1
|
Hayat M, Syed RA, Qaiser H, Uzair M, Al-Regaiey K, Khallaf R, Albassam LAM, Kaleem I, Wang X, Wang R, Bhatti MS, Bashir S. Decoding molecular mechanisms: brain aging and Alzheimer's disease. Neural Regen Res 2025; 20:2279-2299. [PMID: 39104174 PMCID: PMC11759015 DOI: 10.4103/nrr.nrr-d-23-01403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 01/23/2024] [Accepted: 07/04/2024] [Indexed: 08/07/2024] Open
Abstract
The complex morphological, anatomical, physiological, and chemical mechanisms within the aging brain have been the hot topic of research for centuries. The aging process alters the brain structure that affects functions and cognitions, but the worsening of such processes contributes to the pathogenesis of neurodegenerative disorders, such as Alzheimer's disease. Beyond these observable, mild morphological shifts, significant functional modifications in neurotransmission and neuronal activity critically influence the aging brain. Understanding these changes is important for maintaining cognitive health, especially given the increasing prevalence of age-related conditions that affect cognition. This review aims to explore the age-induced changes in brain plasticity and molecular processes, differentiating normal aging from the pathogenesis of Alzheimer's disease, thereby providing insights into predicting the risk of dementia, particularly Alzheimer's disease.
Collapse
Affiliation(s)
- Mahnoor Hayat
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rafay Ali Syed
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hammad Qaiser
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad (IIUI), Islamabad, Pakistan
| | - Mohammad Uzair
- Department of Bioengineering, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Khallaf
- Department of Neurology, Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | | | - Imdad Kaleem
- Department of Biosciences, Commission on Science and Technology for Sustainable Development in the South (COMSATS University), Islamabad, Pakistan
| | - Xueyi Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Ran Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Mehwish S. Bhatti
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
2
|
Chun MY, Park YH, Kim HJ, Na DL, Kim JP, Seo SW, Jang H. Distinct Characteristics of Suspected Non-Alzheimer Pathophysiology in Relation to Cognitive Status and Cerebrovascular Burden. Clin Nucl Med 2025; 50:368-380. [PMID: 40025666 PMCID: PMC11969373 DOI: 10.1097/rlu.0000000000005793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/23/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE OF THE REPORT This study investigated the prevalence and clinical characteristics of suspected non-Alzheimer disease pathophysiology (SNAP) across varying cognitive statuses and cerebral small vessel disease (CSVD) burden. PATIENTS AND METHODS We included 1992 participants with cognitive status categorized as cognitively unimpaired, mild cognitive impairment, or dementia. β-amyloid (Aβ, A) positivity was assessed by Aβ PET, and neurodegeneration (N) positivity was determined through hippocampal volume. Participants were further divided by the presence or absence of severe CSVD. The clinical and imaging characteristics of A-N+ (SNAP) group were compared with those of the A-N- and A+N+ groups. RESULTS SNAP participants were older and had more vascular risk factors compared with A-N- and A+N+ in the CSVD(-) cohort. SNAP and A+N+ showed similar cortical thinning. At the dementia stage, SNAP had a cognitive trajectory similar to A+N+ in the CSVD(-) cohort. However, SNAP exhibited less cognitive decline than A+N+ in the CSVD(+) cohort. CONCLUSIONS SNAP is characterized by distinct clinical and imaging characteristics; however, it does not necessarily indicate a benign prognosis, particularly at the dementia stage. These findings highlight the need to assess SNAP in relation to the cognitive stage and CSVD presence to better understand its progression and guide interventions.
Collapse
Affiliation(s)
- Min Young Chun
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine
- Department of Neurology, Yonsei University College of Medicine
| | - Yu Hyun Park
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University
- Department of Digital Health, SAIHST, Sungkyunkwan University
| | - Duk L. Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University
- Department of Digital Health, SAIHST, Sungkyunkwan University
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Gangnam-gu
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-gu, Seoul, South Korea
| |
Collapse
|
3
|
Pan W, Teng Y, Han X, Liu S, Pang X, Wang L, Zhao M. Value of blood neural cell-derived small extracellular vesicles in the diagnosis and prediction of Alzheimer's disease: A systematic revie. J Prev Alzheimers Dis 2025:100193. [PMID: 40316481 DOI: 10.1016/j.tjpad.2025.100193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025]
Abstract
Blood neural cell-derived small extracellular vesicles (sEVs) can directly reflect changes in brain tissue and are easier to obtain than cerebrospinal fluid. This article systematically reviews the alterations of proteins and miRNAs from neural cell-derived sEVs in patients with Alzheimer's disease (AD), and summarizes the biomarkers with clinical diagnostic and predictive value. PubMed, Web of Science, Embase, and Cochrane Library were searched for studies in blood neural cell-derived sEVs in AD patients up to May 2024. According to the inclusion and exclusion criteria, the literature was screened, the information was extracted and the quality was evaluated. Proteins and miRNAs from neural cell-derived sEVs were classified and summarized, focusing on target molecules with high diagnostic and predictive values for AD. A final 34 articles reporting 5601 participants were included. In cross-sectional studies, Aβ- and Tau-related proteins (Aβ42, Aβ42/40, p-S396-Tau, p-Tau181), p-S312-IRS-1, and cathepsin D were increased, conversely, synaptic proteins (neurogranin, synaptotagmin, synaptophysin, synaptopodin, NMDAR2A) and REST were decreased in blood neuron-derived sEVs (NDsEVs) of patients with AD. While miR-29c-3p was increased in blood NDsEVs and glial cell-derived sEVs. Each of these proteins and miRNAs demonstrated high AD diagnostic value. Additionally, blood astrocyte-derived sEVs (ADsEVs) showed increased complement effector proteins and decreased complement regulatory proteins with a moderate diagnostic value. In longitudinal cohort studies, three composite models displayed high predictive efficacy for early AD prediction, and could predict the occurrence of AD within 1-10 years. Therefore, Aβ- and Tau-related proteins, synaptic proteins, and miRNA in blood neural cell-derived sEVs demonstrate high AD diagnostic and predictive values serving as important biomarkers. Especially, synaptic proteins showed significant changes in the early clinical stage, which has early predictive value.
Collapse
Affiliation(s)
- Weibing Pan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowan Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shaojiao Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xingxue Pang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Mingjing Zhao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
4
|
Dubbelman MA, Elias U, Palmer P, Dafni-Merom A, Gazit L, Udeogu OJ, Wang S, Papp KV, Amariglio RE, Arzy S, Marshall GA. Investigating the associations between tau and mental orientation among cognitively unimpaired individuals. J Alzheimers Dis 2025:13872877251334781. [PMID: 40267288 DOI: 10.1177/13872877251334781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundImpairments in orientation in space, time, and person occur frequently in Alzheimer's disease (AD) dementia. Subtle changes in orientation may arise in preclinical and prodromal disease stages. Thus, assessing orientation may help identify those on a trajectory toward AD dementia.ObjectiveTo investigate how orientation, measured using a novel artificial intelligence-based paradigm, relates to AD biomarkers (amyloid and tau) in cognitively unimpaired older adults.MethodsUsing an automated chatbot, 53 cognitively unimpaired participants (74.0 ± 5.5 years; 60% female) provided details about memories and relationships, recognition of historical event dates, and geographical locations. These details were then used to assess orientation to space, time, and person. For each domain separately, orientation accuracy was calculated by dividing the number of correct responses by response time. All participants underwent Pittsburgh compound-B (amyloid) and flortaucipir (tau) positron emission tomography. We analyzed the relationship between performance on the three orientation domains and retrosplenial, precuneus, neocortical, and medial temporal tau, and global amyloid.ResultsHigher retrosplenial and precuneus tau burden were associated with worse temporal orientation (β = -0.32, 95% confidence interval [95%CI] = [-0.59, -0.05] and β = -0.29, 95%CI = [-0.57, -0.01], respectively). Spatial or social orientation were not associated with amyloid or tau.ConclusionsThese results suggest that impaired temporal orientation is related to AD pathological processes, even before the onset of overt cognitive impairment, and may infer a role for personalized assessment of orientation in early diagnosis of AD.
Collapse
Affiliation(s)
- Mark A Dubbelman
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Uri Elias
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| | - Phebe Palmer
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amnon Dafni-Merom
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| | - Lidor Gazit
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| | - Onyinye J Udeogu
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sharon Wang
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathryn V Papp
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca E Amariglio
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shahar Arzy
- The Computational Neuropsychiatry Lab, Department of Medical Neurobiology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Neurology, Hadassah Hebrew University Medical School, Jerusalem, Israel
| | - Gad A Marshall
- Department of Neurology, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Abdullah L, Zhou Z, Hall J, Petersen M, Zhang F, O'Bryant S. Association of Alzheimer's disease biomarkers with low premorbid intellectual functioning in a multi-ethnic community-dwelling cohort: A cross-sectional study of HABS-HD. J Alzheimers Dis 2025; 104:1201-1211. [PMID: 40116640 DOI: 10.1177/13872877251322966] [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: 03/23/2025]
Abstract
Background: Individuals with intellectual disability (ID) may have a five-fold increased risk for developing Alzheimer's disease (AD). However, studies investigating brain aging among individuals with ID without Down syndrome (DS) are lacking. To begin addressing this gap, our study utilized word reading, a widely recognized indicator of an individual's premorbid intellectual ability (pIQ), to examine the effects of ID without DS on plasma AD biomarker outcomes. Objective: To investigate the relationship between premorbid intellectual ability (pIQ) and plasma AD biomarkers in individuals with ID without DS, while considering ethnic differences in these associations. Methods: Participants from the Health & Aging Brain Study - Health Disparities (HABS-HD) were categorized into low (z ≤ -2.00) or average (z = 0.00 ± 1.00) pIQ groups based on word reading scores. Plasma biomarkers including Aβ40, Aβ42, Aβ42/40, phosphorylated tau 181 (p-Tau181), neurofilament light chain (NfL), and total tau (t-tau) were assayed using Simoa technology. Results: Individuals with low pIQ exhibited significantly higher levels of p-Tau181 (p < 0.05), NfL (p < 0.05), and t-tau (p < 0.05) compared to those with average pIQ. Stratified analysis by ethnicity revealed differential associations, with Hispanic and non-Hispanic White (NHW) participants showing distinct biomarker profiles relative to non-Hispanic Black (NHB) individuals. Conclusions: The findings demonstrate that low pIQ is a reliable factor associated with plasma AD biomarker outcomes. Ethnicity appears to modulate these associations, suggesting complex interactions between factors driving AD susceptibility across diverse populations. This study highlights the importance of considering both pIQ and ethnicity in neurodegenerative processes, particularly in individuals with non-DS intellectual developmental disability.
Collapse
Affiliation(s)
- Lubnaa Abdullah
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Zhengyang Zhou
- Department of Public Health, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James Hall
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Melissa Petersen
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Fan Zhang
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sid O'Bryant
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
6
|
Jauregi-Zinkunegi A, Wilson RE, E Langhough R, Ashton NJ, Blennow K, Johnson SC, Zetterberg H, Bruno D, Mueller KD. Associations between the logical memory test story recall metrics and plasma biomarkers for Alzheimer's disease in individuals free of dementia. Clin Neuropsychol 2025:1-20. [PMID: 40114424 DOI: 10.1080/13854046.2025.2481119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Objective: Blood-based biomarkers are valued for their lower cost and less invasive nature, though issues with widespread implementation and accessibility remain. Process-based scores from story recall have been shown to detect neuronal network disturbances typical of Alzheimer's disease (AD) pathology more effectively than traditional metrics. This study examined the associations between process-based scores and concurrent plasma AD biomarkers in older adults without dementia, while also comparing them to traditional metrics. Additionally, it also investigated the diagnostic utility of these metrics in detecting plasma p-tau217 positivity. Methods: Data from 416 participants (mean age = 66.6 ± 7) free of dementia were extracted from the Wisconsin Registry for Alzheimer's Prevention (WRAP). Logical Memory Test (LMT) and plasma p-tau217, p-tau181, p-tau231, Aβ42/Aβ40 ratio, GFAP and NfL levels were analyzed. Bayesian regression models assessed associations between plasma biomarkers and both process-based and traditional LMT scores, controlling for the covariates. Results: The best-fitting model for plasma p-tau217 included Total ratio (Tr) and Immediate recall (BF10=573), but Tr showed stronger evidence of association (mean coefficient = 0.208; BFinclusion=14.4) than Immediate recall (mean coefficient=-0.007; BFinclusion=1.7). Tr was also the best predictor of plasma p-tau181 (mean coefficient = 0.144; BF10=10.5) and GFAP (mean coefficient = 0.141; BF10=5.8), outperforming traditional LMT scores. No memory scores were associated with plasma p-tau231 or Aβ42/40 ratio levels. Tr score was the strongest single predictor of p-tau217 positivity (BF10=38). Conclusions: These findings suggest that process-based memory scores might be useful in enhancing the detection of neuronal network disturbances associated with AD pathology, especially in settings where biomarker testing is unavailable.
Collapse
Affiliation(s)
| | - Rachael E Wilson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Rebecca E Langhough
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, 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, Hong Kong, China
| | - Davide Bruno
- School of Psychology, Liverpool John Moores University, Liverpool, UK
| | - Kimberly D Mueller
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
- Department of Communication Sciences and Disorders, University of Wisconsin - Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| |
Collapse
|
7
|
Paulsen AJ, Driscoll I, Breidenbach BM, Glittenberg MP, Lose SR, Ma Y, Sager MA, Carlsson CM, Gallagher CL, Hermann BP, Blennow K, Zetterberg H, Asthana S, Johnson SC, Betthauser TJ, Christian BT, Cook DB, Okonkwo OC. The impact of cardiorespiratory fitness on Alzheimer's disease biomarkers and their relationships with cognitive decline. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.03.25323245. [PMID: 40093252 PMCID: PMC11908334 DOI: 10.1101/2025.03.03.25323245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
INTRODUCTION Relationships between core Alzheimer's disease (AD) biomarker accumulation and cognitive decline are well-established and the literature generally suggests a favorable relationship of cardiorespiratory fitness (CRF) on AD biomarker accumulation and cognition. Differences in risk of biomarker status conversion or accumulation rates by CRF, or their potential interactive relationships with cognitive decline remain largely unknown. METHODS Participants (N=533; MeanAGE=65, 70% female) from the Wisconsin Alzheimer's Disease Research Center and the Wisconsin Registry for Alzheimer's Prevention underwent serial blood draws, and cognitive and imaging assessments (MeanFollow-up=6.0 years). PET imaging of amyloid-β (Aβ) and tau (T) and plasma phosphorylated tau-217 (pTau-217) were used to determine biomarker status (+/-). Sex-specific estimated CRF (eCRF) tertiles were created using a validated equation. Kaplan-Meier survival curves and Cox-proportional hazards models characterized the risk of becoming biomarker-positive. Linear mixed effects models estimated associations between baseline eCRF and core AD biomarker accumulation and whether eCRF modified relationships between biomarker accumulation and cognitive decline. Analyses were stratified by biomarker +/- status. RESULTS No significant relationships were observed between eCRF and biomarker trajectories. However, those in the high eCRF group who were also Aβ- (HR[95%CI]=0.42[0.20, 0.88]) and pTau-217-(HR[95%CI]=0.45[0.21, 0.97]) at baseline had a significantly lower risk of becoming biomarker-positive. There was a significant attenuation of the detrimental relationship between Aβ accumulation and cognitive decline for those with high eCRF and Aβ+/T+. DISCUSSION While CRF did not influence core AD biomarker accumulation trajectories, high CRF did seem to protect against becoming biomarker-positive and attenuate the known deleterious relationship between biomarker accumulation and cognitive decline in Aβ+/T+.
