary by ethnocultural group and education.
Collapse
|
10
|
Martino-Adami PV, Chatterjee M, Kleineidam L, Weyerer S, Bickel H, Wiese B, Riedel-Heller SG, Scherer M, Blennow K, Zetterberg H, Wagner M, Schneider A, Ramirez A. Prognostic value of Alzheimer's disease plasma biomarkers in the oldest-old: a prospective primary care-based study. THE LANCET REGIONAL HEALTH. EUROPE 2024; 45:101030. [PMID: 39253733 PMCID: PMC11381503 DOI: 10.1016/j.lanepe.2024.101030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 09/11/2024]
Abstract
Background Blood-based biomarkers offer a promising, less invasive, and more cost-effective alternative for Alzheimer's disease screening compared to cerebrospinal fluid or imaging biomarkers. However, they have been extensively studied only in memory clinic-based cohorts. We aimed to validate them in a more heterogeneous, older patient population from primary care. Methods We measured plasma Aβ42/Aβ40, P-tau181, NfL, and GFAP in 1007 individuals without dementia, aged 79-94 years, from the longitudinal, primary care-based German AgeCoDe study. We assessed the association with cognitive decline, disease progression, and the capacity to predict future dementia of the Alzheimer's type (DAT). We also evaluated biomarker dynamics in 305 individuals with a follow-up sample (∼8 years later). Findings Higher levels of P-tau181 (HR = 1.32 [95% CI: 1.17-1.51]), NfL (HR = 1.19 [95% CI: 1.03-1.36]), and GFAP (HR = 1.36 [95% CI: 1.22-1.52]), and a lower Aβ42/Aβ40 ratio (HR = 0.80 [95% CI: 0.68-0.95]) were associated with an increased risk of progressing to clinically-diagnosed DAT. Additionally, higher levels of P-tau181 (β = -0.49 [95% CI: -0.71 to 0.26]), NfL (β = -0.29 [95% CI: -0.52 to 0.06]), and GFAP (β = -0.60 [95% CI: -0.83 to 0.38]) were linked to faster cognitive decline. A two-step DAT prediction strategy combining initial MMSE with biomarkers improved the identification of individuals in the prodromal stage for potential treatment eligibility. Biomarker levels changed over time, with increases in P-tau181 (β = 0.19 [95% CI: 0.14-0.25]), NfL (β = 2.88 [95% CI: 2.18-3.59]), and GFAP (β = 8.23 [95% CI: 6.71-9.75]). NfL (β = 2.47 [95% CI: 1.04-3.89]) and GFAP (β = 4.45 [95% CI: 1.38-7.51]) exhibited a faster increase in individuals progressing to DAT. Interpretation Evaluating plasma biomarkers, alongside brief cognitive assessments, might enhance the precision of risk assessment for DAT progression in primary care. Funding Alzheimer Forschung Initiative, Bundesministerium für Bildung und Forschung.
Collapse
Affiliation(s)
- Pamela V Martino-Adami
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Madhurima Chatterjee
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, Medical Faculty, Venusberg-Campus 1, 53127, Bonn, Germany
| | | | - Horst Bickel
- Department of Psychiatry, Technical University of Munich, Germany
| | - Birgitt Wiese
- Institute of General Practice, Hannover Medical School, Germany
| | - Steffi G Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, Germany
| | - Martin Scherer
- Department of Primary Medical Care, Center for Psychosocial Medicine, University Medical Center, Hamburg-Eppendorf, Germany
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, 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, PR China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, 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
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, Medical Faculty, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, Medical Faculty, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, Medical Faculty, Venusberg-Campus 1, 53127, Bonn, Germany
- Department of Psychiatry and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, 7703 Floyd Curl Drive, 78229, San Antonio, TX, USA
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, 50931, Cologne, Germany
| |
Collapse
|
11
|
Guo S, Zhu W, Bian Y, Li Z, Zheng H, Li W, Yang Y, Ji X, Zhang B. Developing diagnostic biomarkers for Alzheimer's disease based on histone lactylation-related gene. Heliyon 2024; 10:e37807. [PMID: 39315143 PMCID: PMC11417585 DOI: 10.1016/j.heliyon.2024.e37807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Background Research underscores the significant influence of histone lactylation pathways in the progression of Alzheimer's disease (AD), though the molecular mechanisms associated with histone lactylation-related genes (HLRGs) in AD are still insufficiently investigated. Methods This study employed datasets GSE85426 and GSE97760 to identify candidate genes by intersecting weighted gene co-expression network analysis (WGCNA) module genes with AD-control differentially expressed genes (DEGs). Subsequently, machine learning refined key genes, validated by receiver operating characteristic (ROC) curve performance. Gene-set enrichment analysis (GSEA) explored the molecular mechanisms of these diagnostic markers. Concurrently, the association between the diagnostic genes and both differential immune cells and immune responses was examined. Furthermore, a ceRNA and gene-drug network was developed. Finally, the expression of the selected genes was validated using brain tissues from AD model mice. Results This study identified five genes (ARID5B, NSMCE4A, SESN1, THADA, and XPA) with significant diagnostic utility, primarily enriched in olfactory transduction and N-glycan biosynthesis pathways. Correlation analysis demonstrated a strong positive association between all diagnostic genes and naive B cells. The ceRNA regulatory network comprised 7 miRNAs, 2 mRNAs, and 25 lncRNAs. Additionally, 33 drugs targeting the diagnostic genes were predicted. Following expression validation through training and validation sets, three genes (ARID5B, SESN1, XPA) were ultimately confirmed as biomarkers for this study. RT-qPCR and Western blot analyses revealed upregulated expression of ARID5B, SESN1, and XPA in the cerebral tissue of AD model mice. Conclusion Three histone lactylation-linked genes (ARID5B, SESN1, XPA) were identified as potential AD biomarkers, indicating a strong association with disease progression.
Collapse
Affiliation(s)
- Shaobo Guo
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Wenhui Zhu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yuting Bian
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Zhikai Li
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Heng Zheng
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
- Zhenjiang Hospital of Chinese Traditional And Western Medicine, Zhenjiang, China
| | - Wenlong Li
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
- Liyang Hospital of Chinese Medicine, Liyang, China
| | - Yi Yang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Xuzheng Ji
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Biao Zhang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Geriatric, Nanjing, China
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
12
|
Karlsson L, Vogel J, Arvidsson I, Åström K, Strandberg O, Seidlitz J, Bethlehem RAI, Stomrud E, Ossenkoppele R, Ashton NJ, Zetterberg H, Blennow K, Palmqvist S, Smith R, Janelidze S, Joie RL, Rabinovici GD, Binette AP, Mattsson-Carlgren N, Hansson O. A machine learning-based prediction of tau load and distribution in Alzheimer's disease using plasma, MRI and clinical variables. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.31.24308264. [PMID: 38853877 PMCID: PMC11160861 DOI: 10.1101/2024.05.31.24308264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Tau positron emission tomography (PET) is a reliable neuroimaging technique for assessing regional load of tau pathology in the brain, commonly used in Alzheimer's disease (AD) research and clinical trials. However, its routine clinical use is limited by cost and accessibility barriers. Here we explore using machine learning (ML) models to predict clinically useful tau-PET composites from low-cost and non-invasive features, e.g., basic clinical variables, plasma biomarkers, and structural magnetic resonance imaging (MRI). Results demonstrated that models including plasma biomarkers yielded the most accurate predictions of tau-PET burden (best model: R-squared=0.66-0.68), with especially high contribution from plasma P-tau217. In contrast, MRI variables stood out as best predictors (best model: R-squared=0.28-0.42) of asymmetric tau load between the two hemispheres (an example of clinically relevant spatial information). The models showed high generalizability to external test cohorts with data collected at multiple sites. Based on these results, we also propose a proof-of-concept two-step classification workflow, demonstrating how the ML models can be translated to a clinical setting. This study uncovers current potential in predicting tau-PET information from scalable cost-effective variables, which could improve diagnosis and prognosis of AD.
Collapse
Affiliation(s)
- Linda Karlsson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Jacob Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Clinical Sciences, SciLifeLab, Lund University, Lund, Sweden
| | - Ida Arvidsson
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Kalle Åström
- Centre for Mathematical Sciences, Lund University, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Jakob Seidlitz
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104 USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104 USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Richard A. I. Bethlehem
- University of Cambridge, Department of Psychology, Cambridge Biomedical Campus, Cambridge, CB2 3EB, UK
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience, King’s College London, London, UK
| | - 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
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, 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
| | - 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
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, 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
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Gil D. Rabinovici
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
13
|
Sun X, Yuan X, Chen H, Li W. PM2.5 is linked to Alzheimer's syndrome and delirium: a mendelian randomization analysis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2024; 17:308-315. [PMID: 39399654 PMCID: PMC11470430 DOI: 10.62347/fmuc9744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/27/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Increasing air pollution has drawn our attention to particulate matter (PM2.5), which has been shown to correlate significantly with respiratory and cardiovascular systems. However, whether PM2.5 is causally associated with Alzheimer's syndrome or delirium is unclear. METHODS We retrieved the genetic summary data of PM2.5 from genome-wide association studies (GWAS). The genetic information for Alzheimer's disease was obtained from the IEU OpenGWAS project, and that for delirium was obtained from FinnGen. We used two-sample Mendelian randomization analysis (MR) to associate PM2.5 with Alzheimer's disease or delirium. RESULTS The odds ratio (OR) for Alzheimer's disease was 0.996 with a p-value of 0.443 using the inverse variance weighted algorithm, and the OR associated with the outcome variable of delirium was 0.393 with a p-value of 0.343. CONCLUSION With the exclusion of confounding factors, our findings do not support a genetic association between PM2.5 and Alzheimer's disease or delirium. Further population-based and experimental studies are needed to dissect the complex correlation between PM2.5 and Alzheimer's disease or delirium.
Collapse
Affiliation(s)
- Xiaojin Sun
- Department of Cardiology, Sihong HospitalSuqian 223900, Jiangsu, China
| | - Xiaofan Yuan
- Department of Radiology of The Second Affiliated Hospital of Nanjing Medical UniversityNanjing 210011, Jiangsu, China
| | - Haoyan Chen
- Department of Geriatrics, Jiangsu Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210000, Jiangsu, China
| | - Wenjie Li
- Department of Geriatrics, The First Affiliated Hospital of Ningbo UniversityNingbo 315000, Zhejiang, China
| |
Collapse
|
14
|
Song Y, Kim H, Lee J, Kim K. Oxygen-enriching triphase platform for reliable sensing of femtomolar Alzheimer's neurofilament lights. Biosens Bioelectron 2024; 260:116431. [PMID: 38815462 DOI: 10.1016/j.bios.2024.116431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Accurate quantification of neurofilament lights (NfLs), a prognostic blood biomarker, is highly required to predict neurodegeneration in the presymptomatic stages of Alzheimer's disease. Here, we report self-oxygen-enriching coral structures with triphase interfaces for the label-free photocathodic detection of NfLs in blood plasma with femtomolar sensitivities and high reliability. In conventional photocathodic immunoassays, the poor solubility and sluggish diffusion rate of the dissolved oxygen serving as electron acceptors have necessitated the incorporation of additional electron acceptors or aeration procedures. To address the challenge, we designed the coral-like copper bismuth oxides (CBO) with robust solid-liquid-air contact boundaries that enrich the interfacial oxygen levels without an external aeration source. By optimally assembling the perfluorododecyltrichlorosilane (FTCS) and platinum (Pt) co-catalysts into the silver-doped CBO (Ag:CBO), the stable solid-liquid-air contact boundaries were formed within the sensor interfaces, which allowed for the abundant supply of air phase oxygen through an air pocket connected to the atmosphere. The Pt/FTCS-Ag:CBO exhibited the stable background signals independent of the dissolved oxygen fluctuations and amplified photocurrent signals by 1.76-fold, which were attributed to the elevated interfacial oxygen levels and 11.15 times-lowered mass transport resistance. Under the illumination of white light-emitting diode, the oxygen-enriching photocathodic sensor composed of Pt/FTCS-Ag:CBO conjugated with NfLs-specific antibodies precisely quantified the NfLs in plasma with a low coefficient of variation (≤2.97%), a high degree of recovery (>97.0%), and a limit of detection of 40.38 fg/mL, which was 140 times lower than the typical photocathodic sensor with diphase interfaces.
Collapse
Affiliation(s)
- Yunji Song
- Department of Fiber Convergence Material Engineering, Dankook University, Gyeonggi-Do, 16890, Republic of Korea
| | - Hayeon Kim
- Department of Fiber Convergence Material Engineering, Dankook University, Gyeonggi-Do, 16890, Republic of Korea
| | - Joonseok Lee
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Kayoung Kim
- Department of Fiber Convergence Material Engineering, Dankook University, Gyeonggi-Do, 16890, Republic of Korea.
| |
Collapse
|
15
|
Bader I, Groot C, Tan HS, Milongo JMA, Haan JD, Verberk IMW, Yong K, Orellina J, Campbell S, Wilson D, van Harten AC, Kok PHB, van der Flier WM, Pijnenburg YAL, Barkhof F, van de Giessen E, Teunissen CE, Bouwman FH, Ossenkoppele R. Rationale and design of the BeyeOMARKER study: prospective evaluation of blood- and eye-based biomarkers for early detection of Alzheimer's disease pathology in the eye clinic. Alzheimers Res Ther 2024; 16:190. [PMID: 39169442 PMCID: PMC11340081 DOI: 10.1186/s13195-024-01545-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a common, complex and multifactorial disease that may require screening across multiple routes of referral to enable early detection and subsequent future implementation of tailored interventions. Blood- and eye-based biomarkers show promise as low-cost, scalable and patient-friendly tools for early AD detection given their ability to provide information on AD pathophysiological changes and manifestations in the retina, respectively. Eye clinics provide an intriguing real-world proof-of-concept setting to evaluate the performance of these potential AD screening tools given the intricate connections between the eye and brain, presumed enrichment for AD pathology in the aging population with eye disorders, and the potential for an accelerated diagnostic pathway for under-recognized patient groups. METHODS The BeyeOMARKER study is a prospective, observational, longitudinal cohort study aiming to include individuals visiting an eye-clinic. Inclusion criteria entail being ≥ 50 years old and having no prior dementia diagnosis. Excluded eye-conditions include traumatic insults, superficial inflammation, and conditions in surrounding structures of the eye that are not engaged in vision. The BeyeOMARKER cohort (n = 700) will undergo blood collection to assess plasma p-tau217 levels and a brief cognitive screening at the eye clinic. All participants will subsequently be invited for annual longitudinal follow-up including remotely administered cognitive screening and questionnaires. The BeyeOMARKER + cohort (n = 150), consisting of 100 plasma p-tau217 positive participants and 50 matched negative controls selected from the BeyeOMARKER cohort, will additionally undergo Aβ-PET and tau-PET, MRI, retinal imaging including hyperspectral imaging (primary), widefield imaging, optical coherence tomography (OCT) and OCT-Angiography (secondary), and cognitive and cortical vision assessments. RESULTS We aim to implement the current protocol between April 2024 until March 2027. Primary outcomes include the performance of plasma p-tau217 and hyperspectral retinal imaging to detect AD pathology (using Aβ- and tau-PET visual read as reference standard) and to detect cognitive decline. Initial follow-up is ~ 2 years but may be extended with additional funding. CONCLUSIONS We envision that the BeyeOMARKER study will demonstrate the feasibility of early AD detection based on blood- and eye-based biomarkers in alternative screening settings, and will improve our understanding of the eye-brain connection. TRIAL REGISTRATION The BeyeOMARKER study (Eudamed CIV ID: CIV-NL-23-09-044086; registration date: 19th of March 2024) is approved by the ethical review board of the Amsterdam UMC.
Collapse
Affiliation(s)
- Ilse Bader
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands.
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands.
| | - Colin Groot
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - H Stevie Tan
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, 1081 HV, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Amsterdam UMC Location VUmc, Amsterdam Reproduction and Development Research Institute, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Jean-Marie A Milongo
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
| | - Jurre den Haan
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Laboratory Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Keir Yong
- Queen Square Institute of Neurology, Dementia Research Centre, London, WC1N 3BG, UK
| | | | | | | | - Argonde C van Harten
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Pauline H B Kok
- Department of Ophthalmology, Bergman Clinics, Amsterdam, 1101 BM, The Netherlands
| | - Wiesje M van der Flier
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Yolande A L Pijnenburg
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Frederik Barkhof
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HZ, The Netherlands
- UCL Queen Square Institute of Neurology and Centre for Medical Image Computing, University College, London, WC1N 3BG, UK
| | - Elsmarieke van de Giessen
- Amsterdam Neuroscience, Brain Imaging, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Neurochemistry Laboratory, Laboratory Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081 HV, The Netherlands
| | - Femke H Bouwman
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Rik Ossenkoppele
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV, The Netherlands.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands.
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
| |
Collapse
|
16
|
Yoon D, Myong Y, Kim YG, Sim Y, Cho M, Oh BM, Kim S. Latent diffusion model-based MRI superresolution enhances mild cognitive impairment prognostication and Alzheimer's disease classification. Neuroimage 2024; 296:120663. [PMID: 38843963 DOI: 10.1016/j.neuroimage.2024.120663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/01/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024] Open
Abstract
INTRODUCTION Timely diagnosis and prognostication of Alzheimer's disease (AD) and mild cognitive impairment (MCI) are pivotal for effective intervention. Artificial intelligence (AI) in neuroradiology may aid in such appropriate diagnosis and prognostication. This study aimed to evaluate the potential of novel diffusion model-based AI for enhancing AD and MCI diagnosis through superresolution (SR) of brain magnetic resonance (MR) images. METHODS 1.5T brain MR scans of patients with AD or MCI and healthy controls (NC) from Alzheimer's Disease Neuroimaging Initiative 1 (ADNI1) were superresolved to 3T using a novel diffusion model-based generative AI (d3T*) and a convolutional neural network-based model (c3T*). Comparisons of image quality to actual 1.5T and 3T MRI were conducted based on signal-to-noise ratio (SNR), naturalness image quality evaluator (NIQE), and Blind/Referenceless Image Spatial Quality Evaluator (BRISQUE). Voxel-based volumetric analysis was then conducted to study whether 3T* images offered more accurate volumetry than 1.5T images. Binary and multiclass classifications of AD, MCI, and NC were conducted to evaluate whether 3T* images offered superior AD classification performance compared to actual 1.5T MRI. Moreover, CNN-based classifiers were used to predict conversion of MCI to AD, to evaluate the prognostication performance of 3T* images. The classification performances were evaluated using accuracy, sensitivity, specificity, F1 score, Matthews correlation coefficient (MCC), and area under the receiver-operating curves (AUROC). RESULTS Analysis of variance (ANOVA) detected significant differences in image quality among the 1.5T, c3T*, d3T*, and 3T groups across all metrics. Both c3T* and d3T* showed superior image quality compared to 1.5T MRI in NIQE and BRISQUE with statistical significance. While the hippocampal volumes measured in 3T* and 3T images were not significantly different, the hippocampal volume measured in 1.5T images showed significant difference. 3T*-based AD classifications showed superior performance across all performance metrics compared to 1.5T-based AD classification. Classification performance between d3T* and actual 3T was not significantly different. 3T* images offered superior accuracy in predicting the conversion of MCI to AD than 1.5T images did. CONCLUSIONS The diffusion model-based MRI SR enhances the resolution of brain MR images, significantly improving diagnostic and prognostic accuracy for AD and MCI. Superresolved 3T* images closely matched actual 3T MRIs in quality and volumetric accuracy, and notably improved the prediction performance of conversion from MCI to AD.
Collapse
Affiliation(s)
- Dan Yoon
- Interdisciplinary Program in Bioengineering, Seoul National University Graduate School, Seoul 03080, Republic of Korea
| | - Youho Myong
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Young Gyun Kim
- Interdisciplinary Program in Bioengineering, Seoul National University Graduate School, Seoul 03080, Republic of Korea
| | - Yongsik Sim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Minwoo Cho
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Medicine, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Byung-Mo Oh
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea.
| | - Sungwan Kim
- Interdisciplinary Program in Bioengineering, Seoul National University Graduate School, Seoul 03080, Republic of Korea; Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| |
Collapse
|
17
|
Baumeister H, Vogel JW, Insel PS, Kleineidam L, Wolfsgruber S, Stark M, Gellersen HM, Yakupov R, Schmid MC, Lüsebrink F, Brosseron F, Ziegler G, Freiesleben SD, Preis L, Schneider LS, Spruth EJ, Altenstein S, Lohse A, Fliessbach K, Vogt IR, Bartels C, Schott BH, Rostamzadeh A, Glanz W, Incesoy EI, Butryn M, Janowitz D, Rauchmann BS, Kilimann I, Goerss D, Munk MH, Hetzer S, Dechent P, Ewers M, Scheffler K, Wuestefeld A, Strandberg O, van Westen D, Mattsson-Carlgren N, Janelidze S, Stomrud E, Palmqvist S, Spottke A, Laske C, Teipel S, Perneczky R, Buerger K, Schneider A, Priller J, Peters O, Ramirez A, Wiltfang J, Heneka MT, Wagner M, Düzel E, Jessen F, Hansson O, Berron D. A generalizable data-driven model of atrophy heterogeneity and progression in memory clinic settings. Brain 2024; 147:2400-2413. [PMID: 38654513 PMCID: PMC11224599 DOI: 10.1093/brain/awae118] [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: 09/07/2023] [Revised: 02/02/2024] [Accepted: 03/03/2024] [Indexed: 04/26/2024] Open
Abstract
Memory clinic patients are a heterogeneous population representing various aetiologies of pathological ageing. It is not known whether divergent spatiotemporal progression patterns of brain atrophy, as previously described in Alzheimer's disease patients, are prevalent and clinically meaningful in this group of older adults. To uncover distinct atrophy subtypes, we applied the Subtype and Stage Inference (SuStaIn) algorithm to baseline structural MRI data from 813 participants enrolled in the DELCODE cohort (mean ± standard deviation, age = 70.67 ± 6.07 years, 52% females). Participants were cognitively unimpaired (n = 285) or fulfilled diagnostic criteria for subjective cognitive decline (n = 342), mild cognitive impairment (n = 118) or dementia of the Alzheimer's type (n = 68). Atrophy subtypes were compared in baseline demographics, fluid Alzheimer's disease biomarker levels, the Preclinical Alzheimer Cognitive Composite (PACC-5) as well as episodic memory and executive functioning. PACC-5 trajectories over up to 240 weeks were examined. To test whether baseline atrophy subtype and stage predicted clinical trajectories before manifest cognitive impairment, we analysed PACC-5 trajectories and mild cognitive impairment conversion rates of cognitively unimpaired participants and those with subjective cognitive decline. Limbic-predominant and hippocampal-sparing atrophy subtypes were identified. Limbic-predominant atrophy initially affected the medial temporal lobes, followed by further temporal regions and, finally, the remaining cortical regions. At baseline, this subtype was related to older age, more pathological Alzheimer's disease biomarker levels, APOE ε4 carriership and an amnestic cognitive impairment. Hippocampal-sparing atrophy initially occurred outside the temporal lobe, with the medial temporal lobe spared up to advanced atrophy stages. This atrophy pattern also affected individuals with positive Alzheimer's disease biomarkers and was associated with more generalized cognitive impairment. Limbic-predominant atrophy, in all participants and in only unimpaired participants, was linked to more negative longitudinal PACC-5 slopes than observed in participants without or with hippocampal-sparing atrophy and increased the risk of mild cognitive impairment conversion. SuStaIn modelling was repeated in a sample from the Swedish BioFINDER-2 cohort. Highly similar atrophy progression patterns and associated cognitive profiles were identified. Cross-cohort model generalizability, at both the subject and the group level, was excellent, indicating reliable performance in previously unseen data. The proposed model is a promising tool for capturing heterogeneity among older adults at early at-risk states for Alzheimer's disease in applied settings. The implementation of atrophy subtype- and stage-specific end points might increase the statistical power of pharmacological trials targeting early Alzheimer's disease.
Collapse
Affiliation(s)
- Hannah Baumeister
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Jacob W Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 222 42 Lund, Sweden
| | - Philip S Insel
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Melina Stark
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Helena M Gellersen
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Renat Yakupov
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Matthias C Schmid
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Institute for Medical Biometry, University Hospital Bonn, 53127 Bonn, Germany
| | - Falk Lüsebrink
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Frederic Brosseron
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Gabriel Ziegler
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Silka D Freiesleben
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Department of Psychiatry and Neurosciences, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Lukas Preis
- Department of Psychiatry and Neurosciences, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Luisa-Sophie Schneider
- Department of Psychiatry and Neurosciences, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Eike J Spruth
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Department of Psychiatry and Neurosciences, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Department of Psychiatry and Neurosciences, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Andrea Lohse
- Department of Psychiatry and Neurosciences, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Ina R Vogt
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Claudia Bartels
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Björn H Schott
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075 Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany
- Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Ayda Rostamzadeh
- Department of Psychiatry, Medical Faculty, University of Cologne, 50937 Cologne, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Enise I Incesoy
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, 39120 Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Michaela Butryn
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität, 80336 Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK
- Department of Neuroradiology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), 18147 Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, 18147 Rostock, Germany
| | - Doreen Goerss
- German Center for Neurodegenerative Diseases (DZNE), 18147 Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, 18147 Rostock, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany
| | - Stefan Hetzer
- Berlin Center for Advanced Neuroimaging, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University Göttingen, 37075 Göttingen, Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, 72076 Tübingen, Germany
| | - Anika Wuestefeld
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 222 42 Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 222 42 Lund, Sweden
| | - Danielle van Westen
- Diagnostic Radiology, Institution of Clinical Sciences Lund, Lund University, 211 84 Lund, Sweden
- Image and Function, Skåne University Hospital, 211 84 Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 222 42 Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, 211 84 Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 222 42 Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Neurology, University of Bonn, 53127 Bonn, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research, 72076 Tübingen, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), 18147 Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, 18147 Rostock, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität, 80336 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London SW7 2AZ, UK
| | - Katharina Buerger
- Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-Universität, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Department of Psychiatry and Neurosciences, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Department of Psychiatry and Psychotherapy, Technical University of Munich, 81675 Munich, Germany
- Centre for Clinical Brain Sciences, University of Edinburgh and UK DRI, Edinburgh EH16 4SB, UK
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Department of Psychiatry and Neurosciences, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, 50931 Cologne, Germany
- Department of Psychiatry & Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, The University of Texas at San Antonio, San Antonio, TX 78229, USA
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075 Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), 37075 Göttingen, Germany
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362, Belvaux, Luxembourg
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Psychiatry, Medical Faculty, University of Cologne, 50937 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, 205 02 Malmö, Sweden
| | - David Berron
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 222 42 Lund, Sweden
- Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke University Magdeburg, 39106 Magdeburg, Germany
| |
Collapse
|
18
|
Ma M, Liu Z, Zhao H, Zhang H, Ren J, Qu X. Polyoxometalates: metallodrug agents for combating amyloid aggregation. Natl Sci Rev 2024; 11:nwae226. [PMID: 39081537 PMCID: PMC11288190 DOI: 10.1093/nsr/nwae226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/02/2024] [Accepted: 06/19/2024] [Indexed: 08/02/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease that affects ∼50 million people globally. The accumulation of amyloid-β (Aβ) plaques, a predominant pathological feature of AD, plays a crucial role in AD pathogenesis. In this respect, Aβ has been regarded as a highly promising therapeutic target for AD treatment. Polyoxometalates (POMs) are a novel class of metallodrugs being developed as modulators of Aβ aggregation, owing to their negative charge, polarity, and three-dimensional structure. Unlike traditional discrete inorganic complexes, POMs contain tens to hundreds of metal atoms, showcasing remarkable tunability and diversity in nuclearities, sizes, and shapes. The easily adjustable and structurally variable nature of POMs allows for their favorable interactions with Aβ. This mini-review presents a balanced overview of recent progress in using POMs to mitigate amyloidosis. Clear correlations between anti-amyloid activities and structural features of POMs are also elaborated in detail. Finally, we discuss the current challenges and future prospects of POMs in combating AD.
Collapse
Affiliation(s)
- Mengmeng Ma
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Zhenqi Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Huisi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Haochen Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
19
|
Tranfa M, Lorenzini L, Collij LE, Vállez García D, Ingala S, Pontillo G, Pieperhoff L, Maranzano A, Wolz R, Haller S, Blennow K, Frisoni G, Sudre CH, Chételat G, Ewers M, Payoux P, Waldman A, Martinez‐Lage P, Schwarz AJ, Ritchie CW, Wardlaw JM, Gispert JD, Brunetti A, Mutsaerts HJMM, Wink AM, Barkhof F. Alzheimer's Disease and Small Vessel Disease Differentially Affect White Matter Microstructure. Ann Clin Transl Neurol 2024; 11:1541-1556. [PMID: 38757392 PMCID: PMC11187968 DOI: 10.1002/acn3.52071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 05/18/2024] Open
Abstract
OBJECTIVE Alzheimer's disease (AD) and cerebral small vessel disease (cSVD), the two most common causes of dementia, are characterized by white matter (WM) alterations diverging from the physiological changes occurring in healthy aging. Diffusion tensor imaging (DTI) is a valuable tool to quantify WM integrity non-invasively and identify the determinants of such alterations. Here, we investigated main effects and interactions of AD pathology, APOE-ε4, cSVD, and cardiovascular risk on spatial patterns of WM alterations in non-demented older adults. METHODS Within the prospective European Prevention of Alzheimer's Dementia study, we selected 606 participants (64.9 ± 7.2 years, 376 females) with baseline cerebrospinal fluid samples of amyloid β1-42 and p-Tau181 and MRI scans, including DTI scans. Longitudinal scans (mean follow-up time = 1.3 ± 0.5 years) were obtained in a subset (n = 223). WM integrity was assessed by extracting fractional anisotropy and mean diffusivity in relevant tracts. To identify the determinants of WM disruption, we performed a multimodel inference to identify the best linear mixed-effects model for each tract. RESULTS AD pathology, APOE-ε4, cSVD burden, and cardiovascular risk were all associated with WM integrity within several tracts. While limbic tracts were mainly impacted by AD pathology and APOE-ε4, commissural, associative, and projection tract integrity was more related to cSVD burden and cardiovascular risk. AD pathology and cSVD did not show any significant interaction effect. INTERPRETATION Our results suggest that AD pathology and cSVD exert independent and spatially different effects on WM microstructure, supporting the role of DTI in disease monitoring and suggesting independent targets for preventive medicine approaches.