Collapse
Affiliation(s)
- A J Paulsen
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
| | - I Driscoll
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
| | - B M Breidenbach
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
| | - M P Glittenberg
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
| | - S R Lose
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
| | - Y Ma
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
| | - M A Sager
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
| | - C M Carlsson
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
- Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, 2500 Overlook Terrace, Madison, Wisconsin, USA, 53705
| | - C L Gallagher
- Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, 2500 Overlook Terrace, Madison, Wisconsin, USA, 53705
- Departmentof Neurology, University of Wisconsin School of Medicine and Public Health, 1685 Highland Avenue, Madison, Wisconsin, 53705, USA
| | - B P Hermann
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
- Departmentof Neurology, University of Wisconsin School of Medicine and Public Health, 1685 Highland Avenue, Madison, Wisconsin, 53705, USA
| | - K Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Box 100, 405 30 Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-43180, Göteborg, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, 47 Bd de l'Hôpital, 75013, 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, No.96, JinZhai Road Baohe District, Hefei, Anhui, 230026, P.R. China
| | - H Zetterberg
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Box 100, 405 30 Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, SE-43180, Göteborg, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, Gower Street, London, WC1E 6BT, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Units 1501-1502, 1512-1518, 15/F Building 17W, 17 Science Park W Ave, Science Park, Hong Kong, PR China
| | - S Asthana
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
- Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, 2500 Overlook Terrace, Madison, Wisconsin, USA, 53705
| | - S C Johnson
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
| | - T J Betthauser
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
| | - B T Christian
- Department of Medical Physics, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 111 Highland Ave, Room 1005, Madison, Wisconsin, USA, 53705
| | - D B Cook
- William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, Wisconsin, USA, 53705
- Department of Kinesiology, University of Wisconsin School of Education, 1300 University Avenue, 285 Medical Sciences Center, Madison, Wisconsin, USA, 53706
| | - O C Okonkwo
- Wisconsin Alzheimer's Disease Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC2420, Madison, Wisconsin, 53792, USA
- Wisconsin Alzheimer's Institute, 610 Walnut St, Suite 957, Madison, Wisconsin, 53726, USA
- Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, 2500 Overlook Terrace, Madison, Wisconsin, USA, 53705
| |
Collapse
|
8
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated Appropriate Use Criteria for Amyloid and Tau PET: A Report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. J Nucl Med 2025:jnumed.124.268756. [PMID: 39778970 DOI: 10.2967/jnumed.124.268756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 01/11/2025] Open
Abstract
The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. Methods: The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. Results: For amyloid PET, 7 scenarios were rated as appropriate, 2 as uncertain, and 8 as rarely appropriate. For tau PET, 5 scenarios were rated as appropriate, 6 as uncertain, and 6 as rarely appropriate. Conclusion: AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Gil D Rabinovici
- Department of Neurology and Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California;
| | - David S Knopman
- Mayo Clinic Neurology and Neurosurgery, Rochester, Minnesota
| | - Javier Arbizu
- Department of Nuclear Medicine, University of Navarra Clinic, Pamplona, Spain
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri; Knight Alzheimer's Disease Research Center, School of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Kevin J Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Peter Herscovitch
- Positron Emission Tomography Department, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Phillip H Kuo
- Medical Imaging, Medicine, and Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Jennifer H Lingler
- Department of Health and Community Systems, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Satoshi Minoshima
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah
| | | | - Julie C Price
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Stephen P Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine, Brown University, Providence, Rhode Island
- Butler Hospital Memory and Aging Program, Providence, Rhode Island
| | | | | | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
- Molecular Neuroimaging, Massachusetts General Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
- Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
9
|
Al Amin M, Dehbia Z, Nafady MH, Zehravi M, Kumar KP, Haque MA, Baig MS, Farhana A, Khan SL, Afroz T, Koula D, Tutone M, Nainu F, Ahmad I, Emran TB. Flavonoids and Alzheimer’s disease: reviewing the evidence for neuroprotective potential. Mol Cell Biochem 2025; 480:43-73. [PMID: 38568359 DOI: 10.1007/s11010-023-04922-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2025]
|
10
|
Stephenson HG, Betthauser TJ, Langhough R, Jonaitis E, Du L, Van Hulle C, Kollmorgen G, Quijano‐Rubio C, Chin NA, Okonkwo OC, Carlsson CM, Asthana S, Johnson SC, Blennow K, Zetterberg H, Bendlin BB. Amyloid is associated with accelerated atrophy in cognitively unimpaired individuals. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70089. [PMID: 39996035 PMCID: PMC11848556 DOI: 10.1002/dad2.70089] [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: 08/16/2024] [Revised: 12/13/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION This study examined the association of longitudinal atrophy with baseline cerebrospinal fluid (CSF) amyloid beta (Aβ, A) and phosphorylated tau (p-tau, T) biomarkers (Aβ42/40, p-tau181) in 406 cognitively unimpaired (CU) individuals (6.670 years of follow-up on average, up to 13 imaging visits) to assess whether A+ is associated with Alzheimer's disease-like atrophy and whether this depends on p-tau181 levels. METHODS An A-T- CU group free from abnormal neurodegeneration (N) was identified using a robust normative approach and used to model normal age-related atrophy via z-scoring. Linear mixed-effects models tested differences in longitudinal atrophy between A+ and A-T-N- individuals and between A/T subgroups. RESULTS A+ was associated with worse atrophy within and beyond the medial temporal lobe, even at low levels of p-tau181. DISCUSSION Neurodegeneration likely begins soon after the onset of abnormal Aβ pathology. Clinical intervention at the earliest signs of Aβ pathology may be needed to mitigate further neurodegeneration. Highlights An A-T-N- control group was identified using a robust normative approachA+ was associated with accelerated atrophy in cognitively unimpaired individualsAtrophy was observed even at low p-tau181 levels.
Collapse
|
11
|
Rabinovici GD, Knopman DS, Arbizu J, Benzinger TLS, Donohoe KJ, Hansson O, Herscovitch P, Kuo PH, Lingler JH, Minoshima S, Murray ME, Price JC, Salloway SP, Weber CJ, Carrillo MC, Johnson KA. Updated appropriate use criteria for amyloid and tau PET: A report from the Alzheimer's Association and Society for Nuclear Medicine and Molecular Imaging Workgroup. Alzheimers Dement 2025; 21:e14338. [PMID: 39776249 PMCID: PMC11772739 DOI: 10.1002/alz.14338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION The Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging convened a multidisciplinary workgroup to update appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. METHODS The workgroup identified key research questions that guided a systematic literature review on clinical amyloid/tau PET. Building on this review, the workgroup developed 17 clinical scenarios in which amyloid or tau PET may be considered. A modified Delphi approach was used to rate each scenario by consensus as "rarely appropriate," "uncertain," or "appropriate." Ratings were performed separately for amyloid and tau PET as stand-alone modalities. RESULTS For amyloid PET, seven scenarios were rated as appropriate, two as uncertain, and eight as rarely appropriate. For tau PET, five scenarios were rated as appropriate, six as uncertain, and six as rarely appropriate. DISCUSSION AUC for amyloid and tau PET provide expert recommendations for clinical use of these technologies in the evolving landscape of diagnostics and therapeutics for Alzheimer's disease. HIGHLIGHTS A multidisciplinary workgroup convened by the Alzheimer's Association and the Society of Nuclear Medicine and Molecular Imaging updated the appropriate use criteria (AUC) for amyloid positron emission tomography (PET) and to develop AUC for tau PET. The goal of these updated AUC is to assist clinicians in identifying clinical scenarios in which amyloid or tau PET may be useful for guiding the diagnosis and management of patients who have, or are at risk for, cognitive decline These updated AUC are intended for dementia specialists who spend a significant proportion of their clinical effort caring for patients with cognitive complaints, as well as serve as a general reference for a broader audience interested in implementation of amyloid and tau PET in clinical practice.
Collapse
Affiliation(s)
- Gil D. Rabinovici
- Department of Neurology and Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Javier Arbizu
- Department of Nuclear MedicineUniversity of Navarra ClinicPamplonaSpain
| | - Tammie L. S. Benzinger
- Mallinckrodt Institute of RadiologyWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
- Knight Alzheimer's Disease Research CenterWashington University in St. Louis School of MedicineSt. LouisMissouriUSA
| | - Kevin J. Donohoe
- Nuclear Medicine, Beth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Oskar Hansson
- Department of Clinical Sciences MalmöClinical Memory Research UnitFaculty of MedicineLund UniversityLundSweden
- Memory Clinic, Skåne University HospitalSkånes universitetssjukhusMalmöSweden
| | - Peter Herscovitch
- Positron Emission Tomography DepartmentNational Institutes of Health Clinical CenterBethesdaMarylandUSA
| | - Phillip H. Kuo
- Medical Imaging, Medicine, and Biomedical EngineeringUniversity of ArizonaTucsonArizonaUSA
| | - Jennifer H. Lingler
- Department of Health and Community SystemsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Satoshi Minoshima
- Department of Radiology and Imaging SciencesUniversity of UtahSalt Lake CityUtahUSA
| | | | - Julie C. Price
- Department of RadiologyMassachusetts General Hospital, BostonCharlestownMassachusettsUSA
| | - Stephen P. Salloway
- Department of Neurology and Psychiatry the Warren Alpert School of Medicine at Brown UniversityProvidenceRhode IslandUSA
- Butler Hospital Memory and Aging ProgramProvidenceRhode IslandUSA
| | | | - Maria C. Carrillo
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Keith A. Johnson
- Center for Alzheimer Research and TreatmentDepartment of NeurologyBrigham and Women's HospitalBostonMassachusettsUSA
- Molecular Neuroimaging, Massachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
- Departments of Neurology and RadiologyMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
12
|
Boza-Calvo C, Faustin A, Zhang Y, Briggs AQ, Bernard MA, Bubu OM, Rao JA, Gurin L, Tall SO, Osorio RS, Marsh K, Shao Y, Masurkar AV. Two-Year Longitudinal Outcomes of Subjective Cognitive Decline in Hispanics Compared to Non-hispanic Whites. J Geriatr Psychiatry Neurol 2025; 38:23-31. [PMID: 39043156 DOI: 10.1177/08919887241263097] [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] [Indexed: 07/25/2024]
Abstract
BACKGROUND Subjective cognitive decline (SCD), considered a preclinical dementia stage, is less understood in Hispanics, a high-risk group for dementia. We investigated SCD to mild cognitive impairment (MCI) progression risk, as well as baseline and longitudinal features of depressive symptoms, SCD complaints, and objective cognitive performance among Hispanics compared to non-Hispanic Whites (NHW). METHODS Hispanic (n = 23) and NHW (n = 165) SCD participants were evaluated at baseline and 2-year follow-up. Evaluations assessed function, depressive symptoms, SCD, and objective cognitive performance. RESULTS Hispanics were at increased risk of progression to MCI (OR: 6.10, 95% CI 1.09-34.20, P = .040). Hispanic participants endorsed more depressive symptoms at baseline (P = .048) that worsened more longitudinally (OR: 3.16, 95% CI 1.18-8.51, P = .023). Hispanic participants had increased SCD complaints on the Brief Cognitive Rating Scale (BCRS) (β = .40 SE: .17, P = .023), and in specific BCRS domains: concentration (β = .13, SE: .07, P = .047), past memory (β = .13, SE: .06, P = .039) and functional abilities (β = .10, SE: .05, P = .037). In objective cognitive performance, Hispanic ethnicity associated with decline in MMSE (β = -.27, SE: .13, P = .039), MoCA (β = -.80 SE: .34, P = .032), Trails A (β = 2.75, SE: .89, P = .002), Trails B (β = 9.18, SE: 2.71, P = .001) and Guild Paragraph Recall Delayed (β = -.80 SE: .28, P = .005). Conclusions: Hispanic ethnicity associated with a significantly increased risk of 2-year progression of SCD to MCI compared to NHW. This increased risk associated with increased depressive symptoms, distinctive SCD features, and elevated amnestic and non-amnestic objective cognitive decline. This supports further research to refine the assessment of preclinical dementia in this high-risk group.
Collapse
Affiliation(s)
- Carolina Boza-Calvo
- Centro de Investigación en Hematología y Trastornos Afines (CIHATA), Universidad de Costa Rica, San José, Costa Rica
- Escuela de Medicina, Universidad de Costa Rica, San José, Costa Rica
- NYU Alzheimer's Disease Research Center, NY, USA
| | - Arline Faustin
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Yian Zhang
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Anthony Q Briggs
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Mark A Bernard
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Omonigho M Bubu
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
- Center for Sleep and Brain Health, New York, NY, USA
| | - Julia A Rao
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Lindsey Gurin
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Sakina Ouedraogo Tall
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Ricardo S Osorio
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
- Center for Sleep and Brain Health, New York, NY, USA
| | - Karyn Marsh
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Yongzhao Shao
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Arjun V Masurkar
- NYU Alzheimer's Disease Research Center, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
13
|
Kim YJ, Yun J, Seo SW, Kim JP, Jang H, Kim HJ, Na DL, Woo S, Chun MY. Difference in trajectories according to early amyloid accumulation in cognitively unimpaired elderly. Eur J Neurol 2024; 31:e16482. [PMID: 39275969 PMCID: PMC11555158 DOI: 10.1111/ene.16482] [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: 03/07/2024] [Revised: 08/12/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024]
Abstract
BACKGROUND AND PURPOSE Amyloid β (Aβ), a major biomarker of Alzheimer's disease, leads to tau accumulation, neurodegeneration and cognitive decline. Modelling the trajectory of Aβ accumulation in cognitively unimpaired (CU) individuals is crucial, as treatments targeting Aβ are anticipated. The evolution of Aβ levels was investigated to determine whether it could lead to classification into different groups by studying longitudinal Aβ changes in older CU individuals, and differences between the groups were compared. METHODS A total of 297 CU participants were included from the Alzheimer's Disease Neuroimaging Initiative database, and these participants underwent apolipoprotein E (APOE) genotyping, neuropsychological testing, brain magnetic resonance imaging, and an average of 3.03 follow-up 18F-florbetapir positron emission tomography scans. Distinct Aβ trajectory patterns were classified using latent class growth analysis, and longitudinal cognitive performances across these patterns were assessed with a linear mixed effects model. RESULTS The optimal model consisted of three classes, with a high entropy value of 0.947. The classes were designated as follows: class 1, non-accumulation group (n = 197); class 2, late accumulation group (n = 70); and class 3, early accumulation group (n = 30). The late accumulation and early accumulation groups had more APOE ε4 carriers than the non-accumulation group. The longitudinal analysis of cognitive performance revealed that the early accumulation group showed the steepest decline (modified Preclinical Alzheimer's Cognitive Composite with digit symbol substitution [mPACCdigit], p < 0.001; modified Preclinical Alzheimer's Cognitive Composite with trails B [mPACCtrailsB], p < 0.001) and the late accumulation group showed a steeper decline (mPACCdigit, p = 0.014; mPACCtrailsB, p = 0.007) compared to the non-accumulation group. CONCLUSIONS Our study showed the heterogeneity of Aβ accumulation trajectories in CU older individuals. The prognoses for cognitive decline differ according to the Aβ trajectory patterns.