Collapse
Affiliation(s)
- Mario Tranfa
- Department of Advanced Biomedical SciencesUniversity “Federico II”NaplesItaly
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentreVrije UniversiteitAmsterdamThe Netherlands
| | - Luigi Lorenzini
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentreVrije UniversiteitAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain ImagingAmsterdamThe Netherlands
| | - Lyduine E. Collij
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentreVrije UniversiteitAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain ImagingAmsterdamThe Netherlands
- Clinical Memory Research Unit, Department of Clinical SciencesLund UniversityMalmöSweden
| | - David Vállez García
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentreVrije UniversiteitAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain ImagingAmsterdamThe Netherlands
| | - Silvia Ingala
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentreVrije UniversiteitAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain ImagingAmsterdamThe Netherlands
- Department of RadiologyCopenhagen University Hospital RigshospitaletCopenhagenDenmark
- Cerebriu A/SCopenhagenDenmark
| | - Giuseppe Pontillo
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentreVrije UniversiteitAmsterdamThe Netherlands
| | - Leonard Pieperhoff
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentreVrije UniversiteitAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain ImagingAmsterdamThe Netherlands
| | - Alessio Maranzano
- Department of Neurology and Laboratory of NeuroscienceIRCCS Istituto Auxologico ItalianoMilanItaly
| | | | - Sven Haller
- CIMC ‐ Centre d'Imagerie Médicale de CornavinGenevaSwitzerland
- Department of Surgical Sciences, RadiologyUppsala UniversityUppsalaSweden
- Department of Radiology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Giovanni Frisoni
- Laboratory Alzheimer's Neuroimaging & EpidemiologyIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- University Hospitals and University of GenevaGenevaSwitzerland
| | - Carole H. Sudre
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of Medical Physics and Biomedical Engineering, Centre for Medical Image Computing (CMIC)University College London (UCL)LondonUK
- MRC Unit for Lifelong Health & Ageing at UCLUniversity College LondonLondonUK
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
| | - Gael Chételat
- Normandie Univ, Unicaen, Inserm, U1237, PhIND “Physiopathology and Imaging of Neurological Disorders”, institut Blood‐and‐Brain @ Caen‐Normandie, CyceronUniversité de NormandieCaenFrance
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| | - Pierre Payoux
- Department of Nuclear MedicineToulouse University HospitalToulouseFrance
- ToNIC, Toulouse NeuroImaging CenterUniversity of Toulouse, Inserm, UPSToulouseFrance
| | - Adam Waldman
- Centre for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- Department of MedicineImperial College LondonLondonUK
| | - Pablo Martinez‐Lage
- Centro de Investigación y Terapias Avanzadas, Neurología, CITA‐Alzheimer FoundationSan SebastiánSpain
| | - Adam J. Schwarz
- Takeda Pharmaceuticals, Ltd.CambridgeMassachusettsUSA
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Craig W. Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, Outpatient Department 2, Western General HospitalUniversity of EdinburghEdinburghUK
- Brain Health ScotlandEdinburghUK
| | - Joanna M. Wardlaw
- Centre for Clinical Brain SciencesThe University of EdinburghEdinburghUK
- UK Dementia Research Institute Centre at the University of EdinburghEdinburghUK
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- CIBER Bioingeniería, Biomateriales y Nanomedicina (CIBER‐BBN)MadridSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- Universitat Pompeu FabraBarcelonaSpain
| | - Arturo Brunetti
- Department of Advanced Biomedical SciencesUniversity “Federico II”NaplesItaly
| | - Henk J. M. M. Mutsaerts
- Amsterdam Neuroscience, Brain ImagingAmsterdamThe Netherlands
- Ghent Institute for Functional and Metabolic Imaging (GIfMI)Ghent UniversityGhentBelgium
| | - Alle Meije Wink
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentreVrije UniversiteitAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain ImagingAmsterdamThe Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical CentreVrije UniversiteitAmsterdamThe Netherlands
- Institute of Neurology and Healthcare EngineeringUniversity College LondonLondonUK
| |
Collapse
|
20
|
Noda T, Aschauer DF, Chambers AR, Seiler JPH, Rumpel S. Representational maps in the brain: concepts, approaches, and applications. Front Cell Neurosci 2024; 18:1366200. [PMID: 38584779 PMCID: PMC10995314 DOI: 10.3389/fncel.2024.1366200] [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: 01/05/2024] [Accepted: 03/08/2024] [Indexed: 04/09/2024] Open
Abstract
Neural systems have evolved to process sensory stimuli in a way that allows for efficient and adaptive behavior in a complex environment. Recent technological advances enable us to investigate sensory processing in animal models by simultaneously recording the activity of large populations of neurons with single-cell resolution, yielding high-dimensional datasets. In this review, we discuss concepts and approaches for assessing the population-level representation of sensory stimuli in the form of a representational map. In such a map, not only are the identities of stimuli distinctly represented, but their relational similarity is also mapped onto the space of neuronal activity. We highlight example studies in which the structure of representational maps in the brain are estimated from recordings in humans as well as animals and compare their methodological approaches. Finally, we integrate these aspects and provide an outlook for how the concept of representational maps could be applied to various fields in basic and clinical neuroscience.
Collapse
Affiliation(s)
- Takahiro Noda
- Institute of Physiology, Focus Program Translational Neurosciences, University Medical Center, Johannes Gutenberg University-Mainz, Mainz, Germany
| | - Dominik F. Aschauer
- Institute of Physiology, Focus Program Translational Neurosciences, University Medical Center, Johannes Gutenberg University-Mainz, Mainz, Germany
| | - Anna R. Chambers
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, United States
- Eaton Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Boston, MA, United States
| | - Johannes P.-H. Seiler
- Institute of Physiology, Focus Program Translational Neurosciences, University Medical Center, Johannes Gutenberg University-Mainz, Mainz, Germany
| | - Simon Rumpel
- Institute of Physiology, Focus Program Translational Neurosciences, University Medical Center, Johannes Gutenberg University-Mainz, Mainz, Germany
| |
Collapse
|
21
|
Pauwels EK, Boer GJ. Alzheimer's Disease: A Suitable Case for Treatment with Precision Medicine? Med Princ Pract 2024; 33:000538251. [PMID: 38471490 PMCID: PMC11324226 DOI: 10.1159/000538251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/06/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most common cause of neurodegenerative impairment in elderly people. Clinical characteristics include short-term memory loss, confusion, hallucination, agitation, and behavioural disturbance. Owing to evolving research in biomarkers AD can be discovered at early onset, but the disease is currently considered a continuum, which suggests that pharmacotherapy is most efficacious in the preclinical phase, possibly 15 - 20 years before discernible onset. Present developments in AD therapy aim to respond to this understanding and go beyond the drug families that relieve clinical symptoms. Another important factor in this development is the emergence of precision medicine that aims to tailor treatment to specific patients or patient subgroups. This relatively new platform would categorize AD patients on the basis of parameters like clinical aspects, brain imaging, genetic profiling, clinical genetics and epidemiological factors. This review enlarges on recent progress in the design and clinical use of antisense molecules, antibodies, antioxidants, small molecules and gene editing to stop AD progress and possibly reverse the disease on the basis of relevant biomarkers.
Collapse
Affiliation(s)
- Ernest K.J. Pauwels
- Leiden University and Leiden University Medical Center, Leiden, The Netherlands
| | - Gerard J. Boer
- Netherlands Institute for Brain Research, Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Chudzik A, Śledzianowski A, Przybyszewski AW. Machine Learning and Digital Biomarkers Can Detect Early Stages of Neurodegenerative Diseases. SENSORS (BASEL, SWITZERLAND) 2024; 24:1572. [PMID: 38475108 PMCID: PMC10934426 DOI: 10.3390/s24051572] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer's Disease (AD) and Parkinson's Disease (PD) are devastating conditions that can develop without noticeable symptoms, causing irreversible damage to neurons before any signs become clinically evident. NDs are a major cause of disability and mortality worldwide. Currently, there are no cures or treatments to halt their progression. Therefore, the development of early detection methods is urgently needed to delay neuronal loss as soon as possible. Despite advancements in Medtech, the early diagnosis of NDs remains a challenge at the intersection of medical, IT, and regulatory fields. Thus, this review explores "digital biomarkers" (tools designed for remote neurocognitive data collection and AI analysis) as a potential solution. The review summarizes that recent studies combining AI with digital biomarkers suggest the possibility of identifying pre-symptomatic indicators of NDs. For instance, research utilizing convolutional neural networks for eye tracking has achieved significant diagnostic accuracies. ROC-AUC scores reached up to 0.88, indicating high model performance in differentiating between PD patients and healthy controls. Similarly, advancements in facial expression analysis through tools have demonstrated significant potential in detecting emotional changes in ND patients, with some models reaching an accuracy of 0.89 and a precision of 0.85. This review follows a structured approach to article selection, starting with a comprehensive database search and culminating in a rigorous quality assessment and meaning for NDs of the different methods. The process is visualized in 10 tables with 54 parameters describing different approaches and their consequences for understanding various mechanisms in ND changes. However, these methods also face challenges related to data accuracy and privacy concerns. To address these issues, this review proposes strategies that emphasize the need for rigorous validation and rapid integration into clinical practice. Such integration could transform ND diagnostics, making early detection tools more cost-effective and globally accessible. In conclusion, this review underscores the urgent need to incorporate validated digital health tools into mainstream medical practice. This integration could indicate a new era in the early diagnosis of neurodegenerative diseases, potentially altering the trajectory of these conditions for millions worldwide. Thus, by highlighting specific and statistically significant findings, this review demonstrates the current progress in this field and the potential impact of these advancements on the global management of NDs.
Collapse
Affiliation(s)
- Artur Chudzik
- Polish-Japanese Academy of Information Technology, Faculty of Computer Science, 86 Koszykowa Street, 02-008 Warsaw, Poland; (A.C.); (A.Ś.)
| | - Albert Śledzianowski
- Polish-Japanese Academy of Information Technology, Faculty of Computer Science, 86 Koszykowa Street, 02-008 Warsaw, Poland; (A.C.); (A.Ś.)
| | - Andrzej W. Przybyszewski
- Polish-Japanese Academy of Information Technology, Faculty of Computer Science, 86 Koszykowa Street, 02-008 Warsaw, Poland; (A.C.); (A.Ś.)
- UMass Chan Medical School, Department of Neurology, 65 Lake Avenue, Worcester, MA 01655, USA
| |
Collapse
|
23
|
Michno W, Bowman A, Jha D, Minta K, Ge J, Koutarapu S, Zetterberg H, Blennow K, Lashley T, Heeren RMA, Hanrieder J. Spatial Neurolipidomics at the Single Amyloid-β Plaque Level in Postmortem Human Alzheimer's Disease Brain. ACS Chem Neurosci 2024; 15:877-888. [PMID: 38299453 PMCID: PMC10885149 DOI: 10.1021/acschemneuro.4c00006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/02/2024] Open
Abstract
Lipid dysregulations have been critically implicated in Alzheimer's disease (AD) pathology. Chemical analysis of amyloid-β (Aβ) plaque pathology in transgenic AD mouse models has demonstrated alterations in the microenvironment in the direct proximity of Aβ plaque pathology. In mouse studies, differences in lipid patterns linked to structural polymorphism among Aβ pathology, such as diffuse, immature, and mature fibrillary aggregates, have also been reported. To date, no comprehensive analysis of neuronal lipid microenvironment changes in human AD tissue has been performed. Here, for the first time, we leverage matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) through a high-speed and spatial resolution commercial time-of-light instrument, as well as a high-mass-resolution in-house-developed orbitrap system to characterize the lipid microenvironment in postmortem human brain tissue from AD patients carrying Presenilin 1 mutations (PSEN1) that lead to familial forms of AD (fAD). Interrogation of the spatially resolved MSI data on a single Aβ plaque allowed us to verify nearly 40 sphingolipid and phospholipid species from diverse subclasses being enriched and depleted, in relation to the Aβ deposits. This included monosialo-gangliosides (GM), ceramide monohexosides (HexCer), ceramide-1-phosphates (CerP), ceramide phosphoethanolamine conjugates (PE-Cer), sulfatides (ST), as well as phosphatidylinositols (PI), phosphatidylethanolamines (PE), and phosphatidic acid (PA) species (including Lyso-forms). Indeed, many of the sphingolipid species overlap with the species previously seen in transgenic AD mouse models. Interestingly, in comparison to the animal studies, we observed an increased level of localization of PE and PI species containing arachidonic acid (AA). These findings are highly relevant, demonstrating for the first time Aβ plaque pathology-related alteration in the lipid microenvironment in humans. They provide a basis for the development of potential lipid biomarkers for AD characterization and insight into human-specific molecular pathway alterations.
Collapse
Affiliation(s)
- Wojciech Michno
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
- Department
of Neuroscience, Physiology and Pharmacology, University College London, London WC1E6BT, United
Kingdom
- Department
of Public Health and Caring Sciences, Uppsala
University, Uppsala 75237, Sweden
- Science
for Life Laboratory (SciLife), Uppsala University, Uppsala 75237, Sweden
| | - Andrew Bowman
- Maastricht
MultiModal Molecular Imaging Institute (M4I), Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Durga Jha
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
| | - Karolina Minta
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
| | - Junyue Ge
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
| | - Srinivas Koutarapu
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
| | - Henrik Zetterberg
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
- Clinical
Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal 43180, Sweden
- Department
of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, United
Kingdom
- UK
Dementia Research Institute at UCL, London WC1E 6BT, United Kingdom
- Hong
Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong 999077, China
- Wisconsin
Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University
of Wisconsin-Madison, Madison, Wisconsin 53726, United States
| | - Kaj Blennow
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
- Clinical
Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal 43180, Sweden
- Paris Brain
Institute, ICM, Pitié-Salpêtrière
Hospital, Sorbonne University, Paris 75005, France
- Neurodegenerative
Disorder Research Center, Division of Life Sciences
and Medicine, Department of Neurology, Institute on Aging and Brain
Disorders, University of Science and Technology
of China and First Affiliated Hospital of USTC, Hefei 230001, P. R. China
| | - Tammaryn Lashley
- Department
of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, United
Kingdom
- Queen Square Brain Bank for Neurological
Disorders, Department of
Clinical and Movement Neurosciences, Institute of Neurology, University College London, London WC1N 1PJ, United Kingdom
| | - Ron M. A. Heeren
- Maastricht
MultiModal Molecular Imaging Institute (M4I), Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Jörg Hanrieder
- Department
of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal 43180, Sweden
- Clinical
Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal 43180, Sweden
- Department
of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queen Square, London WC1N 3BG, United
Kingdom
- Science for Life
Laboratory (SciLife), University of Gothenburg, Gothenburg 40530, Sweden
| |
Collapse
|
24
|
Wang C, Tachimori H, Yamaguchi H, Sekiguchi A, Li Y, Yamashita Y. A multimodal deep learning approach for the prediction of cognitive decline and its effectiveness in clinical trials for Alzheimer's disease. Transl Psychiatry 2024; 14:105. [PMID: 38383536 PMCID: PMC10882004 DOI: 10.1038/s41398-024-02819-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024] Open
Abstract
Alzheimer's disease is one of the most important health-care challenges in the world. For decades, numerous efforts have been made to develop therapeutics for Alzheimer's disease, but most clinical trials have failed to show significant treatment effects on slowing or halting cognitive decline. Among several challenges in such trials, one recently noticed but unsolved is biased allocation of fast and slow cognitive decliners to treatment and placebo groups during randomization caused by the large individual variation in the speed of cognitive decline. This allocation bias directly results in either over- or underestimation of the treatment effect from the outcome of the trial. In this study, we propose a stratified randomization method using the degree of cognitive decline predicted by an artificial intelligence model as a stratification index to suppress the allocation bias in randomization and evaluate its effectiveness by simulation using ADNI data set.
Collapse
Affiliation(s)
- Caihua Wang
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, Ashigarakami-gun, Kanagawa, Japan
| | - Hisateru Tachimori
- Department of Information Medicine, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Endowed Course for Health System Innovation, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Yamaguchi
- Department of Information Medicine, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Psychiatry, Yokohama City University School of Medicine, Yokohama, Japan
| | - Atsushi Sekiguchi
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuanzhong Li
- Bio Science & Engineering Laboratories, FUJIFILM Corporation, Ashigarakami-gun, Kanagawa, Japan.
| | - Yuichi Yamashita
- Department of Information Medicine, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
25
|
Koike R, Soeda Y, Kasai A, Fujioka Y, Ishigaki S, Yamanaka A, Takaichi Y, Chambers JK, Uchida K, Watanabe H, Takashima A. Path integration deficits are associated with phosphorylated tau accumulation in the entorhinal cortex. Brain Commun 2024; 6:fcad359. [PMID: 38347945 PMCID: PMC10859636 DOI: 10.1093/braincomms/fcad359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 11/14/2023] [Accepted: 01/04/2024] [Indexed: 02/15/2024] Open
Abstract
Alzheimer's disease is a devastating disease that is accompanied by dementia, and its incidence increases with age. However, no interventions have exhibited clear therapeutic effects. We aimed to develop and characterize behavioural tasks that allow the earlier identification of signs preceding dementia that would facilitate the development of preventative and therapeutic interventions for Alzheimer's disease. To this end, we developed a 3D virtual reality task sensitive to the activity of grid cells in the entorhinal cortex, which is the region that first exhibits neurofibrillary tangles in Alzheimer's disease. We investigated path integration (assessed by error distance) in a spatial navigation task sensitive to grid cells in the entorhinal cortex in 177 volunteers, aged 20-89 years, who did not have self-reported dementia. While place memory was intact even in old age, path integration deteriorated with increasing age. To investigate the relationship between neurofibrillary tangles in the entorhinal cortex and path integration deficit, we examined a mouse model of tauopathy (P301S mutant tau-overexpressing mice; PS19 mice). At 6 months of age, PS19 mice showed a significant accumulation of phosphorylated tau only in the entorhinal cortex, associated with impaired path integration without impairments in spatial cognition. These data are consistent with the idea that path integration deficit is caused by the accumulation of phosphorylated tau in the entorhinal cortex. This method may allow the early identification of individuals likely to develop Alzheimer's disease.
Collapse
Affiliation(s)
- Riki Koike
- Laboratory for Alzheimer’s Disease, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo 171-8588, Japan
| | - Yoshiyuki Soeda
- Laboratory for Alzheimer’s Disease, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo 171-8588, Japan
| | - Atsushi Kasai
- Deapartment of Research and Development, MIG (Medical Innovation Group) Inc, Shibuya, Tokyo 150-0031, Japan
| | - Yusuke Fujioka
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Shinsuke Ishigaki
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Yuta Takaichi
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - James K Chambers
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kazuyuki Uchida
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Hirohisa Watanabe
- Department of Neurology, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Akihiko Takashima
- Laboratory for Alzheimer’s Disease, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo 171-8588, Japan
| |
Collapse
|
26
|
Woo MS, Tissot C, Lantero‐Rodriguez J, Snellman A, Therriault J, Rahmouni N, Macedo AC, Servaes S, Wang Y, Arias JF, Hosseini SA, Chamoun M, Lussier FZ, Benedet AL, Ashton NJ, Karikari TK, Triana‐Baltzer G, Kolb HC, Stevenson J, Mayer C, Kobayashi E, Massarweh G, Friese MA, Pascoal TA, Gauthier S, Zetterberg H, Blennow K, Rosa‐Neto P. Plasma pTau-217 and N-terminal tau (NTA) enhance sensitivity to identify tau PET positivity in amyloid-β positive individuals. Alzheimers Dement 2024; 20:1166-1174. [PMID: 37920945 PMCID: PMC10916953 DOI: 10.1002/alz.13528] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION We set out to identify tau PET-positive (A+T+) individuals among amyloid-beta (Aβ) positive participants using plasma biomarkers. METHODS In this cross-sectional study we assessed 234 participants across the AD continuum who were evaluated by amyloid PET with [18 F]AZD4694 and tau-PET with [18 F]MK6240 and measured plasma levels of total tau, pTau-181, pTau-217, pTau-231, and N-terminal tau (NTA-tau). We evaluated the performances of plasma biomarkers to predict tau positivity in Aβ+ individuals. RESULTS Highest associations with tau positivity in Aβ+ individuals were found for plasma pTau-217 (AUC [CI95% ] = 0.89 [0.82, 0.96]) and NTA-tau (AUC [CI95% ] = 0.88 [0.91, 0.95]). Combining pTau-217 and NTA-tau resulted in the strongest agreement (Cohen's Kappa = 0.74, CI95% = 0.57/0.90, sensitivity = 92%, specificity = 81%) with PET for classifying tau positivity. DISCUSSION The potential for identifying tau accumulation in later Braak stages will be useful for patient stratification and prognostication in treatment trials and in clinical practice. HIGHLIGHTS We found that in a cohort without pre-selection pTau-181, pTau-217, and NTA-tau showed the highest association with tau PET positivity. We found that in Aβ+ individuals pTau-217 and NTA-tau showed the highest association with tau PET positivity. Combining pTau-217 and NTA-tau resulted in the strongest agreement with the tau PET-based classification.
Collapse
|
27
|
Wang H, Yan X, Zhang Y, Wang P, Li J, Zhang X. Mitophagy in Alzheimer's Disease: A Bibliometric Analysis from 2007 to 2022. J Alzheimers Dis Rep 2024; 8:101-128. [PMID: 38312534 PMCID: PMC10836605 DOI: 10.3233/adr-230139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/15/2023] [Indexed: 02/06/2024] Open
Abstract
Background The investigation of mitophagy in Alzheimer's disease (AD) remains relatively underexplored in bibliometric analysis. Objective To delve into the progress of mitophagy, offering a comprehensive overview of research trends and frontiers for researchers. Methods Basic bibliometric information, targets, and target-drug-clinical trial-disease extracted from publications identified in the Web of Science Core Collection from 2007 to 2022 were assessed using bibliometric software. Results The study encompassed 5,146 publications, displaying a consistent 16-year upward trajectory. The United States emerged as the foremost contributor in publications, with the Journal of Alzheimer's Disease being the most prolific journal. P. Hemachandra Reddy, George Perry, and Xiongwei Zhu are the top 3 most prolific authors. PINK1 and Parkin exhibited an upward trend in the last 6 years. Keywords (e.g., insulin, aging, epilepsy, tauopathy, and mitochondrial quality control) have recently emerged as focal points of interest within the past 3 years. "Mitochondrial dysfunction" is among the top terms in disease clustering. The top 10 drugs/molecules (e.g., curcumin, insulin, and melatonin) were summarized, accompanied by their clinical trials and related targets. Conclusions This study presents a comprehensive overview of the mitophagy research landscape in AD over the past 16 years, underscoring mitophagy as an emerging molecular mechanism and a crucial focal point for potential drug in AD. This study pioneers the inclusion of targets and their correlations with drugs, clinical trials, and diseases in bibliometric analysis, providing valuable insights and inspiration for scholars and readers of JADR interested in understanding the potential mechanisms and clinical trials in AD.
Collapse
Affiliation(s)
- Hongqi Wang
- Department of Neurology, Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, China
- Department of Anatomy, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaodong Yan
- Department of Anatomy, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yiming Zhang
- Department of Anatomy, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Peifu Wang
- Department of Neurology, Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, China
| | - Jilai Li
- Department of Neurology, Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, China
| | - Xia Zhang
- Department of Neurology, Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, China
- Department of Anatomy, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Ben-Shlomo Y, Darweesh S, Llibre-Guerra J, Marras C, San Luciano M, Tanner C. The epidemiology of Parkinson's disease. Lancet 2024; 403:283-292. [PMID: 38245248 PMCID: PMC11123577 DOI: 10.1016/s0140-6736(23)01419-8] [Citation(s) in RCA: 218] [Impact Index Per Article: 218.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 04/26/2023] [Accepted: 07/05/2023] [Indexed: 01/22/2024]
Abstract
The epidemiology of Parkinson's disease shows marked variations in time, geography, ethnicity, age, and sex. Internationally, prevalence has increased over and above demographic changes. There are several potential reasons for this increase, including the decline in other competing causes of death. Whether incidence is increasing, especially in women or in many low-income and middle-income countries where there is a shortage of high-quality data, is less certain. Parkinson's disease is more common in older people and men, and a variety of environmental factors have been suggested to explain why, including exposure to neurotoxic agents. Within countries, there appear to be ethnic differences in disease risk, although these differences might reflect differential access to health care. The cause of Parkinson's disease is multifactorial, and involves genetic and environmental factors. Both risk factors (eg, pesticides) and protective factors (eg, physical activity and tendency to smoke) have been postulated to have a role in Parkinson's disease, although elucidating causality is complicated by the long prodromal period. Following the establishment of public health strategies to prevent cardiovascular diseases and some cancers, chronic neurodegenerative diseases such as Parkinson's disease and dementia are gaining a deserved higher priority. Multipronged prevention strategies are required that tackle population-based primary prevention, high-risk targeted secondary prevention, and Parkinson's disease-modifying therapies for tertiary prevention. Future international collaborations will be required to triangulate evidence from basic, applied, and epidemiological research, thereby enhancing the understanding and prevention of Parkinson's disease at a global level.
Collapse
Affiliation(s)
- Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Sirwan Darweesh
- Centre of Expertise for Parkinson and Movement Disorders, Department of Neurology, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, Netherlands
| | | | - Connie Marras
- The Edmond J Safra Program in Parkinson's Disease, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
| | - Marta San Luciano
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Caroline Tanner
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
29
|
Huang Y, Chen H, Gao M, Lv X, Pang T, Rong S, Xu X, Yuan C. Self- and interviewer-reported cognitive problems in relation to cognitive decline and dementia: results from two prospective studies. BMC Med 2024; 22:23. [PMID: 38229039 PMCID: PMC10792911 DOI: 10.1186/s12916-023-03147-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/01/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Little is known regarding the association of interviewer-reported cognitive problems (ICP) with age-related cognitive decline. We aimed to investigate the independent associations of ICP and the combined associations of ICP and self-reported cognitive problems (SCP) with subsequent cognitive decline and dementia in two prospective cohort studies. METHODS We included 10,976 Chinese (age = 57.7 ± 8.7) and 40,499 European (age = 64.6 ± 9.4) adults without dementia from the China Health and Retirement Longitudinal Study (CHARLS) and the Survey of Health, Ageing, and Retirement in Europe (SHARE). Self-rated memory (5-point scale) and interviewer-rated frequencies of asking for clarification (6-point scale) were used to define SCP and ICP (dichotomized). Outcomes included objective cognitive test scores (z-score transformation) and incident dementia. Generalized estimating equation models were performed to evaluate mean differences in objective cognitive decline. Logistic and Cox regression models were used to estimate the relative risk of dementia. Results from two cohorts were pooled using the random-effects models. RESULTS ICP was associated with faster cognitive decline in CHARLS (βCHARLS = -0.025 [-0.044, -0.006] z-score/year). ICP and SCP were also independently associated with higher risk of dementia in two cohorts (pooled relative risk for SCP = 1.73 [1.30, 2.29]; pooled relative risk for ICP = 1.40 [1.10, 1.79]). In the joint analysis, participants with coexistence of SCP and ICP had the fastest cognitive decline (βCHARLS = -0.051 [-0.080, -0.021]; βSHARE = -0.024 [-0.043, -0.004]; pooled β = -0.035 [-0.061, -0.009] z-score/year) and highest risk of dementia (ORCHARLS = 1.77 [1.42, 2.20]; HRSHARE = 2.94 [2.42, 3.59]; pooled relative risk = 2.29 [1.38, 3.77]). CONCLUSIONS The study suggested that interviewer-reported cognitive problems may be early indicators of cognitive decline and dementia in middle-aged and older adults. A combination of self- and interviewer-reported cognitive problems showed the strongest associations with cognitive decline and dementia.