Collapse
Affiliation(s)
- Young Ju Kim
- Department of Neurology, Samsung Medical CentreSungkyunkwan University School of MedicineSeoulSouth Korea
- Neuroscience CentreSamsung Medical CentreSeoulSouth Korea
| | - Jihwan Yun
- Department of Neurology, Samsung Medical CentreSungkyunkwan University School of MedicineSeoulSouth Korea
- Department of Neurology, Soonchunhyang University Bucheon HospitalSoonchunhyang University School of MedicineBucheonSouth Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical CentreSungkyunkwan University School of MedicineSeoulSouth Korea
- Neuroscience CentreSamsung Medical CentreSeoulSouth Korea
- Department of Digital Health, SAIHSTSungkyunkwan UniversitySeoulSouth Korea
- Department of Health Sciences and Technology, SAIHSTSungkyunkwan UniversitySeoulSouth Korea
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical CentreSungkyunkwan University School of MedicineSeoulSouth Korea
- Neuroscience CentreSamsung Medical CentreSeoulSouth Korea
| | - Hyemin Jang
- Department of Neurology, Samsung Medical CentreSungkyunkwan University School of MedicineSeoulSouth Korea
- Department of Neurology, Seoul National University HospitalSeoul National University School of MedicineSeoulSouth Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical CentreSungkyunkwan University School of MedicineSeoulSouth Korea
- Neuroscience CentreSamsung Medical CentreSeoulSouth Korea
- Department of Digital Health, SAIHSTSungkyunkwan UniversitySeoulSouth Korea
- Department of Health Sciences and Technology, SAIHSTSungkyunkwan UniversitySeoulSouth Korea
| | - Duk L. Na
- Department of Neurology, Samsung Medical CentreSungkyunkwan University School of MedicineSeoulSouth Korea
- Neuroscience CentreSamsung Medical CentreSeoulSouth Korea
| | - Sookyoung Woo
- Biostatistics TeamSamsung Biomedical Research InstituteSeoulSouth Korea
| | - Min Young Chun
- Department of Neurology, Samsung Medical CentreSungkyunkwan University School of MedicineSeoulSouth Korea
- Department of NeurologyYonsei University College of MedicineSeoulSouth Korea
- Department of Neurology, Yongin Severance HospitalYonsei University Health SystemYonginSouth Korea
| | | |
Collapse
|
14
|
van Gils AM, Tolonen A, van Harten AC, Vigneswaran S, Barkhof F, Visser LNC, Koikkalainen J, Herukka SK, Hasselbalch SG, Mecocci P, Remes AM, Soininen H, Lemstra AW, Teunissen CE, Jönsson L, Lötjönen J, van der Flier WM, Rhodius-Meester HFM. Computerized decision support to optimally funnel patients through the diagnostic pathway for dementia. Alzheimers Res Ther 2024; 16:256. [PMID: 39587679 PMCID: PMC11590510 DOI: 10.1186/s13195-024-01614-5] [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: 03/15/2024] [Accepted: 10/31/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND The increasing prevalence of dementia and the introduction of disease-modifying therapies (DMTs) highlight the need for efficient diagnostic pathways in memory clinics. We present a data-driven approach to efficiently guide stepwise diagnostic testing for three clinical scenarios: 1) syndrome diagnosis, 2) etiological diagnosis, and 3) eligibility for DMT. METHODS We used data from two memory clinic cohorts (ADC, PredictND), including 504 patients with dementia (302 Alzheimer's disease, 107 frontotemporal dementia, 35 vascular dementia, 60 dementia with Lewy bodies), 191 patients with mild cognitive impairment, and 188 cognitively normal controls (CN). Tests included digital cognitive screening (cCOG), neuropsychological and functional assessment (NP), MRI with automated quantification, and CSF biomarkers. Sequential testing followed a predetermined order, guided by diagnostic certainty. Diagnostic certainty was determined using a clinical decision support system (CDSS) that generates a disease state index (DSI, 0-1), indicating the probability of the syndrome diagnosis or underlying etiology. Diagnosis was confirmed if the DSI exceeded a predefined threshold based on sensitivity/specificity cutoffs relevant to each clinical scenario. Diagnostic accuracy and the need for additional testing were assessed at each step. RESULTS Using cCOG as a prescreener for 1) syndrome diagnosis has the potential to accurately reduce the need for extensive NP (42%), resulting in syndrome diagnosis in all patients, with a diagnostic accuracy of 0.71, which was comparable to using NP alone. For 2) etiological diagnosis, stepwise testing resulted in an etiological diagnosis in 80% of patients with a diagnostic accuracy of 0.77, with MRI needed in 77%, and CSF in 37%. When 3) determining DMT eligibility, stepwise testing (100% cCOG, 83% NP, 75% MRI) selected 60% of the patients for confirmatory CSF testing and eventually identified 90% of the potentially eligible patients with AD dementia. CONCLUSIONS Different diagnostic pathways are accurate and efficient depending on the setting. As such, a data-driven tool holds promise for assisting clinicians in selecting tests of added value across different clinical contexts. This becomes especially important with DMT availability, where the need for more efficient diagnostic pathways is crucial to maintain the accessibility and affordability of dementia diagnoses.
Collapse
Affiliation(s)
- Aniek M van Gils
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands.
| | | | - Argonde C van Harten
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
| | - Sinthujah Vigneswaran
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, 1081HV, The Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Leonie N C Visser
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
- Department of Medical Psychology, Amsterdam UMC, Amsterdam, 1081HV, The Netherlands
- Amsterdam Public Health, Quality of Care, Amsterdam, 1081HV, The Netherlands
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Steen Gregers Hasselbalch
- Danish Dementia Research Centre, University of Copenhagen, Blegdamsvej 9, 2100, RigshospitaletCopenhagen, Denmark
| | - Patrizia Mecocci
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06129, Perugia, Italy
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, SE, Sweden
| | - Anne M Remes
- Research Unit of Clinical Medicine, Neurology, University of Oulu, 90014, Oulu, Finland
| | - Hilkka Soininen
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Afina W Lemstra
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Neurochemistry Laboratory, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, 1081HV, The Netherlands
| | - Linus Jönsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | - Wiesje M van der Flier
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
- Department of Epidemiology and Data Sciences, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, 1081HV, the Netherlands
| | - Hanneke F M Rhodius-Meester
- Alzheimer Center Amsterdam and Department of Neurology, VU University Medical Center, Amsterdam UMC, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081HV, The Netherlands
- Department of Internal Medicine, Geriatric Medicine Section, Amsterdam Cardiovascular Sciences Institute, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, 1081HV, The Netherlands
- Department of Geriatric Medicine, The Memory Clinic, Oslo University Hospital, 0379, Oslo, Norway
| |
Collapse
|
15
|
Chua J, Tan B, Wong D, Garhöfer G, Liew XW, Popa-Cherecheanu A, Loong Chin CW, Milea D, Li-Hsian Chen C, Schmetterer L. Optical coherence tomography angiography of the retina and choroid in systemic diseases. Prog Retin Eye Res 2024; 103:101292. [PMID: 39218142 DOI: 10.1016/j.preteyeres.2024.101292] [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: 05/17/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Optical coherence tomography angiography (OCTA) has transformed ocular vascular imaging, revealing microvascular changes linked to various systemic diseases. This review explores its applications in diabetes, hypertension, cardiovascular diseases, and neurodegenerative diseases. While OCTA provides a valuable window into the body's microvasculature, interpreting the findings can be complex. Additionally, challenges exist due to the relative non-specificity of its findings where changes observed in OCTA might not be unique to a specific disease, variations between OCTA machines, the lack of a standardized normative database for comparison, and potential image artifacts. Despite these limitations, OCTA holds immense potential for the future. The review highlights promising advancements like quantitative analysis of OCTA images, integration of artificial intelligence for faster and more accurate interpretation, and multi-modal imaging combining OCTA with other techniques for a more comprehensive characterization of the ocular vasculature. Furthermore, OCTA's potential future role in personalized medicine, enabling tailored treatment plans based on individual OCTA findings, community screening programs for early disease detection, and longitudinal studies tracking disease progression over time is also discussed. In conclusion, OCTA presents a significant opportunity to improve our understanding and management of systemic diseases. Addressing current limitations and pursuing these exciting future directions can solidify OCTA as an indispensable tool for diagnosis, monitoring disease progression, and potentially guiding treatment decisions across various systemic health conditions.
Collapse
Affiliation(s)
- Jacqueline Chua
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Bingyao Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore
| | - Damon Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore; Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Gerhard Garhöfer
- Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria
| | - Xin Wei Liew
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Alina Popa-Cherecheanu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Emergency University Hospital, Department of Ophthalmology, Bucharest, Romania
| | - Calvin Woon Loong Chin
- Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Dan Milea
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Fondation Ophtalmologique Adolphe De Rothschild, Paris, France
| | - Christopher Li-Hsian Chen
- Memory Aging and Cognition Centre, Departments of Pharmacology and Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Leopold Schmetterer
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore; Academic Clinical Program, Duke-NUS Medical School, National University of Singapore, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore; Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland; Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria; Fondation Ophtalmologique Adolphe De Rothschild, Paris, France; Center for Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna, Austria.
| |
Collapse
|
16
|
Antoniades M, Srinivasan D, Wen J, Erus G, Abdulkadir A, Mamourian E, Melhem R, Hwang G, Cui Y, Govindarajan ST, Chen AA, Zhou Z, Yang Z, Chen J, Pomponio R, Sotardi S, An Y, Bilgel M, LaMontagne P, Singh A, Benzinger T, Beason-Held L, Marcus DS, Yaffe K, Launer L, Morris JC, Tosun D, Ferrucci L, Bryan RN, Resnick SM, Habes M, Wolk D, Fan Y, Nasrallah IM, Shou H, Davatzikos C. Relationship between MRI brain-age heterogeneity, cognition, genetics and Alzheimer's disease neuropathology. EBioMedicine 2024; 109:105399. [PMID: 39437659 PMCID: PMC11536027 DOI: 10.1016/j.ebiom.2024.105399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Brain ageing is highly heterogeneous, as it is driven by a variety of normal and neuropathological processes. These processes may differentially affect structural and functional brain ageing across individuals, with more pronounced ageing (older brain age) during midlife being indicative of later development of dementia. Here, we examined whether brain-ageing heterogeneity in unimpaired older adults related to neurodegeneration, different cognitive trajectories, genetic and amyloid-beta (Aβ) profiles, and to predicted progression to Alzheimer's disease (AD). METHODS Functional and structural brain age measures were obtained for resting-state functional MRI and structural MRI, respectively, in 3460 cognitively normal individuals across an age range spanning 42-85 years. Participants were categorised into four groups based on the difference between their chronological and predicted age in each modality: advanced age in both (n = 291), resilient in both (n = 260) or advanced in one/resilient in the other (n = 163/153). With the resilient group as the reference, brain-age groups were compared across neuroimaging features of neuropathology (white matter hyperintensity volume, neuronal loss measured with Neurite Orientation Dispersion and Density Imaging, AD-specific atrophy patterns measured with the Spatial Patterns of Abnormality for Recognition of Early Alzheimer's Disease index, amyloid burden using amyloid positron emission tomography (PET), progression to mild cognitive impairment and baseline and longitudinal cognitive measures (trail making task, mini mental state examination, digit symbol substitution task). FINDINGS Individuals with advanced structural and functional brain-ages had more features indicative of neurodegeneration and they had poor cognition. Individuals with a resilient brain-age in both modalities had a genetic variant that has been shown to be associated with age of onset of AD. Mixed brain-age was associated with selective cognitive deficits. INTERPRETATION The advanced group displayed evidence of increased atrophy across all neuroimaging features that was not found in either of the mixed groups. This is in line with biomarkers of preclinical AD and cerebrovascular disease. These findings suggest that the variation in structural and functional brain ageing across individuals reflects the degree of underlying neuropathological processes and may indicate the propensity to develop dementia in later life. FUNDING The National Institute on Aging, the National Institutes of Health, the Swiss National Science Foundation, the Kaiser Foundation Research Institute and the National Heart, Lung, and Blood Institute.
Collapse
Affiliation(s)
- Mathilde Antoniades
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Dhivya Srinivasan
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA
| | - Guray Erus
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Department of Clinical Neuroscience, Center for Research in Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - Elizabeth Mamourian
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yuhan Cui
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja Tirumalai Govindarajan
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew A Chen
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Zhen Zhou
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiong Chen
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymond Pomponio
- Department of Biostatistics, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Susan Sotardi
- Department of Radiology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ashish Singh
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, USA
| | - Tammie Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lori Beason-Held
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Lenore Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institute of Health, Baltimore, MD 21224, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mohamad Habes
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - David Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, USA
| | - Christos Davatzikos
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Desai Y, Karunakaran D, Singh J, Noronha AR, Poojary G, Chettri B, Shenoy R, Nampoothiri M, Bojja SL. Exploring the mechanism of sunflower seed oil against Alzheimer's disease through experimental and network pharmacology studies. Indian J Pharmacol 2024; 56:396-404. [PMID: 39973828 PMCID: PMC11913338 DOI: 10.4103/ijp.ijp_761_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVES With the prevalence of Alzheimer's disease (AD) increasing exponentially, there has been a shift in the focus of drug discovery for AD from treating the symptoms to preventing the development of the disease. Several natural compounds are extensively studied as neuroprotectives in preventing disease progression. Helianthus annuus seed oil (HA) is widely used as cooking oil and is abundant in antioxidant activity. Therefore, we evaluated the effect of HA in mice model of scopolamine-induced amnesia and explored the potential underlying mechanisms. METHODS Twenty-four male mice were administered orally with either distilled water (control and scopolamine groups) or treatment groups (HA 100 and HA 200 mg/kg) for 8 consecutive days. All groups, except the control group, received an intraperitoneal injection of scopolamine at a dose of 1 mg/kg. Subsequently, novel object recognition task for cognition assessment and open field tests for locomotory activity were performed. In addition, network analysis was performed to identify the key bioactives and targets of HA against AD. Further, the binding affinity of HA bioactives to the key targets was verified by molecular docking analysis. RESULTS HA (100 mg/kg and 200 mg/kg) significantly ameliorated recognition memory compared to the scopolamine group, suggesting the protective effect of HA against cognitive impairment. Network analysis indicated that the key bioactives of HA, chlorogenic acid, and oleic acid act through multiple targets and pathways, particularly the mitogen-activated protein kinase (MAPK) pathway, to ameliorate AD symptoms. Importantly, chlorogenic acid showed good binding affinity with MAPKs, TP53, and EP300. CONCLUSION HA has therapeutic benefits in AD acting through the MAPK pathway. However, further studies need to be done to confirm the results derived and translate the potential use of HA as a dietary supplement for preventing AD.