Collapse
Affiliation(s)
- Yuhui Huang
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hui Chen
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mengyan Gao
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaozhen Lv
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders, NHC Key Laboratory of Mental Health (Peking University), Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
| | - Ting Pang
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuang Rong
- Department of Nutrition and Food Hygiene, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Xin Xu
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Memory, Ageing and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Changzheng Yuan
- School of Public Health, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
30
|
Chandler JM, Ye W, Mi X, Doty EG, Johnston JA. Potential Impact of Slowing Disease Progression in Early Symptomatic Alzheimer's Disease on Patient Quality of Life, Caregiver Time, and Total Societal Costs: Estimates Based on Findings from GERAS-US Study. J Alzheimers Dis 2024; 100:563-578. [PMID: 38875031 PMCID: PMC11307086 DOI: 10.3233/jad-231166] [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] [Accepted: 05/08/2024] [Indexed: 06/16/2024]
Abstract
Background Impact of Alzheimer's disease (AD) progression on patient health-related quality of life (HRQoL), caregiver time, and societal costs is not well characterized in early AD. Objective To assess the association of change in cognition with HRQoL, caregiver time, and societal costs over 36 months, and estimate the impact of slowing disease progression on these outcomes. Methods This post-hoc analysis included patients with amyloid-positive mild cognitive impairment (MCI) and mild AD dementia (MILD AD) from the 36-month GERAS-US study. Disease progression was assessed using the Mini-Mental State Examination score. Change in outcomes associated with slowing AD progression was estimated using coefficients from generalized linear models. Results At baseline, 300 patients had MCI and 317 had MILD AD. Observed natural progression over 36 months was associated with: 5.1 point decline in the Bath Assessment of Subjective Quality of Life in Dementia (BASQID) score (for HRQoL), increase in 1,050 hours of total caregiver time, and $8,504 total societal costs for MCI; 6.6 point decline in the BASQID score, increase in 1,929 hours of total caregiver time, and $12,795 total societal costs for MILD AD per person. Slowing AD progression by 30% could result in per person savings in HRQoL decline, total caregiver time, and total societal costs: for MCI: 1.5 points, 315 hours, and $2,638; for MILD AD: 2.0 points, 579 hours, and $3,974. Conclusions Slowing AD progression over 36 months could slow decline in HRQoL and save caregiver time and societal cost in patients with MCI and MILD AD.
Collapse
Affiliation(s)
| | - Wenyu Ye
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Xiaojuan Mi
- TechData Services Company, King of Prussia, PA, USA
| | | | | |
Collapse
|
31
|
Zhang W, Zhang L, Lv M, Fu Y, Meng X, Wang M, Wang H. Advances in Developing Small Molecule Drugs for Alzheimer's Disease. Curr Alzheimer Res 2024; 21:221-231. [PMID: 39136501 DOI: 10.2174/0115672050329828240805074938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 10/25/2024]
Abstract
Alzheimer's disease (AD) is the most common type of dementia among middle-aged and elderly individuals. Accelerating the prevention and treatment of AD has become an urgent problem. New technology including Computer-aided drug design (CADD) can effectively reduce the medication cost for patients with AD, reduce the cost of living, and improve the quality of life of patients, providing new ideas for treating AD. This paper reviews the pathogenesis of AD, the latest developments in CADD and other small-molecule docking technologies for drug discovery and development; the current research status of small-molecule compounds for AD at home and abroad from the perspective of drug action targets; the future of AD drug development.
Collapse
Affiliation(s)
- Wei Zhang
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Liujie Zhang
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Mingti Lv
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Yun Fu
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Xiaowen Meng
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Mingyong Wang
- School of Medical Technology, Xinxiang Medical University, Xinxiang, China
- Department of Medical Technology, Shangqiu Medical College, Shangqiu, Henan, China
| | - Hecheng Wang
- School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
- School of Life and Pharmaceutical Science, Dalin University of Technology, Panjin, China
| |
Collapse
|
32
|
Irwin C, Tjandra D, Hu C, Aggarwal V, Lienau A, Giordani B, Wiens J, Migrino RQ. Predicting 5-year dementia conversion in veterans with mild cognitive impairment. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12572. [PMID: 38545542 PMCID: PMC10965752 DOI: 10.1002/dad2.12572] [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: 08/31/2023] [Revised: 01/29/2024] [Accepted: 02/18/2024] [Indexed: 11/11/2024]
Abstract
INTRODUCTION Identifying mild cognitive impairment (MCI) patients at risk for dementia could facilitate early interventions. Using electronic health records (EHRs), we developed a model to predict MCI to all-cause dementia (ACD) conversion at 5 years. METHODS Cox proportional hazards model was used to identify predictors of ACD conversion from EHR data in veterans with MCI. Model performance (area under the receiver operating characteristic curve [AUC] and Brier score) was evaluated on a held-out data subset. RESULTS Of 59,782 MCI patients, 15,420 (25.8%) converted to ACD. The model had good discriminative performance (AUC 0.73 [95% confidence interval (CI) 0.72-0.74]), and calibration (Brier score 0.18 [95% CI 0.17-0.18]). Age, stroke, cerebrovascular disease, myocardial infarction, hypertension, and diabetes were risk factors, while body mass index, alcohol abuse, and sleep apnea were protective factors. DISCUSSION EHR-based prediction model had good performance in identifying 5-year MCI to ACD conversion and has potential to assist triaging of at-risk patients. Highlights Of 59,782 veterans with mild cognitive impairment (MCI), 15,420 (25.8%) converted to all-cause dementia within 5 years.Electronic health record prediction models demonstrated good performance (area under the receiver operating characteristic curve 0.73; Brier 0.18).Age and vascular-related morbidities were predictors of dementia conversion.Synthetic data was comparable to real data in modeling MCI to dementia conversion. Key Points An electronic health record-based model using demographic and co-morbidity data had good performance in identifying veterans who convert from mild cognitive impairment (MCI) to all-cause dementia (ACD) within 5 years.Increased age, stroke, cerebrovascular disease, myocardial infarction, hypertension, and diabetes were risk factors for 5-year conversion from MCI to ACD.High body mass index, alcohol abuse, and sleep apnea were protective factors for 5-year conversion from MCI to ACD.Models using synthetic data, analogs of real patient data that retain the distribution, density, and covariance between variables of real patient data but are not attributable to any specific patient, performed just as well as models using real patient data. This could have significant implications in facilitating widely distributed computing of health-care data with minimized patient privacy concern that could accelerate scientific discoveries.
Collapse
Affiliation(s)
- Chase Irwin
- Phoenix Veterans Affairs Health Care SystemPhoenixArizonaUSA
- University of Arizona College of Medicine‐PhoenixPhoenixArizonaUSA
| | - Donna Tjandra
- Phoenix Veterans Affairs Health Care SystemPhoenixArizonaUSA
- University of MichiganAnn ArborMichiganUSA
| | - Chengcheng Hu
- University of Arizona College of Medicine‐PhoenixPhoenixArizonaUSA
- College of Public HealthUniversity of ArizonaTucsonArizonaUSA
| | - Vinod Aggarwal
- MDClone LimitedBe'er ShevaIsrael
- VHA Office of Healthcare Innovation and LearningVA Central OfficeWashington, District of ColumbiaUSA
| | - Amanda Lienau
- VHA Office of Healthcare Innovation and LearningVA Central OfficeWashington, District of ColumbiaUSA
| | | | | | - Raymond Q. Migrino
- Phoenix Veterans Affairs Health Care SystemPhoenixArizonaUSA
- University of Arizona College of Medicine‐PhoenixPhoenixArizonaUSA
| |
Collapse
|
33
|
Zeng B, Tang C, Wang J, Yang Q, Ren Q, Liu X. Pharmacologic and Nutritional Interventions for Early Alzheimer's Disease: A Systematic Review and Network Meta-Analysis of Randomized Controlled Trials. J Alzheimers Dis 2024; 99:1173-1186. [PMID: 38759015 PMCID: PMC11191524 DOI: 10.3233/jad-240161] [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] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Abstract
Background Early intervention is essential for meaningful disease modification in Alzheimer's disease (AD). Objective We aimed to determine the efficacy and safety of pharmacologic and nutritional interventions for early AD. Methods PubMed, Embase, the Cochrane Library, and ClinicalTrials.gov were searched from database inception until 1 September 2023. We included randomized controlled trials that evaluated the efficacy of interventions in early AD. Only interventions that demonstrated efficacy compared to placebo were included in the network meta-analysis (NMA). Then we performed frequentist fixed-effects NMA to rank the interventions. GRADE criteria were used to evaluate the level of evidence. Results Fifty-eight trials including a total of 33,864 participants and 48 interventions were eligible for inclusion. Among the 48 interventions analyzed, only 6 (12.5%) treatments- ranging from low to high certainty- showed significant improvement in cognitive decline compared to placebo. High certainty evidence indicated that donanemab (standardized mean difference [SMD] -0.239, 95% confidence interval [CI] -0.343 to -0.134) and lecanemab (SMD -0.194, 95% CI -0.279 to -0.108) moderately slowed the clinical progression in patients with amyloid pathology. Additionally, methylphenidate, donepezil, LipiDiDiet, and aducanumab with low certainty showed significant improvement in cognitive decline compared to placebo. However, there was no significant difference in serious adverse events as reported between the six interventions and placebo. Conclusions Only 12.5% of interventions studied demonstrated efficacy in reducing cognitive impairment in early AD. Donanemab and lecanemab have the potential to moderately slow the clinical progression in patients with amyloid pathology. Further evidence is required for early intervention in AD.
Collapse
Affiliation(s)
- Baoqi Zeng
- Central Laboratory, Tianjin Fifth Central Hospital (Peking University Binhai Hospital), Tianjin, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Tianjin Key Laboratory of Epigenetic for Organ Development of Preterm Infants, Tianjin Fifth Central Hospital, Tianjin, China
- Emergency Department, Tianjin Fifth Central Hospital, Tianjin, China
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People’s Hospital, Huangnan, Qinghai, China
| | - Chunbian Tang
- Central Laboratory, Tianjin Fifth Central Hospital (Peking University Binhai Hospital), Tianjin, China
- Tianjin Key Laboratory of Epigenetic for Organ Development of Preterm Infants, Tianjin Fifth Central Hospital, Tianjin, China
- Medical School of Tianjin University, Tianjin, China
| | - Junjian Wang
- Emergency Department, Tianjin Fifth Central Hospital, Tianjin, China
| | - Qingqing Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases, Peking University, Ministry of Education, Beijing, China
| | - Qingcuo Ren
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People’s Hospital, Huangnan, Qinghai, China
| | - Xiaozhi Liu
- Central Laboratory, Tianjin Fifth Central Hospital (Peking University Binhai Hospital), Tianjin, China
- Tianjin Key Laboratory of Epigenetic for Organ Development of Preterm Infants, Tianjin Fifth Central Hospital, Tianjin, China
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People’s Hospital, Huangnan, Qinghai, China
| |
Collapse
|
34
|
Ohno M. A Strategy for Allowing Earlier Diagnosis and Rigorous Evaluation of BACE1 Inhibitors in Preclinical Alzheimer's Disease. J Alzheimers Dis 2024; 99:431-445. [PMID: 38701146 DOI: 10.3233/jad-231451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Given continued failure of BACE1 inhibitor programs at symptomatic and prodromal stages of Alzheimer's disease (AD), clinical trials need to target the earlier preclinical stage. However, trial design is complex in this population with negative diagnosis of classical hippocampal amnesia on standard memory tests. Besides recent advances in brain imaging, electroencephalogram, and fluid-based biomarkers, new cognitive markers should be established for earlier diagnosis that can optimize recruitment to BACE1 inhibitor trials in presymptomatic AD. Notably, accelerated long-term forgetting (ALF) is emerging as a sensitive cognitive measure that can discriminate between asymptomatic individuals with high risks for developing AD and healthy controls. ALF is a form of declarative memory impairment characterized by increased forgetting rates over longer delays (days to months) despite normal storage within the standard delays of testing (20-60 min). Therefore, ALF may represent a harbinger of preclinical dementia and the impairment of systems memory consolidation, during which memory traces temporarily stored in the hippocampus become gradually integrated into cortical networks. This review provides an overview of the utility of ALF in a rational design of next-generation BACE1 inhibitor trials in preclinical AD. I explore potential mechanisms underlying ALF and relevant early-stage biomarkers useful for BACE1 inhibitor evaluation, including synaptic protein alterations, astrocytic dysregulation and neuron hyperactivity in the hippocampal-cortical network. Furthermore, given the physiological role of the isoform BACE2 as an AD-suppressor gene, I also discuss the possible association between the poor selectivity of BACE1 inhibitors and their side effects (e.g., cognitive worsening) in prior clinical trials.
Collapse
Affiliation(s)
- Masuo Ohno
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
| |
Collapse
|
35
|
Raman R, Hussen K, Donohue MC, Ernstrom K, Holdridge KC, Langford O, Molina-Henry DP, Pierce AL, Sims JR, Smith A, Yaari R, Aisen PS, Sperling R, Grill JD. Pre-Randomization Predictors of Study Discontinuation in a Preclinical Alzheimer's Disease Randomized Controlled Trial. J Prev Alzheimers Dis 2024; 11:874-880. [PMID: 39044496 PMCID: PMC11266258 DOI: 10.14283/jpad.2024.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/13/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Participant discontinuation from study treatment in a clinical trial can leave a trial underpowered, produce bias in statistical analysis, and limit interpretability of study results. Retaining participants in clinical trials for the full study duration is therefore as important as participant recruitment. OBJECTIVE This analysis aims to identify associations of pre-randomization characteristics of participants with premature discontinuation during the blinded phase of the Anti-Amyloid treatment in Asymptomatic AD (A4) Study. DESIGN All A4 trial randomized participants were classified as having prematurely discontinued study during the blinded period of the study for any reason (dropouts) or completed the blinded phase of the study on treatment (completers). SETTING The trial was conducted across 67 study sites in the United States, Canada, Japan and Australia through the global COVID-19 pandemic. PARTICIPANTS The sample consisted of all 1169 A4 trial randomized participants. MEASUREMENTS Pre-randomization demographic, clinical, amyloid PET and genetic predictors of study discontinuation were evaluated using a univariate generalized linear mixed model (GLMM), with discontinuation status as the binary outcome, each predictor as a fixed effect, and site as a random effect to account for differences among study sites in the trial. Characteristics significant at p<0.10 were then included in a multivariable GLMM. RESULTS Among randomized participants, 339 (29%) discontinued the study during the blinded period (median follow-up time in trial: 759 days). From the multivariable analysis, the two main predictors of study discontinuation were screening State-Trait Anxiety Inventory (STAI) scores (OR = 1.07 [95%CI = 1.02; 1.12]; p=0.002) and age (OR = 1.06 [95%CI = 1.03; 1.09]; p<0.001). Participants with a family history of dementia (OR = 0.75 [95%CI = 0.55; 1.01]; p=0.063) and APOE ε4 carriers (OR = 0.79 [95%CI = 0.6; 1.04]; p=0.094) were less likely to discontinue from the study, with the association being marginally significant. In these analyses, sex, race and ethnicity, cognitive scores and amyloid/tau PET scores were not associated with study dropout. CONCLUSIONS In the A4 trial, older participants and those with higher levels of anxiety at baseline as measured by the STAI were more likely to discontinue while those who had a family history of dementia or were APOE ε4 carriers were less likely to drop out. These findings have direct implications for future preclinical trial design and selection processes to identify those individuals at greatest risk of dropout and provide information to the study team to develop effective selection and retention strategies in AD prevention studies.