Collapse
Affiliation(s)
- Yashodhan Desai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Cappa SF, Ribaldi F, Chicherio C, Frisoni GB. Subjective cognitive decline: Memory complaints, cognitive awareness, and metacognition. Alzheimers Dement 2024; 20:6622-6631. [PMID: 39051174 PMCID: PMC11497716 DOI: 10.1002/alz.13905] [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: 06/07/2023] [Revised: 03/20/2024] [Accepted: 04/20/2024] [Indexed: 07/27/2024]
Abstract
Cognitive complaints are common in elderly subjects and are a frequent reason for referral to memory clinics. If the complaints are not associated with objective cognitive impairment, the condition is labelled subjective cognitive decline (SCD). SCD is often considered as a stage antedating objective impairment, and an at-risk condition for subsequent dementia. Recent large-scale studies indicate that a significantly increased risk of clinical progression in subjects with SCD is associated with positivity for Alzheimer's disease (AD) biomarkers, a finding supporting the notion that SCD can be due to different mechanisms not associated with neurodegeneration, including functional cognitive disorders. In this paper we present a selective review of research on the relations among SCD, cognitive awareness, and metacognitive abilities. We propose that longitudinal studies of metacognitive efficiency in SCD may provide useful cues about the risk of progression to dementia and the possible presence of a functional cognitive disorder, with different implications for the management of this prevalent aging-related condition. HIGHLIGHTS: Subjective cognitive decline (SCD), a common cause of referral to memory clinics, can be due to multiple conditions. The predictive value of SCD for progression to Alzheimer's disease (AD) dementia is high in association with AD biomarker positivity. The awareness of cognitive decline is the mechanism responsible for the emergence of SCD and metacognition is the underlying neuropsychological function. The awareness of cognitive decline in clinical patients is usually assessed comparing an informant rating to the patient self-assessment, a method that can be affected by informant bias. While there is strong evidence that awareness starts to decline with the onset of objective cognitive impairment, progressively leading to the anosognosia of AD, the status of metacognitive efficiency in SCD needs to be further investigated. Quantitative, performance-based indexes of metacognitive efficiency may contribute both to the assessment of progression risk and to the management of subjects with functional cognitive disorders.
Collapse
Affiliation(s)
- Stefano F. Cappa
- University Institute of Advanced Studies and IRCCS Mondino Foundation PaviaPaviaItaly
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE)University of GenevaGenevaSwitzerland
- Geneva Memory CenterDepartment of Rehabilitation and GeriatricsGeneva University HospitalsGenevaSwitzerland
| | - Christian Chicherio
- Geneva Memory CenterDepartment of Rehabilitation and GeriatricsGeneva University HospitalsGenevaSwitzerland
- Center for Interdisciplinary Study of Gerontology and Vulnerability (CIGEV)University of GenevaGenevaSwitzerland
| | - Giovanni B. Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE)University of GenevaGenevaSwitzerland
- Geneva Memory CenterDepartment of Rehabilitation and GeriatricsGeneva University HospitalsGenevaSwitzerland
| |
Collapse
|
19
|
Wang J, Ackley S, Woodworth DC, Sajjadi SA, Decarli CS, Fletcher EF, Glymour MM, Jiang L, Kawas C, Corrada MM. Associations of Amyloid Burden, White Matter Hyperintensities, and Hippocampal Volume With Cognitive Trajectories in the 90+ Study. Neurology 2024; 103:e209665. [PMID: 39008782 PMCID: PMC11249511 DOI: 10.1212/wnl.0000000000209665] [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: 02/16/2024] [Accepted: 05/10/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Amyloid pathology, vascular disease pathology, and pathologies affecting the medial temporal lobe are associated with cognitive trajectories in older adults. However, only limited evidence exists on how these pathologies influence cognition in the oldest old. We evaluated whether amyloid burden, white matter hyperintensity (WMH) volume, and hippocampal volume (HV) are associated with cognitive level and decline in the oldest old. METHODS This was a longitudinal, observational community-based cohort study. We included participants with 18F-florbetapir PET and MRI data from the 90+ Study. Amyloid load was measured using the standardized uptake value ratio in the precuneus/posterior cingulate with eroded white matter mask as reference. WMH volume was log-transformed. All imaging measures were standardized using sample means and SDs. HV and log-WMH volume were normalized by total intracranial volume using the residual approach. Global cognitive performance was measured by the Mini-Mental State Examination (MMSE) and modified MMSE (3MS) tests, repeated every 6 months. We used linear mixed-effects models with random intercepts; random slopes; and interaction between time, time squared, and imaging variables to estimate the associations of imaging variables with cognitive level and cognitive decline. Models were adjusted for demographics, APOE genotype, and health behaviors. RESULTS The sample included 192 participants. The mean age was 92.9 years, 125 (65.1%) were female, 71 (37.0%) achieved a degree beyond college, and the median follow-up time was 3.0 years. A higher amyloid load was associated with a lower cognitive level (βMMSE = -0.82, 95% CI -1.17 to -0.46; β3MS = -2.77, 95% CI -3.69 to -1.84). A 1-SD decrease in HV was associated with a 0.70-point decrease in the MMSE score (95% CI -1.14 to -0.27) and a 2.27-point decrease in the 3MS score (95% CI -3.40 to -1.14). Clear nonlinear cognitive trajectories were detected. A higher amyloid burden and smaller HV were associated with faster cognitive decline. WMH volume was not significantly associated with cognitive level or decline. DISCUSSION Amyloid burden and hippocampal atrophy are associated with both cognitive level and cognitive decline in the oldest old. Our findings shed light on how different pathologies contributed to driving cognitive function in the oldest old.
Collapse
Affiliation(s)
- Jingxuan Wang
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Sarah Ackley
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Davis C Woodworth
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Seyed Ahmad Sajjadi
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Charles S Decarli
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Evan F Fletcher
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - M Maria Glymour
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Luohua Jiang
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Claudia Kawas
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| | - Maria M Corrada
- From the Department of Epidemiology and Biostatistics (J.W.), University of California, San Francisco; Department of Epidemiology (J.W., S.A., M.M.G.), Boston University, MA; Department of Neurology (D.C.W., S.A.S., C.K., M.M.C.), University of California, Irvine; Imaging of Dementia and Aging Laboratory (C.S.D., E.F.F.), Department of Neurology, University of California, Davis; and Department of Epidemiology and Biostatistics (L.J., M.M.C.), and Department of Neurobiology and Behavior (C.K.), University of California, Irvine
| |
Collapse
|
20
|
Babulal GM, Chen L, Murphy SA, Carr DB, Morris JC. Predicting Driving Cessation Among Cognitively Normal Older Drivers: The Role of Alzheimer Disease Biomarkers and Clinical Assessments. Neurology 2024; 102:e209426. [PMID: 38776513 PMCID: PMC11226325 DOI: 10.1212/wnl.0000000000209426] [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: 01/04/2024] [Accepted: 03/11/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVES With the aging US population and increasing incidence of Alzheimer disease (AD), understanding factors contributing to driving cessation among older adults is crucial for clinicians. Driving is integral for maintaining independence and functional mobility, but the risk factors for driving cessation, particularly in the context of normal aging and preclinical AD, are not well understood. We studied a well-characterized community cohort to examine factors associated with driving cessation. METHODS This prospective, longitudinal observation study enrolled participants from the Knight Alzheimer Disease Research Center and The DRIVES Project. Participants were enrolled if they were aged 65 years or older, drove weekly, and were cognitively normal (Clinical Dementia Rating [CDR] = 0) at baseline. Participants underwent annual clinical, neurologic, and neuropsychological assessments, including β-amyloid PET imaging and CSF (Aβ42, total tau [t-Tau], and phosphorylated tau [p-Tau]) collection every 2-3 years. The primary outcome was time from baseline visit to driving cessation, accounting for death as a competing risk. The cumulative incidence function of driving cessation was estimated for each biomarker. The Fine and Gray subdistribution hazard model was used to examine the association between time to driving cessation and biomarkers adjusting for clinical and demographic covariates. RESULTS Among the 283 participants included in this study, there was a mean follow-up of 5.62 years. Driving cessation (8%) was associated with older age, female sex, progression to symptomatic AD (CDR ≥0.5), and poorer performance on a preclinical Alzheimer cognitive composite (PACC) score. Aβ PET imaging did not independently predict driving cessation, whereas CSF biomarkers, specifically t-Tau/Aβ42 (hazard ratio [HR] 2.82, 95% CI 1.23-6.44, p = 0.014) and p-Tau/Aβ42 (HR 2.91, 95% CI 1.28-6.59, p = 0.012) ratios, were independent predictors in the simple model adjusting for age, education, and sex. However, in the full model, progression to cognitive impairment based on the CDR and PACC score across each model was associated with a higher risk of driving cessation, whereas AD biomarkers were not statistically significant. DISCUSSION Female sex, CDR progression, and neuropsychological measures of cognitive functioning obtained in the clinic were strongly associated with future driving cessation. The results emphasize the need for early planning and conversations about driving retirement in the context of cognitive decline and the immense value of clinical measures in determining functional outcomes.
Collapse
Affiliation(s)
- Ganesh M Babulal
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - Ling Chen
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - Samantha A Murphy
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - David B Carr
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| | - John C Morris
- From the Department of Neurology (G.M.B., S.A.M., J.C.M.), Division of Biostatistics (L.C.), and Department of Medicine (D.B.C.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
21
|
Enzlein T, Lashley T, Sammour DA, Hopf C, Chávez-Gutiérrez L. Integrative Single-Plaque Analysis Reveals Signature Aβ and Lipid Profiles in the Alzheimer's Brain. Anal Chem 2024; 96:9799-9807. [PMID: 38830618 PMCID: PMC11190877 DOI: 10.1021/acs.analchem.3c05557] [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: 12/06/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/05/2024]
Abstract
Cerebral accumulation of amyloid-β (Aβ) initiates molecular and cellular cascades that lead to Alzheimer's disease (AD). However, amyloid deposition does not invariably lead to dementia. Amyloid-positive but cognitively unaffected (AP-CU) individuals present widespread amyloid pathology, suggesting that molecular signatures more complex than the total amyloid burden are required to better differentiate AD from AP-CU cases. Motivated by the essential role of Aβ and the key lipid involvement in AD pathogenesis, we applied multimodal mass spectrometry imaging (MSI) and machine learning (ML) to investigate amyloid plaque heterogeneity, regarding Aβ and lipid composition, in AP-CU versus AD brain samples at the single-plaque level. Instead of focusing on a population mean, our analytical approach allowed the investigation of large populations of plaques at the single-plaque level. We found that different (sub)populations of amyloid plaques, differing in Aβ and lipid composition, coexist in the brain samples studied. The integration of MSI data with ML-based feature extraction further revealed that plaque-associated gangliosides GM2 and GM1, as well as Aβ1-38, but not Aβ1-42, are relevant differentiators between the investigated pathologies. The pinpointed differences may guide further fundamental research investigating the role of amyloid plaque heterogeneity in AD pathogenesis/progression and may provide molecular clues for further development of emerging immunotherapies to effectively target toxic amyloid assemblies in AD therapy. Our study exemplifies how an integrative analytical strategy facilitates the unraveling of complex biochemical phenomena, advancing our understanding of AD from an analytical perspective and offering potential avenues for the refinement of diagnostic tools.
Collapse
Affiliation(s)
- Thomas Enzlein
- Center
for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack Str. 10, Mannheim 68163, Germany
- KU
Leuven-VIB Center for Brain & Disease Research, VIB, Leuven 3000, Belgium
- Department
of Neurosciences, Leuven Institute for Neuroscience and Disease, KU Leuven, Leuven 3000, Belgium
| | - Tammaryn Lashley
- Department
of Neurodegenerative Diseases, UCL Queen
Square Institute of Neurology, London WC1N 3BG, U.K.
| | - Denis Abu Sammour
- Center
for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack Str. 10, Mannheim 68163, Germany
| | - Carsten Hopf
- Center
for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Paul-Wittsack Str. 10, Mannheim 68163, Germany
- Mannheim
Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Heidelberg 69120, Germany
- Medical Faculty, Heidelberg University, Heidelberg 69120, Germany
| | - Lucía Chávez-Gutiérrez
- KU
Leuven-VIB Center for Brain & Disease Research, VIB, Leuven 3000, Belgium
- Department
of Neurosciences, Leuven Institute for Neuroscience and Disease, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
22
|
Iqbal I, Saqib F, Mubarak Z, Latif MF, Wahid M, Nasir B, Shahzad H, Sharifi-Rad J, Mubarak MS. Alzheimer's disease and drug delivery across the blood-brain barrier: approaches and challenges. Eur J Med Res 2024; 29:313. [PMID: 38849950 PMCID: PMC11161981 DOI: 10.1186/s40001-024-01915-3] [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: 02/07/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
Alzheimer's disease (AD) is a diverse disease with a complex pathophysiology. The presence of extracellular β-amyloid deposition as neuritic plaques and intracellular accumulation of hyper-phosphorylated tau as neurofibrillary tangles remain the core neuropathologic criteria for diagnosing Alzheimer's disease. Nonetheless, several recent basic discoveries have revealed significant pathogenic roles for other essential cellular and molecular processes. Previously, there were not so many disease-modifying medications (DMT) available as drug distribution through the blood-brain barrier (BBB) is difficult due to its nature, especially drugs of polypeptides nature and proteins. Recently FDA has approved lecanemab as DMT for its proven efficacy. It is also complicated to deliver drugs for diseases like epilepsy or any brain tumor due to the limitations of the BBB. After the advancements in the drug delivery system, different techniques are used to transport the medication across the BBB. Other methods are used, like enhancement of brain blood vessel fluidity by liposomes, infusion of hyperosmotic solutions, and local intracerebral implants, but these are invasive approaches. Non-invasive approaches include the formulation of nanoparticles and their coating with polymers. This review article emphasizes all the above-mentioned techniques, procedures, and challenges to transporting medicines across the BBB. It summarizes the most recent literature dealing with drug delivery across the BBB.
Collapse
Affiliation(s)
- Iram Iqbal
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
- Primary and Secondary Healthcare Department, Govt of the Punjab, Lahore, Pakistan
| | - Fatima Saqib
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan.
| | - Zobia Mubarak
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
- Primary and Secondary Healthcare Department, Govt of the Punjab, Lahore, Pakistan
| | - Muhammad Farhaj Latif
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Muqeet Wahid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Bushra Nasir
- Department of Pharmaceutics, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Hamna Shahzad
- Department of Biochemistry, Bahauddin Zakariya University Multan, Multan, Pakistan
| | - Javad Sharifi-Rad
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea.
| | - Mohammad S Mubarak
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
23
|
Hu H, Hu H, Jiang J, Bi Y, Sun Y, Ou Y, Tan L, Yu J. Echocardiographic measures of the left heart and cerebrospinal fluid biomarkers of Alzheimer's disease pathology in cognitively intact adults: The CABLE study. Alzheimers Dement 2024; 20:3943-3957. [PMID: 38676443 PMCID: PMC11180853 DOI: 10.1002/alz.13837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 04/28/2024]
Abstract
INTRODUCTION This study delineated the interrelationships between subclinical alterations in the left heart, cerebrospinal fluid (CSF), Alzheimer's disease (AD) biomarkers, and cognition. METHODS Multiple linear regressions were conducted in 1244 cognitively normal participants (mean age = 65.5; 43% female) who underwent echocardiography (left atrial [LA] and left ventricular [LV] morphologic or functional parameters) and CSF AD biomarkers measurements. Mediating effects of AD pathologies were examined. Differences in cardiac parameters across ATN categories were tested using analysis of variance (ANOVA) and logistic regressions. RESULTS LA or LV enlargement (characterized by increased diameters and volumes) and LV hypertrophy (increased interventricular septal or posterior wall thickness and ventricular mass) were associated with higher CSF phosphorylated (p)-tau and total (t)-tau levels, and poorer cognition. Tau pathologies mediated the heart-cognition relationships. Cardiac parameters were higher in stage 2 and suspected non-Alzheimer's pathology groups than controls. DISCUSSION These findings suggested close associations of subclinical cardiac changes with tau pathologies and cognition. HIGHLIGHTS Various subclinical alterations in the left heart related to poorer cognition. Subclinical cardiac changes related to tau pathologies in cognitively normal adults. Tau pathologies mediated the heart-cognition relationships. Subclinical cardiac changes related to the AD continuum, especially to stage 2. The accumulation of cardiac alterations magnified their damage to the brain.
Collapse
Affiliation(s)
- He‐Ying Hu
- Department of NeurologyQingdao Municipal Hospital, Qingdao UniversityQingdaoShandongChina
| | - Hao Hu
- Department of NeurologyQingdao Municipal Hospital, Qingdao UniversityQingdaoShandongChina
| | - Jing Jiang
- Department of Cardiac UltrasonographyQingdao Municipal Hospital, Qingdao UniversityQingdaoShandongChina
| | - Yan‐Lin Bi
- Department of AnesthesiologyQingdao Municipal Hospital, Qingdao UniversityQingdaoShandongChina
| | - Yan Sun
- Department of NeurologyQingdao Municipal Hospital, Qingdao UniversityQingdaoShandongChina
| | - Ya‐Nan Ou
- Department of NeurologyQingdao Municipal Hospital, Qingdao UniversityQingdaoShandongChina
| | - Lan Tan
- Department of NeurologyQingdao Municipal Hospital, Qingdao UniversityQingdaoShandongChina
| | - Jin‐Tai Yu
- Department of Neurology and National Center for Neurological DisordersHuashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan UniversityShanghaiChina
| |
Collapse
|
24
|
Vos SJB, Delvenne A, Jack CR, Thal DR, Visser PJ. The clinical importance of suspected non-Alzheimer disease pathophysiology. Nat Rev Neurol 2024; 20:337-346. [PMID: 38724589 DOI: 10.1038/s41582-024-00962-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 06/06/2024]
Abstract
The development of biomarkers for Alzheimer disease (AD) has led to the origin of suspected non-AD pathophysiology (SNAP) - a heterogeneous biomarker-based concept that describes individuals with normal amyloid and abnormal tau and/or neurodegeneration biomarker status. In this Review, we describe the origins of the SNAP construct, along with its prevalence, diagnostic and prognostic implications, and underlying neuropathology. As we discuss, SNAP can be operationalized using different biomarker modalities, which could affect prevalence estimates and reported characteristics of SNAP in ways that are not yet fully understood. Moreover, the underlying aetiologies that lead to a SNAP biomarker profile, and whether SNAP is the same in people with and without cognitive impairment, remains unclear. Improved insight into the clinical characteristics and pathophysiology of SNAP is of major importance for research and clinical practice, as well as for trial design to optimize care and treatment of individuals with SNAP.
Collapse
Affiliation(s)
- Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.
| | - Aurore Delvenne
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Dietmar R Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology and Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
25
|
Huszár Z, Engh MA, Pavlekovics M, Sato T, Steenkamp Y, Hanseeuw B, Terebessy T, Molnár Z, Hegyi P, Csukly G. Risk of conversion to mild cognitive impairment or dementia among subjects with amyloid and tau pathology: a systematic review and meta-analysis. Alzheimers Res Ther 2024; 16:81. [PMID: 38610055 PMCID: PMC11015617 DOI: 10.1186/s13195-024-01455-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 04/08/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Measurement of beta-amyloid (Aβ) and phosphorylated tau (p-tau) levels offers the potential for early detection of neurocognitive impairment. Still, the probability of developing a clinical syndrome in the presence of these protein changes (A+ and T+) remains unclear. By performing a systematic review and meta-analysis, we investigated the risk of mild cognitive impairment (MCI) or dementia in the non-demented population with A+ and A- alone and in combination with T+ and T- as confirmed by PET or cerebrospinal fluid examination. METHODS A systematic search of prospective and retrospective studies investigating the association of Aβ and p-tau with cognitive decline was performed in three databases (MEDLINE via PubMed, EMBASE, and CENTRAL) on January 9, 2024. The risk of bias was assessed using the Cochrane QUIPS tool. Odds ratios (OR) and Hazard Ratios (HR) were pooled using a random-effects model. The effect of neurodegeneration was not studied due to its non-specific nature. RESULTS A total of 18,162 records were found, and at the end of the selection process, data from 36 cohorts were pooled (n= 7,793). Compared to the unexposed group, the odds ratio (OR) for conversion to dementia in A+ MCI patients was 5.18 [95% CI 3.93; 6.81]. In A+ CU subjects, the OR for conversion to MCI or dementia was 5.79 [95% CI 2.88; 11.64]. Cerebrospinal fluid Aβ42 or Aβ42/40 analysis and amyloid PET imaging showed consistent results. The OR for conversion in A+T+ MCI subjects (11.60 [95% CI 7.96; 16.91]) was significantly higher than in A+T- subjects (2.73 [95% CI 1.65; 4.52]). The OR for A-T+ MCI subjects was non-significant (1.47 [95% CI 0.55; 3.92]). CU subjects with A+T+ status had a significantly higher OR for conversion (13.46 [95% CI 3.69; 49.11]) than A+T- subjects (2.04 [95% CI 0.70; 5.97]). Meta-regression showed that the ORs for Aβ exposure decreased with age in MCI. (beta = -0.04 [95% CI -0.03 to -0.083]). CONCLUSIONS Identifying Aβ-positive individuals, irrespective of the measurement technique employed (CSF or PET), enables the detection of the most at-risk population before disease onset, or at least at a mild stage. The inclusion of tau status in addition to Aβ, especially in A+T+ cases, further refines the risk assessment. Notably, the higher odds ratio associated with Aβ decreases with age. TRIAL REGISTRATION The study was registered in PROSPERO (ID: CRD42021288100).
Collapse
Affiliation(s)
- Zsolt Huszár
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6, Budapest, 1083, Hungary
| | - Marie Anne Engh
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Márk Pavlekovics
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- Department of Neurology, Jahn Ferenc Teaching Hospital, Köves utca 1, Budapest, 1204, Hungary
| | - Tomoya Sato
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Yalea Steenkamp
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Bernard Hanseeuw
- Department of Neurology and Institute of Neuroscience, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, 1200, Belgium
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02155, USA
| | - Tamás Terebessy
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Zsolt Molnár
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Üllői út 78/A, Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Poznan University of Medical Sciences, 49 Przybyszewskiego St, Poznan, Poland
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, 7624, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Tömő 25-29, Budapest, 1083, Hungary
- Translational Pancreatology Research Group, Interdisciplinary Centre of Excellence for Research Development and Innovation University of Szeged, Budapesti 9, Szeged, 6728, Hungary
| | - Gábor Csukly
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary.
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6, Budapest, 1083, Hungary.
| |
Collapse
|
26
|
Wen Q, Wittens MMJ, Engelborghs S, van Herwijnen MHM, Tsamou M, Roggen E, Smeets B, Krauskopf J, Briedé JJ. Beyond CSF and Neuroimaging Assessment: Evaluating Plasma miR-145-5p as a Potential Biomarker for Mild Cognitive Impairment and Alzheimer's Disease. ACS Chem Neurosci 2024; 15:1042-1054. [PMID: 38407050 PMCID: PMC10921410 DOI: 10.1021/acschemneuro.3c00740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/27/2024] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. New strategies for the early detection of MCI and sporadic AD are crucial for developing effective treatment options. Current techniques used for diagnosis of AD are invasive and/or expensive, so they are not suitable for population screening. Cerebrospinal fluid (CSF) biomarkers such as amyloid β1-42 (Aβ1-42), total tau (T-tau), and phosphorylated tau181 (P-tau181) levels are core biomarkers for early diagnosis of AD. Several studies have proposed the use of blood-circulating microRNAs (miRNAs) as potential novel early biomarkers for AD. We therefore applied a novel approach to identify blood-circulating miRNAs associated with CSF biomarkers and explored the potential of these miRNAs as biomarkers of AD. In total, 112 subjects consisting of 28 dementia due to AD cases, 63 MCI due to AD cases, and 21 cognitively healthy controls were included. We identified seven Aβ1-42-associated plasma miRNAs, six P-tau181-associated plasma miRNAs, and nine Aβ1-42-associated serum miRNAs. These miRNAs were involved in AD-relevant biological processes, such as PI3K/AKT signaling. Based on this signaling pathway, we constructed an miRNA-gene target network, wherein miR-145-5p has been identified as a hub. Furthermore, we showed that miR-145-5p performs best in the prediction of both AD and MCI. Moreover, miR-145-5p also improved the prediction performance of the mini-mental state examination (MMSE) score. The performance of this miRNA was validated using different datasets including an RT-qPCR dataset from plasma samples of 23 MCI cases and 30 age-matched controls. These findings indicate that blood-circulating miRNAs that are associated with CSF biomarkers levels and specifically plasma miR-145-5p alone or combined with the MMSE score can potentially be used as noninvasive biomarkers for AD or MCI screening in the general population, although studies in other AD cohorts are necessary for further validation.
Collapse
Affiliation(s)
- Qingfeng Wen
- Department
of Toxicogenomics, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- MHeNS,
School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Mandy Melissa Jane Wittens
- Department
of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Universiteitsplein 1, BE-2610 Antwerpen, Belgium
- Neuroprotection
and Neuromodulation (NEUR), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
- Department
of Neurology, Universitair Ziekenhuis Brussel
(UZ Brussel), Laarbeeklaan
101, 1090 Brussel, Belgium
| | - Sebastiaan Engelborghs
- Department
of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Universiteitsplein 1, BE-2610 Antwerpen, Belgium
- Neuroprotection
and Neuromodulation (NEUR), Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussel, Belgium
- Department
of Neurology, Universitair Ziekenhuis Brussel
(UZ Brussel), Laarbeeklaan
101, 1090 Brussel, Belgium
| | - Marcel H. M. van Herwijnen
- Department
of Toxicogenomics, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Maria Tsamou
- ToxGenSolutions
(TGS), 6229EV Maastricht, The Netherlands
| | - Erwin Roggen
- ToxGenSolutions
(TGS), 6229EV Maastricht, The Netherlands
| | - Bert Smeets
- Department
of Toxicogenomics, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- MHeNS,
School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Julian Krauskopf
- Department
of Toxicogenomics, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Jacco Jan Briedé
- Department
of Toxicogenomics, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- MHeNS,
School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
27
|
Kamondi A, Grigg-Damberger M, Löscher W, Tanila H, Horvath AA. Epilepsy and epileptiform activity in late-onset Alzheimer disease: clinical and pathophysiological advances, gaps and conundrums. Nat Rev Neurol 2024; 20:162-182. [PMID: 38356056 DOI: 10.1038/s41582-024-00932-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/16/2024]
Abstract
A growing body of evidence has demonstrated a link between Alzheimer disease (AD) and epilepsy. Late-onset epilepsy and epileptiform activity can precede cognitive deterioration in AD by years, and its presence has been shown to predict a faster disease course. In animal models of AD, amyloid and tau pathology are linked to cortical network hyperexcitability that precedes the first signs of memory decline. Thus, detection of epileptiform activity in AD has substantial clinical importance as a potential novel modifiable risk factor for dementia. In this Review, we summarize the epidemiological evidence for the complex bidirectional relationship between AD and epilepsy, examine the effect of epileptiform activity and seizures on cognition in people with AD, and discuss the precision medicine treatment strategies based on the latest research in human and animal models. Finally, we outline some of the unresolved questions of the field that should be addressed by rigorous research, including whether particular clinicopathological subtypes of AD have a stronger association with epilepsy, and the sequence of events between epileptiform activity and amyloid and tau pathology.
Collapse
Affiliation(s)
- Anita Kamondi
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary.