Collapse
Affiliation(s)
- R Raman
- Rema Raman, PhD, Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Elliott ML, Nielsen JA, Hanford LC, Hamadeh A, Hilbert T, Kober T, Dickerson BC, Hyman BT, Mair RW, Eldaief MC, Buckner RL. Precision Brain Morphometry Using Cluster Scanning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.23.23300492. [PMID: 38234845 PMCID: PMC10793507 DOI: 10.1101/2023.12.23.23300492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Measurement error limits the statistical power to detect group differences and longitudinal change in structural MRI morphometric measures (e.g., hippocampal volume, prefrontal thickness). Recent advances in scan acceleration enable extremely fast T1-weighted scans (~1 minute) to achieve morphometric errors that are close to the errors in longer traditional scans. As acceleration allows multiple scans to be acquired in rapid succession, it becomes possible to pool estimates to increase measurement precision, a strategy known as "cluster scanning." Here we explored brain morphometry using cluster scanning in a test-retest study of 40 individuals (12 younger adults, 18 cognitively unimpaired older adults, and 10 adults diagnosed with mild cognitive impairment or Alzheimer's Dementia). Morphometric errors from a single compressed sensing (CS) 1.0mm scan with 6x acceleration (CSx6) were, on average, 12% larger than a traditional scan using the Alzheimer's Disease Neuroimaging Initiative (ADNI) protocol. Pooled estimates from four clustered CSx6 acquisitions led to errors that were 34% smaller than ADNI despite having a shorter total acquisition time. Given a fixed amount of time, a gain in measurement precision can thus be achieved by acquiring multiple rapid scans instead of a single traditional scan. Errors were further reduced when estimates were pooled from eight CSx6 scans (51% smaller than ADNI). Neither pooling across a break nor pooling across multiple scan resolutions boosted this benefit. We discuss the potential of cluster scanning to improve morphometric precision, boost statistical power, and produce more sensitive disease progression biomarkers.
Collapse
Affiliation(s)
- Maxwell L Elliott
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Jared A Nielsen
- Department of Psychology, Neuroscience Center, Brigham Young University, Provo, UT, 84602, USA
| | - Lindsay C Hanford
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Aya Hamadeh
- Baylor College of Medicine, Houston, TX 77030
| | - Tom Hilbert
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tobias Kober
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bradford C Dickerson
- Frontotemporal Disorders Unit
- Alzheimer's Disease Research Center
- Athinoula A. Martinos Center for Biomedical Imaging
- Department of Neurology, Massachusetts General Hospital & Harvard Medical School
- Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Charlestown, MA 02129, USA
| | - Bradley T Hyman
- Alzheimer's Disease Research Center
- Department of Neurology, Massachusetts General Hospital & Harvard Medical School
| | - Ross W Mair
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Athinoula A. Martinos Center for Biomedical Imaging
| | - Mark C Eldaief
- Frontotemporal Disorders Unit
- Alzheimer's Disease Research Center
- Department of Neurology, Massachusetts General Hospital & Harvard Medical School
- Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Charlestown, MA 02129, USA
| | - Randy L Buckner
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Alzheimer's Disease Research Center
- Athinoula A. Martinos Center for Biomedical Imaging
- Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
37
|
Lee S, Kim E, Moon CE, Park C, Lim JW, Baek M, Shin MK, Ki J, Cho H, Ji YW, Haam S. Amplified fluorogenic immunoassay for early diagnosis and monitoring of Alzheimer's disease from tear fluid. Nat Commun 2023; 14:8153. [PMID: 38071202 PMCID: PMC10710446 DOI: 10.1038/s41467-023-43995-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Accurate diagnosis of Alzheimer's disease (AD) in its earliest stage can prevent the disease and delay the symptoms. Therefore, more sensitive, non-invasive, and simple screening tools are required for the early diagnosis and monitoring of AD. Here, we design a self-assembled nanoparticle-mediated amplified fluorogenic immunoassay (SNAFIA) consisting of magnetic and fluorophore-loaded polymeric nanoparticles. Using a discovery cohort of 21 subjects, proteomic analysis identifies adenylyl cyclase-associated protein 1 (CAP1) as a potential tear biomarker. The SNAFIA demonstrates a low detection limit (236 aM), good reliability (R2 = 0.991), and a wide analytical range (0.320-1000 fM) for CAP1 in tear fluid. Crucially, in the verification phase with 39 subjects, SNAFIA discriminates AD patients from healthy controls with 90% sensitivity and 100% specificity in under an hour. Utilizing tear fluid as a liquid biopsy, SNAFIA could potentially aid in long-term care planning, improve clinical trial efficiency, and accelerate therapeutic development for AD.
Collapse
Affiliation(s)
- Sojeong Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Eunjung Kim
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
- Department of Bioengineering & Nano-bioengineering, Research Center for Bio Materials and Process Development, Incheon National University, Incheon, 22012, Republic of Korea
| | - Chae-Eun Moon
- Department of Ophthalmology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, 16995, Republic of Korea
| | - Chaewon Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jong-Woo Lim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Minseok Baek
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, 26426, Republic of Korea
| | - Moo-Kwang Shin
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jisun Ki
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea.
| | - Yong Woo Ji
- Department of Ophthalmology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, 16995, Republic of Korea.
| | - Seungjoo Haam
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
38
|
Bolton CJ, Khan OA, Liu D, Wilhoite S, Dumitrescu L, Peterson A, Blennow K, Zetterberg H, Hohman TJ, Jefferson AL, Gifford KA. Sex and Education Modify the Association Between Subjective Cognitive Decline and Amyloid Pathology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.03.23297795. [PMID: 37961115 PMCID: PMC10635270 DOI: 10.1101/2023.11.03.23297795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Subjective cognitive decline (SCD) may be an early risk factor for dementia, particularly in highly educated individuals and women. This study examined the effect of education and sex on the association between SCD and Alzheimer's disease (AD) biomarkers in non-demented older adults. Method Vanderbilt Memory and Aging Project participants free of clinical dementia or stroke (n=156, 72±6 years, 37% mild cognitive impairment, 33% female) completed fasting lumbar puncture, SCD assessment, and Wide Range Achievement Test-III Reading subtest to assess reading level at baseline as a a proxy for educational quality. Cerebrospinal fluid (CSF) biomarkers for AD (β-amyloid 42 (Aβ42), Aβ42/40 ratio, phosphorylated tau (p-tau), tau, and neurofilament light (NfL)) were analyzed in batch. Linear mixed effects models related SCD to CSF AD biomarkers and follow-up models assessed SCD x sex, SCD x reading level , and SCD x education interactions on AD biomarkers. Result In main effect models, higher SCD was associated with lower Aβ42 and Aβ42/40 ratio (p-values<0.004). SCD was not associated with tau, p-tau, or NfL levels ( p- values>0.38). SCD score interacted with sex on Aβ42/40 ratio ( p =0.03) but no other biomarkers ( p -values>0.10). In stratified models, higher SCD was associated with lower Aβ42/40 ratio in men ( p =0.0003) but not in women ( p =0.48). SCD score interacted with education on Aβ42 ( p =0.005) and Aβ42/40 ratio ( p =0.001) such that higher education was associated with a stronger negative association between SCD and amyloid levels. No SCD score x reading level interaction was found (p-values> 0.51) though significant associations between SCD and amyloid markers were seen in the higher reading level group (p-values<0.004) but not the lower reading level group (p-values>0.12) when stratified by a median split in reading level. Conclusion Among community-dwelling older adults free of clinical dementia, higher SCD was associated with greater cerebral amyloid accumulation, one of the earliest pathological AD changes. SCD appears most useful in detecting early AD-related brain changes in men and individuals with higher quantity and quality of education. SCD was not associated with CSF markers of tau pathology or neurodegeneration. These findings suggest that considering sex and education is important when assessing SCD in older adults.
Collapse
|
39
|
Bader I, Bader I, Lopes Alves I, Vállez García D, Vellas B, Dubois B, Boada M, Marquié M, Altomare D, Scheltens P, Vandenberghe R, Hanseeuw B, Schöll M, Frisoni GB, Jessen F, Nordberg A, Kivipelto M, Ritchie CW, Grau-Rivera O, Molinuevo JL, Ford L, Stephens A, Gismondi R, Gispert JD, Farrar G, Barkhof F, Visser PJ, Collij LE. Recruitment of pre-dementia participants: main enrollment barriers in a longitudinal amyloid-PET study. Alzheimers Res Ther 2023; 15:189. [PMID: 37919783 PMCID: PMC10621165 DOI: 10.1186/s13195-023-01332-4] [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: 05/30/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND The mismatch between the limited availability versus the high demand of participants who are in the pre-dementia phase of Alzheimer's disease (AD) is a bottleneck for clinical studies in AD. Nevertheless, potential enrollment barriers in the pre-dementia population are relatively under-reported. In a large European longitudinal biomarker study (the AMYPAD-PNHS), we investigated main enrollment barriers in individuals with no or mild symptoms recruited from research and clinical parent cohorts (PCs) of ongoing observational studies. METHODS Logistic regression was used to predict study refusal based on sex, age, education, global cognition (MMSE), family history of dementia, and number of prior study visits. Study refusal rates and categorized enrollment barriers were compared between PCs using chi-squared tests. RESULTS 535/1856 (28.8%) of the participants recruited from ongoing studies declined participation in the AMYPAD-PNHS. Only for participants recruited from clinical PCs (n = 243), a higher MMSE-score (β = - 0.22, OR = 0.80, p < .05), more prior study visits (β = - 0.93, OR = 0.40, p < .001), and positive family history of dementia (β = 2.08, OR = 8.02, p < .01) resulted in lower odds on study refusal. General study burden was the main enrollment barrier (36.1%), followed by amyloid-PET related burden (PCresearch = 27.4%, PCclinical = 9.0%, X2 = 10.56, p = .001), and loss of research interest (PCclinical = 46.3%, PCresearch = 16.5%, X2 = 32.34, p < .001). CONCLUSIONS The enrollment rate for the AMYPAD-PNHS was relatively high, suggesting an advantage of recruitment via ongoing studies. In this observational cohort, study burden reduction and tailored strategies may potentially improve participant enrollment into trial readiness cohorts such as for phase-3 early anti-amyloid intervention trials. The AMYPAD-PNHS (EudraCT: 2018-002277-22) was approved by the ethical review board of the VU Medical Center (VUmc) as the Sponsor site and in every affiliated site.
Collapse
Affiliation(s)
- Ilse Bader
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV, Amsterdam, The Netherlands.
| | - Ilona Bader
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, 1081 HV, The Netherlands
| | - Isadora Lopes Alves
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, 1081 HV, The Netherlands
- Brain Research Center, 1081 GN, Amsterdam, The Netherlands
| | - David Vállez García
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, 1081 HV, The Netherlands
| | - Bruno Vellas
- Gérontopole of Toulouse, University Hospital of Toulouse (CHU-Toulouse), 31300, Toulouse, France
- UMR INSERM 1027, University of Toulouse III, 31062, Toulouse, France
| | - Bruno Dubois
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain Institute, Salpetriere Hospital, Sorbonne University, 75013, Paris, France
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08028, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08028, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Daniele Altomare
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25123, Brescia, Italy
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV, Amsterdam, The Netherlands
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Leuven Brain Institute, KU Leuven, 3001, Louvain, Belgium
| | - Bernard Hanseeuw
- Institute of Neuroscience, Université Catholique de Louvain, 1200, Brussels, Belgium
- Department of Neurology, Clinique Universitaires Saint-Luc, 1200, Brussels, Belgium
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, 02155, USA
- WELBIO Department, WEL Research Institute, Avenue Pasteur, 6, 1300, Wavre, Belgium
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 405 30, Gothenburg, Sweden
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30, Gothenburg, Sweden
- Dementia Research Centre, Queen Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, 1205, Geneva, Switzerland
- Geneva Memory Center, Geneva University Hospitals, 1205, Geneva, Switzerland
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society (NVS), Karolinska Institutet, 171 77, Stockholm, Sweden
- Theme Inflammation, Karolinska University Hospital, Stockholm, 171 77, Sweden
- Theme Aging, Karolinska University Hospital, Stockholm, 171 77, Sweden
| | - Miia Kivipelto
- Kuopio University Hospital, 70210, Kuopio, Finland
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society (NVS), Karolinska Institutet, 171 77, Stockholm, Sweden
- Imperial College London, London, SW7 2AZ, UK
| | | | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005, Barcelona, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005, Barcelona, Spain
- H. Lundbeck A/S, 2500, Copenhagen, Denmark
| | - Lisa Ford
- Janssen Research and Development, Titusville, NJ, 08560, USA
| | | | | | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005, Barcelona, Spain
| | - Gill Farrar
- GE Healthcare, Pharmaceutical Diagnostics, Amersham, HP7 9LL, UK
| | - Frederik Barkhof
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, 1081 HV, The Netherlands
- Institutes of Neurology and Healthcare Engineering, UCL, London, WC1N 3BG, UK
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV, Amsterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Lyduine E Collij
- Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, 1081 HZ, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, 1081 HV, The Netherlands
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, 221 00, Malmö, Sweden
| |
Collapse
|
40
|
Bu Y, Wang K, Yang X, Nie G. Sensitive dual-mode sensing platform for Amyloid β detection: Combining dual Z-scheme heterojunction enhanced photoelectrochemistry analysis and dual-wavelength ratiometric electrochemiluminescence strategy. Biosens Bioelectron 2023; 237:115507. [PMID: 37437453 DOI: 10.1016/j.bios.2023.115507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/19/2023] [Accepted: 06/30/2023] [Indexed: 07/14/2023]
Abstract
As a tumor biomarker, the accumulation of amyloid β oligomers (Aβo) in the brain has been suggested as a key feature in the pathogenesis and progression of Alzheimer's disease (AD). In this work, we designed a novel photoelectrochemical (PEC) and electrochemiluminescence resonance energy transfer (ECL-RET) dual-mode biosensor to achieve ultra-sensitive detection of Aβo. Specifically, the electrode surface modified Carbon Dots (C Dots) and the electrodeposited polyaniline (PANI) film formed a Z-scheme heterojunction reversing the photocurrent signal, and then the Aβo specific recognition peptide was attached to the surface via amide bonding between the amino group of PANI and carbonyl group of peptide. After that, in the presence of CdTe labeled specific recognition aptamer for Aβ (CdTe-Apt), Aβo was captured to construct a sandwich-type biosensor and exhibited a significantly enhanced cathodic photocurrent response because the formed dual Z-scheme heterojunction promoted charge separation efficiency. Interestingly, the proposed biosensor also caused a ratiometric change in the ECL intensity at 555 nm and 640 nm. Therefore, the developed biosensor achieved dual-mode detection of Aβo, where the PEC detection range of Aβo was from 10 fM to 0.1 μM (with a detection limit of 4.27 fM) and the ECL method provided a linear detection range of 10 fM to 10 nM (with a detection limit of 6.41 fM). The stability and reliability of the experimental results indicate that this has been a promising biosensing pattern and could be extended to the analysis of other biomarkers.