- Department of Neurology, Semmelweis University, Budapest, Hungary.
| | | | - Wolfgang Löscher
- Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany
| | - Heikki Tanila
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Andras Attila Horvath
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
28
|
Jia J, Ning Y, Chen M, Wang S, Yang H, Li F, Ding J, Li Y, Zhao B, Lyu J, Yang S, Yan X, Wang Y, Qin W, Wang Q, Li Y, Zhang J, Liang F, Liao Z, Wang S. Biomarker Changes during 20 Years Preceding Alzheimer's Disease. N Engl J Med 2024; 390:712-722. [PMID: 38381674 DOI: 10.1056/nejmoa2310168] [Citation(s) in RCA: 120] [Impact Index Per Article: 120.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
BACKGROUND Biomarker changes that occur in the period between normal cognition and the diagnosis of sporadic Alzheimer's disease have not been extensively investigated in longitudinal studies. METHODS We conducted a multicenter, nested case-control study of Alzheimer's disease biomarkers in cognitively normal participants who were enrolled in the China Cognition and Aging Study from January 2000 through December 2020. A subgroup of these participants underwent testing of cerebrospinal fluid (CSF), cognitive assessments, and brain imaging at 2-year-to-3-year intervals. A total of 648 participants in whom Alzheimer's disease developed were matched with 648 participants who had normal cognition, and the temporal trajectories of CSF biochemical marker concentrations, cognitive testing, and imaging were analyzed in the two groups. RESULTS The median follow-up was 19.9 years (interquartile range, 19.5 to 20.2). CSF and imaging biomarkers in the Alzheimer's disease group diverged from those in the cognitively normal group at the following estimated number of years before diagnosis: amyloid-beta (Aβ)42, 18 years; the ratio of Aβ42 to Aβ40, 14 years; phosphorylated tau 181, 11 years; total tau, 10 years; neurofilament light chain, 9 years; hippocampal volume, 8 years; and cognitive decline, 6 years. As cognitive impairment progressed, the changes in CSF biomarker levels in the Alzheimer's disease group initially accelerated and then slowed. CONCLUSIONS In this study involving Chinese participants during the 20 years preceding clinical diagnosis of sporadic Alzheimer's disease, we observed the time courses of CSF biomarkers, the times before diagnosis at which they diverged from the biomarkers from a matched group of participants who remained cognitively normal, and the temporal order in which the biomarkers became abnormal. (Funded by the Key Project of the National Natural Science Foundation of China and others; ClinicalTrials.gov number, NCT03653156.).
Collapse
Affiliation(s)
- Jianping Jia
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Yuye Ning
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Meilin Chen
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Shuheng Wang
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Hao Yang
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Fangyu Li
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Jiayi Ding
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Yan Li
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Bote Zhao
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Jihui Lyu
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Shanshan Yang
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Xin Yan
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Yue Wang
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Wei Qin
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Qi Wang
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Ying Li
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Jintao Zhang
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Furu Liang
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Zhengluan Liao
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| | - Shan Wang
- From the Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital (J.J., Y.N., M.C., Shuheng Wang, H.Y., F. Li, J.D., Yan Li, B.Z., W.Q., Q.W., Ying Li), Beijing Key Laboratory of Geriatric Cognitive Disorders, Clinical Center for Neurodegenerative Disease and Memory Impairment (J.J.), the Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders (J.J.), and the Department of Neurology, Beijing Anding Hospital (Y.W.), Capital Medical University, Key Laboratory of Neurodegenerative Diseases, Ministry of Education (J.J.), the Center for Cognitive Disorders, Beijing Geriatric Hospital (J.L.), and the Department of Neurology, Beijing Jishuitan Hospital (X.Y.), Beijing, the Department of Neurology, Daqing Oilfield General Hospital, Daqing (S.Y.), the Department of Neurology, the 960th Hospital of the People's Liberation Army, Jinan (J.Z.), the Department of Neurology, Baotou Central Hospital, Baotou (F. Liang), the Department of Psychiatry, Zhejiang Provincial People's Hospital, Hangzhou (Z.L.), and the Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang (Shan Wang) - all in China
| |
Collapse
|
29
|
Li QY, Hu HY, Zhang GW, Hu H, Ou YN, Huang LY, Wang AY, Gao PY, Ma LY, Tan L, Yu JT. Associations between cardiometabolic multimorbidity and cerebrospinal fluid biomarkers of Alzheimer's disease pathology in cognitively intact adults: the CABLE study. Alzheimers Res Ther 2024; 16:28. [PMID: 38321520 PMCID: PMC10848421 DOI: 10.1186/s13195-024-01396-w] [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: 08/08/2023] [Accepted: 01/21/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Cardiometabolic multimorbidity is associated with an increased risk of dementia, but the pathogenic mechanisms linking them remain largely undefined. We aimed to assess the associations of cardiometabolic multimorbidity with cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) pathology to enhance our understanding of the underlying mechanisms linking cardiometabolic multimorbidity and AD. METHODS This study included 1464 cognitively intact participants from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) database. Cardiometabolic diseases (CMD) are a group of interrelated disorders such as hypertension, diabetes, heart diseases (HD), and stroke. Based on the CMD status, participants were categorized as CMD-free, single CMD, or CMD multimorbidity. CMD multimorbidity is defined as the coexistence of two or more CMDs. The associations of cardiometabolic multimorbidity and CSF biomarkers were examined using multivariable linear regression models with demographic characteristics, the APOE ε4 allele, and lifestyle factors as covariates. Subgroup analyses stratified by age, sex, and APOE ε4 status were also performed. RESULTS A total of 1464 individuals (mean age, 61.80 years; age range, 40-89 years) were included. The markers of phosphorylated tau-related processes (CSF P-tau181: β = 0.165, P = 0.037) and neuronal injury (CSF T-tau: β = 0.065, P = 0.033) were significantly increased in subjects with CMD multimorbidity (versus CMD-free), but not in those with single CMD. The association between CMD multimorbidity with CSF T-tau levels remained significant after controlling for Aβ42 levels. Additionally, significantly elevated tau-related biomarkers were observed in patients with specific CMD combinations (i.e., hypertension and diabetes, hypertension and HD), especially in long disease courses. CONCLUSIONS The presence of cardiometabolic multimorbidity was associated with tau phosphorylation and neuronal injury in cognitively normal populations. CMD multimorbidity might be a potential independent target to alleviate tau-related pathologies that can cause cognitive impairment.
Collapse
Affiliation(s)
- Qiong-Yao Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Gao-Wen Zhang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - An-Yi Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Li-Yun Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, No. 12 Wulumuqi Road, Shanghai, China.
| |
Collapse
|
30
|
Li JQ, Song JH, Suckling J, Wang YJ, Zuo CT, Zhang C, Gao J, Song YQ, Xie AM, Tan L, Yu JT. Disease trajectories in older adults with non-AD pathologic change and comparison with Alzheimer's disease pathophysiology: A longitudinal study. Neurobiol Aging 2024; 134:106-114. [PMID: 38056216 DOI: 10.1016/j.neurobiolaging.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
Based on the 'AT(N)' system, individuals with normal amyloid biomarkers but abnormal tauopathy or neurodegeneration biomarkers are classified as non-Alzheimer's disease (AD) pathologic change. This study aimed to assess the long-term clinical and cognitive trajectories of individuals with non-AD pathologic change among older adults without dementia, comparing them to those with normal AD biomarkers and AD pathophysiology. Analyzing Alzheimer's Disease Neuroimaging Initiative data, we evaluated clinical outcomes and conversion risk longitudinally using mixed effects models and multivariate Cox proportional hazard models. We found that compared to individuals with A-T-N-, those with abnormal tauopathy or neurodegeneration biomarkers (A-T + N-, A-T-N + , and A-T + N + ) had a faster rate of cognitive decline and disease progression. Individuals with A-T + N + had a faster rate of decline than those with A-T + N-. Additionally, in individuals with the same baseline tauopathy and neurodegeneration biomarker status, the presence of baseline amyloid could accelerate cognitive decline and clinical progression. These findings provide a foundation for future studies on non-AD pathologic change and its comparison with AD pathophysiology.
Collapse
Affiliation(s)
- Jie-Qiong Li
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China.
| | - Jing-Hui Song
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - John Suckling
- Department of Psychiatry, University of Cambridge, Cambridge CB2 1TN, UK; Medical Research Council and Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge CB2 1TN, UK; Cambridgeshire and Peterborough NHS Trust, UK
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Chuan-Tao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai 200433, China
| | - Can Zhang
- Genetics and Aging Research Unit, Mass GeneralInstitute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown 02138, MA 02129-2060, USA
| | - Jing Gao
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Yu-Qiang Song
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - An-Mu Xie
- Department of Neurology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital,Qingdao University, Qingdao 266000, Shandong, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for NeurologicalDisorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200040, China.
| |
Collapse
|
31
|
Palade J, Alsop E, Courtright-Lim A, Hsieh M, Whitsett TG, Galasko D, Van Keuren-Jensen K. Small RNA Changes in Plasma Have Potential for Early Diagnosis of Alzheimer's Disease before Symptom Onset. Cells 2024; 13:207. [PMID: 38334599 PMCID: PMC10854972 DOI: 10.3390/cells13030207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
Alzheimer's disease (AD), due to its multifactorial nature and complex etiology, poses challenges for research, diagnosis, and treatment, and impacts millions worldwide. To address the need for minimally invasive, repeatable measures that aid in AD diagnosis and progression monitoring, studies leveraging RNAs associated with extracellular vesicles (EVs) in human biofluids have revealed AD-associated changes. However, the validation of AD biomarkers has suffered from the collection of samples from differing points in the disease time course or a lack of confirmed AD diagnoses. Here, we integrate clinical diagnosis and postmortem pathology data to form more accurate experimental groups and use small RNA sequencing to show that EVs from plasma can serve as a potential source of RNAs that reflect disease-related changes. Importantly, we demonstrated that these changes are identifiable in the EVs of preclinical patients, years before symptom manifestation, and that machine learning models based on differentially expressed RNAs can help predict disease conversion or progression. This research offers critical insight into early disease biomarkers and underscores the significance of accounting for disease progression and pathology in human AD studies.
Collapse
Affiliation(s)
- Joanna Palade
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (J.P.); (E.A.); (M.H.); (T.G.W.)
| | - Eric Alsop
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (J.P.); (E.A.); (M.H.); (T.G.W.)
| | | | - Michael Hsieh
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (J.P.); (E.A.); (M.H.); (T.G.W.)
| | - Timothy G. Whitsett
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (J.P.); (E.A.); (M.H.); (T.G.W.)
| | - Douglas Galasko
- Department of Neurosciences, San Diego and Shiley-Marcos Alzheimer’s Disease Research Center, University of California, La Jolla, CA 92037, USA;
| | - Kendall Van Keuren-Jensen
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (J.P.); (E.A.); (M.H.); (T.G.W.)
| |
Collapse
|
32
|
Corradetti G, Oncel D, Kadomoto S, Arakaki X, Kloner RA, Sadun AA, Sadda SR, Chan JW. Choriocapillaris and Retinal Vascular Alterations in Presymptomatic Alzheimer's Disease. Invest Ophthalmol Vis Sci 2024; 65:47. [PMID: 38294804 PMCID: PMC10839815 DOI: 10.1167/iovs.65.1.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
Purpose To compare optical coherence tomography angiography (OCTA) retina metrics between cognitively healthy subjects with pathological versus normal cerebrospinal fluid (CSF) Aβ42/tau ratios. Methods Swept-source OCTA scans were collected using the Zeiss PLEX Elite 9000 and analyzed on 23 cognitively healthy (CH) subjects who had previously undergone CSF analysis. Thirteen subjects had a pathological Aβ42/tau (PAT) ratio of <2.7132, indicative of presymptomatic Alzheimer's disease (AD), and 10 had a normal Aβ42/tau (NAT) ratio of ≥2.7132. OCTA en face images of the superficial vascular complex (SVC) and deep vascular complex were binarized and skeletonized to quantify the perfusion density (PD), vessel length density (VLD), and fractal dimension (FrD). The foveal avascular zone (FAZ) area was calculated using the SVC slab. Choriocapillaris flow deficits (CCFDs) were computed from the en face OCTA slab of the CC. The above parameters were compared between CH-PATs and CH-NATs. Results Compared to CH-NATs, CH-PATs showed significantly decreased PD, VLD, and FrD in the SVC, with a significantly increased FAZ area and CCFDs. Conclusions Swept-source OCTA analysis of the SVC and CC suggests a significant vascular loss at the CH stage of pre-AD that might be an indicator of a neurodegenerative process initiated by the impaired clearance of Aβ42 in the blood vessel wall and by phosphorylated tau accumulation in the perivascular spaces, a process that most likely mirrors that in the brain. If confirmed in larger longitudinal studies, OCTA retinal and inner choroidal metrics may be important biomarkers for assessing presymptomatic AD.
Collapse
Affiliation(s)
- Giulia Corradetti
- Doheny Eye Institute, Pasadena, California, United States
- Department of Ophthalmology David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States
| | - Deniz Oncel
- Doheny Eye Institute, Pasadena, California, United States
| | - Shin Kadomoto
- Doheny Eye Institute, Pasadena, California, United States
| | - Xianghong Arakaki
- Cognition and Brain Integration Laboratory, Department of Neurosciences, Huntington Medical Research Institutes, Pasadena, California, United States
| | - Robert A. Kloner
- Clinical Neuroscience, Department of Neurosciences, Huntington Medical Research Institutes, Pasadena, California, United States
- Cardiovascular Research Institute, Huntington Medical Research Institutes, Pasadena, California, United States
- Cardiovascular Division, Department of Medicine Keck School of Medicine of University of Southern California, Los Angeles, California, United States
| | - Alfredo A. Sadun
- Doheny Eye Institute, Pasadena, California, United States
- Department of Ophthalmology David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States
| | - SriniVas R. Sadda
- Doheny Eye Institute, Pasadena, California, United States
- Department of Ophthalmology David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States
| | - Jane W. Chan
- Doheny Eye Institute, Pasadena, California, United States
- Department of Ophthalmology David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States
| |
Collapse
|
33
|
Cardoso S, Guerreiro M, Montalvo A, Silva D, Alves L, de Mendonça A. Amyloid-Negative, Neurodegeneration-Negative Amnestic Mild Cognitive Impairment. J Alzheimers Dis 2024; 101:369-377. [PMID: 39177603 DOI: 10.3233/jad-240621] [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: 08/24/2024]
Abstract
Background The concept of amnestic mild cognitive impairment (aMCI) was developed to identify patients at an initial stage of Alzheimer's disease (AD). However, some patients with aMCI do not present biomarkers of amyloid pathology or neuronal injury. Objective To know the natural history of amyloid-negative and neurodegeneration-negative patients with aMCI, namely to ascertain: 1) whether these patients remain cognitively stable or they present a slow decline in neuropsychological tests; 2) whether the memory complaints subside with the apparently benign clinical course of the disorder or if they persist along the time. Methods Patients who fulfilled criteria for aMCI with no biomarkers of amyloid pathology or neuronal injury were selected from a large cohort of non-demented patients with cognitive complaints, and were followed with clinical and neuropsychological assessments. Results Twenty-one amyloid-negative and neurodegeneration-negative aMCI patients were followed for 7.1±3.7 years. At the baseline they had more pronounced deficits in verbal learning (California Verbal Learning Test) and were also impaired in Word Recall and Logical Memory. However, they did not decline in any cognitive test during follow-up. The patients maintained a high level of subjective memory complaints from baseline (9.7±4.1) to the follow-up visit (9.2±4.1, a non-significant difference), in spite of a statistically significant decrease in the depressive symptoms, with Geriatric Depression Scale (15 items) score 4.9±2.8 at baseline and 3.2±1.8 at the follow-up visit. Conclusions Amyloid-negative, neurodegeneration-negative aMCI is a chronic clinical condition characterized by the long-term persistence of cognitive deficits and distressing memory complaints. Adequate strategies to treat this condition are needed.
Collapse
Affiliation(s)
- Sandra Cardoso
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | | | | | - Dina Silva
- Department of Psychology and Educational Sciences and Centre for Biomedical Research (CBMR), Cognitive Neuroscience Research Group, Universidade do Algarve, Faro, Portugal
| | - Luísa Alves
- Chronic Diseases Research Centre, NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal
| | | |
Collapse
|
34
|
Aschenbrenner AJ, Hassenstab JJ, Schindler SE, Janelidze S, Hansson O, Morris JC, Grober E. Free Recall Outperforms Story Recall in Associations with Plasma Biomarkers in Preclinical Alzheimer Disease. J Prev Alzheimers Dis 2024; 11:1696-1702. [PMID: 39559880 PMCID: PMC11573877 DOI: 10.14283/jpad.2024.130] [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: 11/20/2024]
Abstract
BACKGROUND A decline in episodic memory is one of the earliest cognitive characteristics of Alzheimer disease and memory tests are heavily featured in cognitive composite endpoints that are used to demonstrate treatment efficacy. Assessments of episodic memory can take many forms including free recall, associate learning, and paragraph or story recall. Plasma biomarkers of Alzheimer disease are now widely available and will likely form the backbone of cohort enrichment strategies for future clinical trials. Thus, it is critical to evaluate which episodic memory measures are most sensitive to plasma markers of Alzheimer disease pathology. OBJECTIVES To compare the associations of common episodic memory tests with plasma biomarkers of Alzheimer disease. DESIGN Longitudinal cohort study. SETTING Academic medical center in the midwestern United States. PARTICIPANTS A total of 161 cognitively normal older adults with at least one plasma biomarker assessment and two or more annual clinical and cognitive assessments which included up to three different tests of episodic memory. MEASUREMENTS Episodic memory performance using free recall, paired associates recall or paragraph recall. Plasma Aβ42, Aβ40, ptau217, and neurofilament light chain were measured. RESULTS Free recall on the Free and Cued Selective Reminding Test with Immediate Recall (FCSRT + IR) was substantially more sensitive to longitudinal cognitive change associated with abnormal baseline plasma Aβ42/Aβ40 and ptau217 compared to other measures of episodic memory. A cognitive composite that included only free recall showed larger decline associated with baseline Aβ42/Aβ40 when compared to those that included paragraph recall. Differences in decline across composites were minimal when considering baseline ptau217 or NfL. CONCLUSION Episodic memory is a critical domain to assess in preclinical Alzheimer disease. Methods of assessing memory are not equal and longitudinal change in free recall substantially outperformed both paired associates and paragraph recall. Clinical trial results will depend critically on the episodic memory test(s) that are chosen for a composite endpoint and free recall from the FCSRT + IR is an optimal memory measure to include rather than paired associates or paragraph recall.
Collapse
Affiliation(s)
- A J Aschenbrenner
- Andrew Aschenbrenner, PhD, 4488 Forest Park Ave, STE 301, St. Louis, MO, 63108, , 314-273-1041
| | | | | | | | | | | | | |
Collapse
|
35
|
Na S, Lee C, Ho S, Hong YJ, Jeong JH, Park KH, Kim S, Wang MJ, Choi SH, Han S, Kang SW, Kang S, Yang DW. A Longitudinal Study on Memory Enhancement in Subjective Cognitive Decline Patients: Clinical and Neuroimaging Perspectives. J Alzheimers Dis 2024; 97:193-204. [PMID: 38108349 DOI: 10.3233/jad-230667] [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: 12/19/2023]
Abstract
BACKGROUND Subjective cognitive decline (SCD) refers to the self-reported persistent cognitive decline despite normal objective testing, increasing the risk of dementia compared to cognitively normal individuals. OBJECTIVE This study aims to investigate the attributes of SCD patients who demonstrated memory function improvement. METHODS In this prospective study of SCD, a total of 120 subjects were enrolled as part of a multicenter cohort study aimed at identifying predictors for the clinical progression to mild cognitive impairment or dementia (CoSCo study). All subjects underwent 18F-florbetaben PET and brain MRI scans at baseline and annual neuropsychological tests. At the 24-month follow-up, we classified SCD patients based on changes in memory function, the z-score of the Seoul verbal learning test delayed recall. RESULTS Of the 120 enrolled patients, 107 successfully completed the 24-month follow-up assessment. Among these, 80 patients (74.8%) with SCD exhibited memory function improvements. SCD patients with improved memory function had a lower prevalence of coronary artery disease at baseline and performed better in the trail-making test part B compared to those without improvement. Anatomical and biomarker analysis showed a lower frequency of amyloid PET positivity and larger volumes in the left and right superior parietal lobes in subjects with improved memory function. CONCLUSIONS Our prospective study indicates that SCD patients experiencing memory improvement over a 24-month period had a lower amyloid burden, fewer cardiovascular risk factors, and superior executive cognitive function. Identifying these key factors associated with cognitive improvement may assist clinicians in predicting future memory function improvements in SCD patients.
Collapse
Affiliation(s)
- Seunghee Na
- Department of Neurology, College of Medicine, The Catholic University of Korea, Incheon St. Mary's Hospital, Incheon, South Korea
| | - Chonghwee Lee
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, South Korea
| | - SeongHee Ho
- Department of Neurology, Hanyang University Hanmaeum Changwon Hospital, Changwon, Korea
| | - Yun Jeong Hong
- Department of Neurology, College of Medicine, The Catholic University of Korea, Uijeongbu St. Mary's Hospital, Uijeongbu, South Korea
| | - Jee Hyang Jeong
- Department of Neurology, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, Seoul, South Korea
| | - Kee Hyung Park
- Department of Neurology, Gachon University Gil Hospital, Incheon, South Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | | | - Seong Hye Choi
- Department of Neurology, Inha University School of Medicine, Incheon, South Korea
| | | | - Seung Wan Kang
- Data Center for Korean EEG, College of Nursing, Seoul National University, Seoul, South Korea
- iMediSync Inc. Seoul, South Korea
| | - Sungmin Kang
- Research and Development, PeopleBio Inc., Seongnam-si, Gyeonggi-do, South Korea
| | - Dong Won Yang
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, South Korea
| |
Collapse
|
36
|
Souchet B, Michaïl A, Billoir B, Braudeau J. Biological Diagnosis of Alzheimer's Disease Based on Amyloid Status: An Illustration of Confirmation Bias in Medical Research? Int J Mol Sci 2023; 24:17544. [PMID: 38139372 PMCID: PMC10744068 DOI: 10.3390/ijms242417544] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Alzheimer's disease (AD) was first characterized by Dr. Alois Alzheimer in 1906 by studying a demented patient and discovering cerebral amyloid plaques and neurofibrillary tangles. Subsequent research highlighted the roles of Aβ peptides and tau proteins, which are the primary constituents of these lesions, which led to the amyloid cascade hypothesis. Technological advances, such as PET scans using Florbetapir, have made it possible to visualize amyloid plaques in living patients, thus improving AD's risk assessment. The National Institute on Aging and the Alzheimer's Association introduced biological diagnostic criteria in 2011, which underlined the amyloid deposits diagnostic value. However, potential confirmation bias may have led researchers to over-rely on amyloid markers independent of AD's symptoms, despite evidence of their limited specificity. This review provides a critical examination of the current research paradigm in AD, including, in particular, the predominant focus on amyloid and tau species in diagnostics. We discuss the potential multifaceted consequences of this approach and propose strategies to mitigate its overemphasis in the development of new biomarkers. Furthermore, our study presents comprehensive guidelines aimed at enhancing the creation of biomarkers for accurately predicting AD dementia onset. These innovations are crucial for refining patient selection processes in clinical trial enrollment and for the optimization of therapeutic strategies. Overcoming confirmation bias is essential to advance the diagnosis and treatment of AD and to move towards precision medicine by incorporating a more nuanced understanding of amyloid biomarkers.
Collapse
Affiliation(s)
| | | | | | - Jérôme Braudeau
- AgenT SAS, 4 Rue Pierre Fontaine, 91000 Evry-Courcouronnes, France; (B.S.); (A.M.); (B.B.)
| |
Collapse
|
37
|
Chino B, Torres-Simón L, Żelwetro A, Rodríguez-Rojo IC, Carnes-Vendrell A, Piñol-Ripoll G, Yubero R, Paúl N, Maestú F. Understanding the Episodic Memory and Executive Functioning Axis Impairment in MCI Patients: A Multicenter Study in Comparison with CSF Biomarkers. Biomedicines 2023; 11:3147. [PMID: 38137368 PMCID: PMC10741228 DOI: 10.3390/biomedicines11123147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/06/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND This study aimed to explore the association between a verbal learning task that evaluates the potential mutual dependency between memory and executive functions (i.e., the Test of Memory Strategies, TMS) and cerebrospinal fluid (CSF) Alzheimer's Disease (AD) biomarkers. METHODS A sample of 47 mild cognitive impairment (MCI) participants from Poland and Spain were classified according to the Erlangen Score Diagnostic Algorithm (ESA) into CSF- (n = 16) and CSF+ (n = 31) groups. Correlation analyses between TMS word-list conditions and CSF biomarkers were conducted. Additionally, an analysis of covariance was performed to define the effect on ESA classification in the sample, using as a covariable the country of origin of the participants. RESULTS Significant associations between the TMS-3 condition and Aβ42, t-tau, and p-tau were observed for the whole sample. In addition, the CSF- participants obtained higher cognitive performance in TMS-3 compared to the CSF+ group. This outcome persisted if the groups were based on Aβ42 scores, but not t-tau or p-tau values. CONCLUSIONS These findings could indicate that poor performance on verbal learning tests may be affected by executive dysfunctions. Therefore, future intervention plans focused on training executive functions would be of interest to improve the ability of MCI patients to encode and organize information.
Collapse
Affiliation(s)
- Brenda Chino
- Institute of Neuroscience, Autonomous University of Barcelona (UAB), 08193 Barcelona, Spain;
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.T.-S.); (F.M.)
| | - Lucía Torres-Simón
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.T.-S.); (F.M.)
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Agnieszka Żelwetro
- Interdisciplinary Doctoral School, SWPS University of Social Sciences and Humanities, 53-238 Wrocław, Poland;
- Alzheimer’s Disease Research, Center in Ścinawa, 59-330 Ścinawa, Poland
| | - Inmaculada Concepción Rodríguez-Rojo
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.T.-S.); (F.M.)
- Department of Nursing and Physiotherapy, Faculty of Medicine and Health Sciences, Universidad de Alcalá, 28801 Madrid, Spain
| | - Anna Carnes-Vendrell
- Unitat de Trastorns Cognitius, Cognition and Behavior Study Group, Universitat de Lleida, IRBLleida, 25198 Lleida, Spain; (A.C.-V.); (G.P.-R.)
| | - Gerard Piñol-Ripoll
- Unitat de Trastorns Cognitius, Cognition and Behavior Study Group, Universitat de Lleida, IRBLleida, 25198 Lleida, Spain; (A.C.-V.); (G.P.-R.)
| | - Raquel Yubero
- Neurology Department, Hospital Quirónsalud Madrid, 28223 Madrid, Spain;
| | - Nuria Paúl
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Fernando Maestú
- Center for Cognitive and Computational Neuroscience, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.T.-S.); (F.M.)
- Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense de Madrid, 28040 Madrid, Spain;
- Instituto de Investigación del Hospital Clínico San Carlos, 28040 Madrid, Spain
| |
Collapse
|
38
|
Guo HF, Wu Y, Li J, Pan FF. Analysis of the relationship between blood pressure variability and subtle cognitive decline in older adults. World J Psychiatry 2023; 13:872-883. [DOI: 10.5498/wjp.v13.i11.872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/18/2023] [Accepted: 10/23/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Blood pressure variability (BPV) has been shown to be related to mild cognitive impairment and Alzheimer's disease in a number of studies. However, the relationship between BPV and subtle cognitive decline (SCD) has received minimal attention in this field of research to date and has rarely been reported.
AIM To examine whether SCD is independently associated with changes in BPV in older adults.
METHODS Participants were selected based on having participated in cognitive function evaluation and ambulatory blood pressure measurement at the Shanghai Sixth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine between June 2020 and August 2022. The participants included 182 individuals with SCD as the experimental group and 237 with normal cognitive function as the control group. The basic data, laboratory examinations, scale tests, and ambulatory blood pressure test results of the two groups were analyzed retrospectively, and the relationship between SCD and BPV was subsequently evaluated.
RESULTS Significant differences were observed between the two groups of participants (P < 0.05) in terms of age, education level, prevalence rate of diabetes, fasting blood glucose level, 24-h systolic blood pressure standard deviation and coefficient of variation, 24-h diastolic blood pressure standard deviation and coefficient of variation. The scale monitoring results showed significant differences in the scores for memory, attention, and visual space between the experimental and control groups. Logistic regression analysis indicated that age, education level, blood sugar level, and BPV were factors influencing cognitive decline. Linear regression analysis showed that there was an independent correlation between blood pressure variation and SCD, even after adjusting for related factors. Each of the above differences was still significant.
CONCLUSION This study suggests that increased BPV is associated with SCD.
Collapse
Affiliation(s)
- Hui-Feng Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yi Wu
- Prenatal Diagnosis Center, International Peace Maternity & Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jie Li
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Feng-Feng Pan
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
39
|
Garcia S, Askew RL, Kavcic V, Shair S, Bhaumik AK, Rose E, Campbell S, May N, Hampstead BM, Dodge HH, Heidebrink JL, Paulson HL, Giordani B. Mild Cognitive Impairment Subtype Performance in Comparison to Healthy Older Controls on the NIH Toolbox and Cogstate. Alzheimer Dis Assoc Disord 2023; 37:328-334. [PMID: 37862614 PMCID: PMC10873007 DOI: 10.1097/wad.0000000000000587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 09/06/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Early detection is necessary for the treatment of dementia. Computerized testing has become more widely used in clinical trials; however, it is unclear how sensitive these measures are to early signs of neurodegeneration. We investigated the use of the NIH Toolbox-Cognition (NIHTB-CB) and Cogstate-Brief computerized neuropsychological batteries in the identification of mild cognitive impairment (MCI) versus healthy older adults [healthy control (HC)] and amnestic (aMCI) versus nonamnestic MCI (naMCI). Exploratory analyses include investigating potential racial differences. METHODS Two hundred six older adults were diagnosed as aMCI (n = 58), naMCI (n = 15), or cognitively healthy (HC; n = 133). RESULTS The NIH Toolbox-CB subtests of Flanker, Picture Sequence Memory, and Picture Vocabulary significantly differentiated MCI from HC. Further, subtests from both computerized batteries differentiated patients with aMCI from those with naMCI. Although the main effect of race differences was noted on tests and in diagnostic groups was significant, there were no significant race-by-test interactions. CONCLUSIONS Computer-based subtests vary in their ability to help distinguish MCI subtypes, though these tests provide less expensive and easier-to-administer clinical screeners to help identify patients early who may qualify for more comprehensive evaluations. Further work is needed, however, to refine computerized tests to achieve better precision in distinguishing impairment subtypes.