Collapse
Affiliation(s)
- Yuwei Bu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China
| | - Kun Wang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China
| | - Xiaoyan Yang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| | - Guangming Nie
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE; College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| |
Collapse
|
41
|
Noori M, Mahboobi R, Nabavi-Rad A, Jamshidizadeh S, Fakharian F, Yadegar A, Zali MR. Helicobacter pylori infection contributes to the expression of Alzheimer's disease-associated risk factors and neuroinflammation. Heliyon 2023; 9:e19607. [PMID: 37810022 PMCID: PMC10558876 DOI: 10.1016/j.heliyon.2023.e19607] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 10/10/2023] Open
Abstract
Over time, mounting evidence has demonstrated extra-gastric manifestations of Helicobacter pylori infection. As such, a number of studies demonstrated the potential contribution of H. pylori infection to the incidence and progression of Alzheimer's disease (AD). Considering unanswered questions regarding the effect of H. pylori infection on brain activity, we sought to investigate the impact of H. pylori infection on the expression of AD-associated risk factors. We used two H. pylori clinical strains obtained from two patients with peptic ulcer and evaluated their influence on the expression level of AD-associated genes (APP, ApoE2, ApoE4, ABCA7, BIN1, Clu, CD33) and genes for inflammatory markers (TLR-4, IL-8, TNF-α) by RT-qPCR in human glioblastoma (U87MG) and astrocyte (1321N1) cell lines. The expression of inflammatory cytokines was further assessed by ELISA assay. The exposure of U97MG and 1321N1 cells to H. pylori strains resulted in a significant enhancement in the expression level of the risk allele ApoE4, while reducing the expression of the protective allele ApoE2. H. pylori infection remarkably increased the expression level of main AD-associated risk genes, and also pro-inflammatory cytokines. Furthermore, we noticed a substantial elevation in the mRNA expression level of transmembrane receptor TLR-4 following H. pylori infection. Our findings presented the potential for H. pylori to stimulate the expression of AD-associated risk genes and trigger neuroinflammation in the brain tissue. This, in principle, leads to the recommendation that AD patients should perhaps test for H. pylori infection and receive treatments upon positive detection.
Collapse
Affiliation(s)
- Maryam Noori
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramina Mahboobi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Jamshidizadeh
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzaneh Fakharian
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
van Heusden FC, van Nifterick AM, Souza BC, França ASC, Nauta IM, Stam CJ, Scheltens P, Smit AB, Gouw AA, van Kesteren RE. Neurophysiological alterations in mice and humans carrying mutations in APP and PSEN1 genes. Alzheimers Res Ther 2023; 15:142. [PMID: 37608393 PMCID: PMC10464047 DOI: 10.1186/s13195-023-01287-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/11/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Studies in animal models of Alzheimer's disease (AD) have provided valuable insights into the molecular and cellular processes underlying neuronal network dysfunction. Whether and how AD-related neurophysiological alterations translate between mice and humans remains however uncertain. METHODS We characterized neurophysiological alterations in mice and humans carrying AD mutations in the APP and/or PSEN1 genes, focusing on early pre-symptomatic changes. Longitudinal local field potential recordings were performed in APP/PS1 mice and cross-sectional magnetoencephalography recordings in human APP and/or PSEN1 mutation carriers. All recordings were acquired in the left frontal cortex, parietal cortex, and hippocampus. Spectral power and functional connectivity were analyzed and compared with wildtype control mice and healthy age-matched human subjects. RESULTS APP/PS1 mice showed increased absolute power, especially at higher frequencies (beta and gamma) and predominantly between 3 and 6 moa. Relative power showed an overall shift from lower to higher frequencies over almost the entire recording period and across all three brain regions. Human mutation carriers, on the other hand, did not show changes in power except for an increase in relative theta power in the hippocampus. Mouse parietal cortex and hippocampal power spectra showed a characteristic peak at around 8 Hz which was not significantly altered in transgenic mice. Human power spectra showed a characteristic peak at around 9 Hz, the frequency of which was significantly reduced in mutation carriers. Significant alterations in functional connectivity were detected in theta, alpha, beta, and gamma frequency bands, but the exact frequency range and direction of change differed for APP/PS1 mice and human mutation carriers. CONCLUSIONS Both mice and humans carrying APP and/or PSEN1 mutations show abnormal neurophysiological activity, but several measures do not translate one-to-one between species. Alterations in absolute and relative power in mice should be interpreted with care and may be due to overexpression of amyloid in combination with the absence of tau pathology and cholinergic degeneration. Future studies should explore whether changes in brain activity in other AD mouse models, for instance, those also including tau pathology, provide better translation to the human AD continuum.
Collapse
Affiliation(s)
- Fran C van Heusden
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, The Netherlands
| | - Anne M van Nifterick
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - Bryan C Souza
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, 6525AJ, The Netherlands
| | - Arthur S C França
- Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, 6525AJ, The Netherlands
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, 1105 BA, The Netherlands
| | - Ilse M Nauta
- MS Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - Cornelis J Stam
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, The Netherlands
| | - Alida A Gouw
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, 1081HV, The Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, The Netherlands.
| |
Collapse
|
43
|
Elefante C, Brancati GE, Ismail Z, Ricciardulli S, Beatino MF, Lepri V, Famà A, Ferrari E, Giampietri L, Baldacci F, Ceravolo R, Maremmani I, Lattanzi L, Perugi G. Mild Behavioral Impairment in Psychogeriatric Patients: Clinical Features and Psychopathology Severity. J Clin Med 2023; 12:5423. [PMID: 37629464 PMCID: PMC10455739 DOI: 10.3390/jcm12165423] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
The Mild Behavioral Impairment (MBI) concept was developed to determine whether late-onset persistent neuropsychiatric symptoms (NPSs) may be early manifestations of cognitive decline. Our study aims to investigate the prevalence and differentiating features of MBI with respect to major neurocognitive disorders (MNDs) and primary psychiatric disorders (PPDs). A total of 144 elderly patients who were referred to our psychogeriatric outpatient service were recruited. The severity of mental illness was evaluated by means of the Clinical Global Impression Severity scale, the severity of psychopathology was evaluated by means of the Brief Psychiatric Rating Scale (BPRS), and overall functioning was evaluated by means of the Global Assessment of Functioning scale. The sample included 73 (50.6%) patients with PPDs, 40 (27.8%) patients with MBI, and 31 (21.5%) patients with MNDs. Patients with MNDs reported the greatest severity of mental illness, the highest BPRS Total, Psychosis, Activation, and Negative Symptom scores, and the lowest functioning. Patients with MBI and PPDs had comparable levels of severity of mental illness and overall functioning, but MBI patients reported higher BPRS Total and Negative Symptom scores than PPD patients. Patients with MBI frequently reported specific clinical features, including a higher severity of apathy and motor retardation. These features merit further investigation since they may help the differential diagnosis between MBI and PPDs.
Collapse
Affiliation(s)
- Camilla Elefante
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.E.); (G.E.B.); (S.R.); (M.F.B.); (V.L.); (G.P.)
| | - Giulio Emilio Brancati
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.E.); (G.E.B.); (S.R.); (M.F.B.); (V.L.); (G.P.)
| | - Zahinoor Ismail
- Departments of Psychiatry, Clinical Neurosciences, Community Health Sciences, and Pathology and Laboratory Medicine, Hotchkiss Brain Institute & O’Brien Institute for Public Health, University of Calgary, Calgary, AB T2N 1N4, Canada;
- College of Health and Medicine, University of Exeter, Exeter EX4 4QG, UK
| | - Sara Ricciardulli
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.E.); (G.E.B.); (S.R.); (M.F.B.); (V.L.); (G.P.)
| | - Maria Francesca Beatino
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.E.); (G.E.B.); (S.R.); (M.F.B.); (V.L.); (G.P.)
| | - Vittoria Lepri
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.E.); (G.E.B.); (S.R.); (M.F.B.); (V.L.); (G.P.)
| | - Antonella Famà
- Psychiatry Unit, Azienda Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy; (A.F.); (E.F.); (L.L.)
| | - Elisabetta Ferrari
- Psychiatry Unit, Azienda Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy; (A.F.); (E.F.); (L.L.)
| | - Linda Giampietri
- Neurology Unit, Santa Chiara University Hospital, 56126 Pisa, Italy; (L.G.); (F.B.); (R.C.)
| | - Filippo Baldacci
- Neurology Unit, Santa Chiara University Hospital, 56126 Pisa, Italy; (L.G.); (F.B.); (R.C.)
| | - Roberto Ceravolo
- Neurology Unit, Santa Chiara University Hospital, 56126 Pisa, Italy; (L.G.); (F.B.); (R.C.)
| | - Icro Maremmani
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.E.); (G.E.B.); (S.R.); (M.F.B.); (V.L.); (G.P.)
- G. De Lisio Institute of Behavioral Sciences, 56127 Pisa, Italy
- Addiction Medicine, Saint Camillus International University of Health and Medical Sciences (UniCamillus), 00131 Rome, Italy
| | - Lorenzo Lattanzi
- Psychiatry Unit, Azienda Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy; (A.F.); (E.F.); (L.L.)
| | - Giulio Perugi
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (C.E.); (G.E.B.); (S.R.); (M.F.B.); (V.L.); (G.P.)
- G. De Lisio Institute of Behavioral Sciences, 56127 Pisa, Italy
| |
Collapse
|
44
|
Sims JR, Zimmer JA, Evans CD, Lu M, Ardayfio P, Sparks J, Wessels AM, Shcherbinin S, Wang H, Monkul Nery ES, Collins EC, Solomon P, Salloway S, Apostolova LG, Hansson O, Ritchie C, Brooks DA, Mintun M, Skovronsky DM. Donanemab in Early Symptomatic Alzheimer Disease: The TRAILBLAZER-ALZ 2 Randomized Clinical Trial. JAMA 2023; 330:512-527. [PMID: 37459141 PMCID: PMC10352931 DOI: 10.1001/jama.2023.13239] [Citation(s) in RCA: 1056] [Impact Index Per Article: 528.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/28/2023] [Indexed: 07/20/2023]
Abstract
Importance There are limited efficacious treatments for Alzheimer disease. Objective To assess efficacy and adverse events of donanemab, an antibody designed to clear brain amyloid plaque. Design, Setting, and Participants Multicenter (277 medical research centers/hospitals in 8 countries), randomized, double-blind, placebo-controlled, 18-month phase 3 trial that enrolled 1736 participants with early symptomatic Alzheimer disease (mild cognitive impairment/mild dementia) with amyloid and low/medium or high tau pathology based on positron emission tomography imaging from June 2020 to November 2021 (last patient visit for primary outcome in April 2023). Interventions Participants were randomized in a 1:1 ratio to receive donanemab (n = 860) or placebo (n = 876) intravenously every 4 weeks for 72 weeks. Participants in the donanemab group were switched to receive placebo in a blinded manner if dose completion criteria were met. Main Outcomes and Measures The primary outcome was change in integrated Alzheimer Disease Rating Scale (iADRS) score from baseline to 76 weeks (range, 0-144; lower scores indicate greater impairment). There were 24 gated outcomes (primary, secondary, and exploratory), including the secondary outcome of change in the sum of boxes of the Clinical Dementia Rating Scale (CDR-SB) score (range, 0-18; higher scores indicate greater impairment). Statistical testing allocated α of .04 to testing low/medium tau population outcomes, with the remainder (.01) for combined population outcomes. Results Among 1736 randomized participants (mean age, 73.0 years; 996 [57.4%] women; 1182 [68.1%] with low/medium tau pathology and 552 [31.8%] with high tau pathology), 1320 (76%) completed the trial. Of the 24 gated outcomes, 23 were statistically significant. The least-squares mean (LSM) change in iADRS score at 76 weeks was -6.02 (95% CI, -7.01 to -5.03) in the donanemab group and -9.27 (95% CI, -10.23 to -8.31) in the placebo group (difference, 3.25 [95% CI, 1.88-4.62]; P < .001) in the low/medium tau population and -10.2 (95% CI, -11.22 to -9.16) with donanemab and -13.1 (95% CI, -14.10 to -12.13) with placebo (difference, 2.92 [95% CI, 1.51-4.33]; P < .001) in the combined population. LSM change in CDR-SB score at 76 weeks was 1.20 (95% CI, 1.00-1.41) with donanemab and 1.88 (95% CI, 1.68-2.08) with placebo (difference, -0.67 [95% CI, -0.95 to -0.40]; P < .001) in the low/medium tau population and 1.72 (95% CI, 1.53-1.91) with donanemab and 2.42 (95% CI, 2.24-2.60) with placebo (difference, -0.7 [95% CI, -0.95 to -0.45]; P < .001) in the combined population. Amyloid-related imaging abnormalities of edema or effusion occurred in 205 participants (24.0%; 52 symptomatic) in the donanemab group and 18 (2.1%; 0 symptomatic during study) in the placebo group and infusion-related reactions occurred in 74 participants (8.7%) with donanemab and 4 (0.5%) with placebo. Three deaths in the donanemab group and 1 in the placebo group were considered treatment related. Conclusions and Relevance Among participants with early symptomatic Alzheimer disease and amyloid and tau pathology, donanemab significantly slowed clinical progression at 76 weeks in those with low/medium tau and in the combined low/medium and high tau pathology population. Trial Registration ClinicalTrials.gov Identifier: NCT04437511.
Collapse
Affiliation(s)
| | | | | | - Ming Lu
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | | | | | - Hong Wang
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | - Paul Solomon
- Boston Center for Memory and Boston University Alzheimer’s Disease Center, Boston, Massachusetts
| | - Stephen Salloway
- Department of Neurology and Department of Psychiatry, Alpert Medical School of Brown University, Providence, Rhode Island
- Butler Hospital, Providence, Rhode Island
| | - Liana G. Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Lund, Sweden
| | | | | | - Mark Mintun
- Eli Lilly and Company, Indianapolis, Indiana
| | | |
Collapse
|
45
|
Julkunen V, Schwarz C, Kalapudas J, Hallikainen M, Piironen AK, Mannermaa A, Kujala H, Laitinen T, Kosma VM, Paajanen TI, Kälviäinen R, Hiltunen M, Herukka SK, Kärkkäinen S, Kokkola T, Urjansson M, Perola M, Palotie A, Vuoksimaa E, Runz H. A FinnGen pilot clinical recall study for Alzheimer's disease. Sci Rep 2023; 13:12641. [PMID: 37537264 PMCID: PMC10400697 DOI: 10.1038/s41598-023-39835-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023] Open
Abstract
Successful development of novel therapies requires that clinical trials are conducted in patient cohorts with the highest benefit-to-risk ratio. Population-based biobanks with comprehensive health and genetic data from large numbers of individuals hold promise to facilitate identification of trial participants, particularly when interventions need to start while symptoms are still mild, such as for Alzheimer's disease (AD). This study describes a process for clinical recall studies from FinnGen. We demonstrate the feasibility to systematically ascertain customized clinical data from FinnGen participants with ICD10 diagnosis of AD or mild cognitive disorder (MCD) in a single-center cross-sectional study testing blood-based biomarkers and cognitive functioning in-person, computer-based and remote. As a result, 19% (27/140) of a pre-specified FinnGen subcohort were successfully recalled and completed the study. Hospital records largely validated registry entries. For 8/12 MCD patients, other reasons than AD were identified as underlying diagnosis. Cognitive measures correlated across platforms, with highest consistencies for dementia screening (r = 0.818) and semantic fluency (r = 0.764), respectively, for in-person versus telephone-administered tests. Glial fibrillary acidic protein (GFAP) (p < 0.002) and phosphorylated-tau 181 (pTau-181) (p < 0.020) most reliably differentiated AD from MCD participants. We conclude that informative, customized clinical recall studies from FinnGen are feasible.
Collapse
Affiliation(s)
- Valtteri Julkunen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland.
- Department of Neurology, Neurocenter, Kuopio University Hospital, Kuopio, Finland.
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Claudia Schwarz
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Juho Kalapudas
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Merja Hallikainen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | | - Teemu I Paajanen
- Work Ability and Working Careers, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Reetta Kälviäinen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Sari Kärkkäinen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Tarja Kokkola
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Mia Urjansson
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Markus Perola
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eero Vuoksimaa
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Heiko Runz
- Translational Sciences, Biogen, Cambridge, MA, USA.
| |
Collapse
|
46
|
Elliott ML, Hanford LC, Hamadeh A, Hilbert T, Kober T, Dickerson BC, Mair RW, Eldaief MC, Buckner RL. Brain morphometry in older adults with and without dementia using extremely rapid structural scans. Neuroimage 2023; 276:120173. [PMID: 37201641 PMCID: PMC10330834 DOI: 10.1016/j.neuroimage.2023.120173] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/25/2023] [Accepted: 05/15/2023] [Indexed: 05/20/2023] Open
Abstract
T1-weighted structural MRI is widely used to measure brain morphometry (e.g., cortical thickness and subcortical volumes). Accelerated scans as fast as one minute or less are now available but it is unclear if they are adequate for quantitative morphometry. Here we compared the measurement properties of a widely adopted 1.0 mm resolution scan from the Alzheimer's Disease Neuroimaging Initiative (ADNI = 5'12'') with two variants of highly accelerated 1.0 mm scans (compressed-sensing, CSx6 = 1'12''; and wave-controlled aliasing in parallel imaging, WAVEx9 = 1'09'') in a test-retest study of 37 older adults aged 54 to 86 (including 19 individuals diagnosed with a neurodegenerative dementia). Rapid scans produced highly reliable morphometric measures that largely matched the quality of morphometrics derived from the ADNI scan. Regions of lower reliability and relative divergence between ADNI and rapid scan alternatives tended to occur in midline regions and regions with susceptibility-induced artifacts. Critically, the rapid scans yielded morphometric measures similar to the ADNI scan in regions of high atrophy. The results converge to suggest that, for many current uses, extremely rapid scans can replace longer scans. As a final test, we explored the possibility of a 0'49'' 1.2 mm CSx6 structural scan, which also showed promise. Rapid structural scans may benefit MRI studies by shortening the scan session and reducing cost, minimizing opportunity for movement, creating room for additional scan sequences, and allowing for the repetition of structural scans to increase precision of the estimates.
Collapse
Affiliation(s)
- Maxwell L Elliott
- Department of Psychology, Center for Brain Science, Harvard University, 52 Oxford Street, Northwest Laboratory 280.10, Cambridge, MA 02138, USA.
| | - Lindsay C Hanford
- Department of Psychology, Center for Brain Science, Harvard University, 52 Oxford Street, Northwest Laboratory 280.10, Cambridge, MA 02138, USA
| | - Aya Hamadeh
- Baylor College of Medicine, Houston, TX 77030, USA
| | - Tom Hilbert
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Switzerland; Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tobias Kober
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Switzerland; Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bradford C Dickerson
- Frontotemporal Disorders Unit, Massachusetts General Hospital, USA; Alzheimer's Disease Research Center, Massachusetts General Hospital, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, USA; Department of Neurology, Massachusetts General Hospital & Harvard Medical School, USA; Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Charlestown, MA 02129, USA
| | - Ross W Mair
- Department of Psychology, Center for Brain Science, Harvard University, 52 Oxford Street, Northwest Laboratory 280.10, Cambridge, MA 02138, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, USA
| | - Mark C Eldaief
- Frontotemporal Disorders Unit, Massachusetts General Hospital, USA; Alzheimer's Disease Research Center, Massachusetts General Hospital, USA; Department of Neurology, Massachusetts General Hospital & Harvard Medical School, USA; Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Charlestown, MA 02129, USA
| | - Randy L Buckner
- Department of Psychology, Center for Brain Science, Harvard University, 52 Oxford Street, Northwest Laboratory 280.10, Cambridge, MA 02138, USA; Alzheimer's Disease Research Center, Massachusetts General Hospital, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, USA; Department of Psychiatry, Massachusetts General Hospital & Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
47
|
Lutz J, Pratap A, Lenze EJ, Bestha D, Lipschitz JM, Karantzoulis S, Vaidyanathan U, Robin J, Horan W, Brannan S, Mittoux A, Davis MC, Lakhan SE, Keefe R. Innovative Technologies in CNS Trials: Promises and Pitfalls for Recruitment, Retention, and Representativeness. INNOVATIONS IN CLINICAL NEUROSCIENCE 2023; 20:40-46. [PMID: 37817816 PMCID: PMC10561984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
Objective Recruitment of a sufficiently large and representative patient sample and its retention during central nervous system (CNS) trials presents major challenges for study sponsors. Technological advances are reshaping clinical trial operations to meet these challenges, and the COVID-19 pandemic further accelerated this development. Method of Research The International Society for CNS Clinical Trials and Methodology (ISCTM; www.isctm.org) Innovative Technologies for CNS Trials Working Group surveyed the state of technological innovations for improved recruitment and retention and assessed their promises and pitfalls. Results Online advertisement and electronic patient registries can enhance recruitment, but challenges with sample representativeness, conversion rates from eligible prescreening to enrolled patients, data privacy and security, and patient identification remain hurdles for optimal use of these technologies. Electronic medical records (EMR) mining with artificial intelligence (AI)/machine learning (ML) methods is promising but awaits translation into trials. During the study treatment phase, technological innovations increasingly support participant retention, including adherence with the investigational treatment. Digital tools for adherence and retention support take many forms, including patient-centric communication channels between researchers and participants, real-time study reminders, and digital behavioral interventions to increase study compliance. However, such tools add technical complexities to trials, and their impact on the generalizability of results are largely unknown. Conclusion Overall, the group found a scarcity of systematic data directly assessing the impact of technological innovations on study recruitment and retention in CNS trials, even for strategies with already high adoption, such as online recruitment. Given the added complexity and costs associated with most technological innovations, such data is needed to fully harness technologies for CNS trials and drive further adoption.
Collapse
Affiliation(s)
- Jacqueline Lutz
- Dr. Lutz was with Medical Office, Click Therapeutics, Inc. in New York, New York, at the time of writing; she is now with Biogen Digital Health in Cambridge, Massachusetts, and Boston University School of Medicine in Boston, Massachusetts
| | - Abhishek Pratap
- Dr. Pratap was with Center for Addiction & Mental Health in Toronto, Canada, at the time of writing; he is now with Boehringer Ingelheim in Ridgefield, Connecticut; King's College London in London, United Kingdom; and Department of Biomedical Informatics and Medical Education, University of Washington in Seattle, Washington
| | - Eric J Lenze
- Dr. Lenze is with Department of Psychiatry, Washington University School of Medicine in St. Louis, Missouri
| | - Durga Bestha
- Dr. Bestha is with Atrium Health in Charlotte, North Carolina
| | - Jessica M Lipschitz
- Dr. Lipschitz is with Brigham and Women's Hospital in Boston, Massachusetts, and Harvard Medical School in Boston, Massachusetts
| | | | - Uma Vaidyanathan
- Dr. Vaidyanathan was with Boehringer Ingelheim in Ridgefield, Connecticut, at the time of writing; she is now with Sublimus in Ridgefield, Connecticut
| | - Jessica Robin
- Dr. Robin is with Winterlight Labs, Inc. in Toronto, Canada
| | - William Horan
- Dr. Horan was with WCG VeraSci in Durham, North Carolina, at the time of writing; he is now with Karuna Therapeutics in Boston, Massachusetts, and University of California in Los Angeles, California
| | - Stephen Brannan
- Dr. Brannan is with Karuna Therapeutics in Boston, Massachusetts
| | | | | | - Shaheen E Lakhan
- Dr. Lakhan is with Medical Office, Click Therapeutics, Inc. in New York, New York, and School of Neuroscience, Virginia Tech in Blacksburg, Virginia
| | - Richard Keefe
- Dr. Keefe is with Department of Psychiatry, Duke University Medical Center in Durham, North Carolina
| |
Collapse
|
48
|
Brady ES, Griffiths J, Andrianova L, Bielska M, Saito T, Saido TC, Randall AD, Tamagnini F, Witton J, Craig MT. Alterations to parvalbumin-expressing interneuron function and associated network oscillations in the hippocampal - medial prefrontal cortex circuit during natural sleep in App NL-G-F/NL-G-F mice. Neurobiol Dis 2023; 182:106151. [PMID: 37172910 DOI: 10.1016/j.nbd.2023.106151] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023] Open
Abstract
In the early stages of Alzheimer's disease (AD), the accumulation of the peptide amyloid-β (Aβ) damages synapses and disrupts neuronal activity, leading to the disruption of neuronal oscillations associated with cognition. This is thought to be largely due to impairments in CNS synaptic inhibition, particularly via parvalbumin (PV)-expressing interneurons that are essential for generating several key oscillations. Research in this field has largely been conducted in mouse models that over-express humanised, mutated forms of AD-associated genes that produce exaggerated pathology. This has prompted the development and use of knock-in mouse lines that express these genes at an endogenous level, such as the AppNL-G-F/NL-G-F mouse model used in the present study. These mice appear to model the early stages of Aβ-induced network impairments, yet an in-depth characterisation of these impairments in currently lacking. Therefore, using 16 month-old AppNL-G-F/NL-G-F mice, we analysed neuronal oscillations found in the hippocampus and medial prefrontal cortex (mPFC) during awake behaviour, rapid eye movement (REM) and non-REM (NREM) sleep to assess the extent of network dysfunction. No alterations to gamma oscillations were found to occur in the hippocampus or mPFC during either awake behaviour, REM or NREM sleep. However, during NREM sleep an increase in the power of mPFC spindles and decrease in the power of hippocampal sharp-wave ripples was identified. The latter was accompanied by an increase in the synchronisation of PV-expressing interneuron activity, as measured using two-photon Ca2+ imaging, as well as a decrease in PV-expressing interneuron density. Furthermore, although changes were detected in local network function of mPFC and hippocampus, long-range communication between these regions appeared intact. Altogether, our results suggest that these NREM sleep-specific impairments represent the early stages of circuit breakdown in response to amyloidopathy.
Collapse
Affiliation(s)
- Erica S Brady
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; Gladstone Institute for Neurological Disease, 1650 Owens Street, San Francisco, CA 91458, United States of America
| | - Jessica Griffiths
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK
| | - Lilya Andrianova
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Monika Bielska
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Andrew D Randall
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; School of Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - Francesco Tamagnini
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK
| | - Jonathan Witton
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK.
| | - Michael T Craig
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Prince of Wales Road, Exeter EX4 4PS, England, UK; School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, UK.
| |
Collapse
|
49
|
Li W, Peng X, Mei X, Dong M, Li Y, Dong H. Multifunctional DNA Tetrahedron for Alzheimer's Disease Mitochondria-Targeted Therapy by MicroRNA Regulation. ACS APPLIED MATERIALS & INTERFACES 2023; 15:22977-22984. [PMID: 37145038 DOI: 10.1021/acsami.3c03181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The principal hallmark of Alzheimer's disease (AD) is neuron mitochondrial dysfunction, whereas mitochondrial miRNAs potentially play important roles. Nevertheless, efficacious mitochondria organelle therapeutic agents for treatment and management of AD are highly advisable. Herein, we report a multifunctional DNA tetrahedron-based mitochondria-targeted therapeutic platform, termed tetrahedral DNA framework-based nanoparticles (TDFNs), which was modified with triphenylphosphine (TPP) for mitochondria-targeting, cholesterol (Chol) for crossing the central nervous system, and functional antisense oligonucleotide (ASO) for both AD diagnosis and gene silencing therapy. After injecting intravenously through the tail vein of 3 × Tg-AD model mice, TDFNs can both easily cross the blood brain barrier and accurately arrive at the mitochondria. The functional ASO could not only be detected via the fluorescence signal for diagnosis but also mediate the apoptosis pathway through knocking miRNA-34a down, leading to recovery of the neuron cells. The superior performance of TDFNs suggests the great potential in mitochondria organelle therapeutics.
Collapse
Affiliation(s)
- Weiqun Li
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Xin Peng
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Xuecui Mei
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
| | - Mingjie Dong
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Center, Precision Medicine and Health Research Institute, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Yingchun Li
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen, Guangdong 518055, China
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Haifeng Dong
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Center, Precision Medicine and Health Research Institute, Shenzhen University, Shenzhen, Guangdong 518060, China
| |
Collapse
|
50
|
Rafii MS, Aisen PS. Detection and treatment of Alzheimer's disease in its preclinical stage. NATURE AGING 2023; 3:520-531. [PMID: 37202518 PMCID: PMC11110912 DOI: 10.1038/s43587-023-00410-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/29/2023] [Indexed: 05/20/2023]
Abstract
Longitudinal multimodal biomarker studies reveal that the continuum of Alzheimer's disease (AD) includes a long latent phase, referred to as preclinical AD, which precedes the onset of symptoms by decades. Treatment during the preclinical AD phase offers an optimal opportunity for slowing the progression of disease. However, trial design in this population is complex. In this Review, we discuss the recent advances in accurate plasma measurements, new recruitment approaches, sensitive cognitive instruments and self-reported outcomes that have facilitated the successful launch of multiple phase 3 trials for preclinical AD. The recent success of anti-amyloid immunotherapy trials in symptomatic AD has increased the enthusiasm for testing this strategy at the earliest feasible stage. We provide an outlook for standard screening of amyloid accumulation at the preclinical stage in clinically normal individuals, during which effective therapy to delay or prevent cognitive decline can be initiated.
Collapse
Affiliation(s)
- Michael S Rafii
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine University of Southern California, Los Angeles, CA, USA.
| | - Paul S Aisen
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine University of Southern California, Los Angeles, CA, USA
| |
Collapse
|