Collapse
Affiliation(s)
- Sarah Garcia
- Psychology Department, Stetson University, DeLand, FL, USA
| | | | | | - Sarah Shair
- Departments of Psychiatry, Neurology, Psychology, and School of Nursing, University of Michigan, Ann Arbor, MI, USA
- Michigan Alzheimer’s Disease Research Center, Ann Arbor, MI, USA
| | - Arijit K Bhaumik
- Departments of Psychiatry, Neurology, Psychology, and School of Nursing, University of Michigan, Ann Arbor, MI, USA
- Michigan Alzheimer’s Disease Research Center, Ann Arbor, MI, USA
| | - Edna Rose
- Departments of Psychiatry, Neurology, Psychology, and School of Nursing, University of Michigan, Ann Arbor, MI, USA
- Michigan Alzheimer’s Disease Research Center, Ann Arbor, MI, USA
| | - Stephen Campbell
- Departments of Psychiatry, Neurology, Psychology, and School of Nursing, University of Michigan, Ann Arbor, MI, USA
- Michigan Alzheimer’s Disease Research Center, Ann Arbor, MI, USA
| | - Nicolas May
- Michigan Alzheimer’s Disease Research Center, Ann Arbor, MI, USA
| | - Benjamin M. Hampstead
- Departments of Psychiatry, Neurology, Psychology, and School of Nursing, University of Michigan, Ann Arbor, MI, USA
- Michigan Alzheimer’s Disease Research Center, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Hiroko H. Dodge
- Layton Aging and Alzheimer’s Disease Center, Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Judith L Heidebrink
- Departments of Psychiatry, Neurology, Psychology, and School of Nursing, University of Michigan, Ann Arbor, MI, USA
- Michigan Alzheimer’s Disease Research Center, Ann Arbor, MI, USA
| | - Henry L Paulson
- Departments of Psychiatry, Neurology, Psychology, and School of Nursing, University of Michigan, Ann Arbor, MI, USA
- Michigan Alzheimer’s Disease Research Center, Ann Arbor, MI, USA
| | - Bruno Giordani
- Departments of Psychiatry, Neurology, Psychology, and School of Nursing, University of Michigan, Ann Arbor, MI, USA
- Michigan Alzheimer’s Disease Research Center, Ann Arbor, MI, USA
| |
Collapse
|
40
|
Soldan A, Oh S, Ryu T, Pettigrew C, Zhu Y, Moghekar A, Xiao MF, Pontone GM, Albert M, Na CH, Worley P. NPTX2 in Cerebrospinal Fluid Predicts the Progression From Normal Cognition to Mild Cognitive Impairment. Ann Neurol 2023; 94:620-631. [PMID: 37345460 PMCID: PMC10543570 DOI: 10.1002/ana.26725] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 06/23/2023]
Abstract
OBJECTIVE This study examined whether cerebrospinal fluid (CSF) baseline levels of the synaptic protein NPTX2 predict time to onset of symptoms of mild cognitive impairment (MCI), both alone and when accounting for traditional CSF Alzheimer's disease (AD) biomarker levels. Longitudinal NPTX2 levels were also examined. METHODS CSF was collected longitudinally from 269 cognitively normal BIOCARD Study participants (mean baseline age = 57.7 years; mean follow-up = 16.3 years; n = 77 progressed to MCI/dementia). NPTX2 levels were measured from 3 correlated peptides using quantitative parallel reaction monitoring mass spectrometry. Levels of Aβ42 /Aβ40 , p-tau181 , and t-tau were measured from the same CSF specimens using Lumipulse automated electrochemiluminescence assays. RESULTS In Cox regression models, lower baseline NPTX2 levels were associated with an earlier time to MCI symptom onset (hazard ratio [HR] = 0.76, SE = 0.09, p = 0.023). This association was significant for progression within 7 years (p = 0.036) and after 7 years from baseline (p = 0.001). Baseline NPTX2 levels improved prediction of time to MCI symptom onset after accounting for baseline AD biomarker levels (p < 0.01), and NPTX2 did not interact with the CSF AD biomarkers or APOE-ε4 genetic status. In linear mixed effects models, higher baseline p-tau181 and t-tau levels were associated with higher baseline levels of NPTX2 (both p < 0.001) and greater rates of NPTX2 declines over time. INTERPRETATION NPTX2 may be a valuable prognostic biomarker during preclinical AD that provides additive and independent prediction of MCI onset among individuals who are cognitively normal. We hypothesize that NPTX2-mediated circuit homeostasis confers resilience during the early phase of AD. ANN NEUROL 2023;94:620-631.
Collapse
Affiliation(s)
- Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sungtaek Oh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Taekyung Ryu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yuxin Zhu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Armstrong Institute for Patient Safety and Quality, Johns Hopkins Medicine, Baltimore, MD
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mei-Fang Xiao
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Gregory M. Pontone
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Chan-Hyun Na
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Paul Worley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
41
|
Deng Z, Xie D, Cai J, Jiang J, Pan D, Liao H, Liu X, Xu Y, Li H, Shen Q, Lattanzi S, Xiao S, Tang Y. Different types of milk consumption and the risk of dementia: Analysis from a large-scale cohort study. Clin Nutr 2023; 42:2058-2067. [PMID: 37677911 DOI: 10.1016/j.clnu.2023.08.019] [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: 04/02/2023] [Revised: 08/16/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND & AIMS Previous studies have investigated whether milk consumption has a role in preventing the development of cognitive impairment, but the results were inconsistent. Importantly, most of them have disregarded the role of different types of milk. This study aimed to examine the associations between different types of milk consumption and the risk of dementia. METHODS In this large-scale cohort study, participants without cognitive impairment at baseline were included from the UK Biobank. The type of milk mainly used was self-reported at baseline, including full-cream milk, skimmed-milk, soy milk, other milk, and no milk. The primary outcome was all-cause dementia. Secondary outcomes included Alzheimer's disease and vascular dementia. RESULTS Of the 307,271 participants included in the study (mean age 56.3 [SD 8.1] years), 3789 (1.2%) incident all-cause dementia cases were observed over a median follow-up of 12.3 years. After adjustment for potential confounders, only soy milk consumers had a statistically significantly lower risk of all-cause dementia compared with no milk consumers (hazard ratio [HR], 0.69; 95% confidence interval [CI], 0.54 to 0.90). When compared with soy milk non-consumers consisting of full-cream milk, skimmed-milk, and other milk consumers, soy milk consumers still showed a lower risk of all-cause dementia (HR, 0.76; 95% CI, 0.63 to 0.92), and there was no significant interaction with genetic risk for dementia (P for interaction = 0.15). Soy milk consumers showed a lower risk of Alzheimer's disease (HR, 0.70; 95% CI, 0.51 to 0.94; P = 0.02), while the association was not significant for vascular dementia (HR, 0.72; 95% CI, 0.47 to 1.12; P = 0.14). CONCLUSIONS The main consumption of soy milk was associated with a lower risk of dementia, particularly non-vascular dementia. Additional studies are needed to investigate how this association varies with the dose or frequency of the consumption of soy milk and to examine the generalizability of these findings in different populations.
Collapse
Affiliation(s)
- Zhenhong Deng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dongshu Xie
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jinhua Cai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jingru Jiang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dong Pan
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Huanquan Liao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xingyi Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Honghong Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qingyu Shen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Songhua Xiao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
42
|
Erickson P, Simrén J, Brum WS, Ennis GE, Kollmorgen G, Suridjan I, Langhough R, Jonaitis EM, Van Hulle CA, Betthauser TJ, Carlsson CM, Asthana S, Ashton NJ, Johnson SC, Shaw LM, Blennow K, Andreasson U, Bendlin BB, Zetterberg H. Prevalence and Clinical Implications of a β-Amyloid-Negative, Tau-Positive Cerebrospinal Fluid Biomarker Profile in Alzheimer Disease. JAMA Neurol 2023; 80:2807607. [PMID: 37523162 PMCID: PMC10391361 DOI: 10.1001/jamaneurol.2023.2338] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/05/2023] [Indexed: 08/01/2023]
Abstract
Importance Knowledge is lacking on the prevalence and prognosis of individuals with a β-amyloid-negative, tau-positive (A-T+) cerebrospinal fluid (CSF) biomarker profile. Objective To estimate the prevalence of a CSF A-T+ biomarker profile and investigate its clinical implications. Design, Setting, and Participants This was a retrospective cohort study of the cross-sectional multicenter University of Gothenburg (UGOT) cohort (November 2019-January 2021), the longitudinal multicenter Alzheimer Disease Neuroimaging Initiative (ADNI) cohort (individuals with mild cognitive impairment [MCI] and no cognitive impairment; September 2005-May 2022), and 2 Wisconsin cohorts, Wisconsin Alzheimer Disease Research Center and Wisconsin Registry for Alzheimer Prevention (WISC; individuals without cognitive impairment; February 2007-November 2020). This was a multicenter study, with data collected from referral centers in clinical routine (UGOT) and research settings (ADNI and WISC). Eligible individuals had 1 lumbar puncture (all cohorts), 2 or more cognitive assessments (ADNI and WISC), and imaging (ADNI only) performed on 2 separate occasions. Data were analyzed on August 2022 to April 2023. Exposures Baseline CSF Aβ42/40 and phosphorylated tau (p-tau)181; cognitive tests (ADNI: modified preclinical Alzheimer cognitive composite [mPACC]; WISC: modified 3-test PACC [PACC-3]). Exposures in the ADNI cohort included [18F]-florbetapir amyloid positron emission tomography (PET), magnetic resonance imaging (MRI), [18F]-fluorodeoxyglucose PET (FDG-PET), and cross-sectional tau-PET (ADNI: [18F]-flortaucipir, WISC: [18F]-MK6240). Main Outcomes and Measures Primary outcomes were the prevalence of CSF AT biomarker profiles and continuous longitudinal global cognitive outcome and imaging biomarker trajectories in A-T+ vs A-T- groups. Secondary outcomes included cross-sectional tau-PET. Results A total of 7679 individuals (mean [SD] age, 71.0 [8.4] years; 4101 male [53%]) were included in the UGOT cohort, 970 individuals (mean [SD] age, 73 [7.0] years; 526 male [54%]) were included in the ADNI cohort, and 519 individuals (mean [SD] age, 60 [7.3] years; 346 female [67%]) were included in the WISC cohort. The prevalence of an A-T+ profile in the UGOT cohort was 4.1% (95% CI, 3.7%-4.6%), being less common than the other patterns. Longitudinally, no significant differences in rates of worsening were observed between A-T+ and A-T- profiles for cognition or imaging biomarkers. Cross-sectionally, A-T+ had similar tau-PET uptake to individuals with an A-T- biomarker profile. Conclusion and Relevance Results suggest that the CSF A-T+ biomarker profile was found in approximately 5% of lumbar punctures and was not associated with a higher rate of cognitive decline or biomarker signs of disease progression compared with biomarker-negative individuals.
Collapse
Affiliation(s)
- Pontus Erickson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joel Simrén
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Wagner S. Brum
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gilda E. Ennis
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | | | | | - Rebecca Langhough
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Erin M. Jonaitis
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Carol A. Van Hulle
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Tobey J. Betthauser
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Cynthia M. Carlsson
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Sanjay Asthana
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Nicholas J. Ashton
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, King’s College London, London, England
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, England
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Sterling C. Johnson
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ulf Andreasson
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Barbara B. Bendlin
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison
- Institute of Neurology, Department of Neurodegenerative Disease, University College London, London, England
- UK Dementia Research Institute, University College London, London, England
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| |
Collapse
|
43
|
Xiong C, McCue LM, Buckles V, Grant E, Agboola F, Coble D, Bateman RJ, Fagan AM, Benzinger TL, Hassenstab J, Schindler SE, McDade E, Moulder K, Gordon BA, Cruchaga C, Day GS, Ikeuchi T, Suzuki K, Allegri RF, Vöglein J, Levin J, Morris JC. Cross-sectional and longitudinal comparisons of biomarkers and cognition among asymptomatic middle-aged individuals with a parental history of either autosomal dominant or late-onset Alzheimer's disease. Alzheimers Dement 2023; 19:2923-2932. [PMID: 36640138 PMCID: PMC10345163 DOI: 10.1002/alz.12912] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Comparisons of late-onset Alzheimer's disease (LOAD) and autosomal dominant AD (ADAD) are confounded by age. METHODS We compared biomarkers from cerebrospinal fluid (CSF), magnetic resonance imaging, and amyloid imaging with Pittsburgh Compound-B (PiB) across four groups of 387 cognitively normal participants, 42 to 65 years of age, in the Dominantly Inherited Alzheimer Network (DIAN) and the Adult Children Study (ACS) of LOAD: DIAN mutation carriers (MCs) and non-carriers (NON-MCs), and ACS participants with a positive (FH+) and negative (FH-) family history of LOAD. RESULTS At baseline, MCs had the lowest age-adjusted level of CSF Aβ42 and the highest levels of total and phosphorylated tau-181, and PiB uptake. Longitudinally, MC had similar increase in PiB uptake to FH+, but drastically faster decline in hippocampal volume than others, and was the only group showing cognitive decline. DISCUSSION Preclinical ADAD and LOAD share many biomarker signatures, but cross-sectional and longitudinal differences may exist.
Collapse
Affiliation(s)
- Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Lena M. McCue
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Virginia Buckles
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Elizabeth Grant
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Folasade Agboola
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Dean Coble
- Division of Biostatistics, Washington University, St. Louis, Missouri, USA
| | - Randall J. Bateman
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Anne M Fagan
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Tammie L.S. Benzinger
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Radiology, Washington University, St. Louis, Missouri, USA
- Department of Neurological Surgery, Washington University, St. Louis, Missouri, USA
| | - Jason Hassenstab
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
- Department of Psychology, Washington University, St. Louis, Missouri, USA
| | - Suzanne E. Schindler
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Eric McDade
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Krista Moulder
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | - Brian A. Gordon
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Psychology, Washington University, St. Louis, Missouri, USA
- Department of Radiology, Washington University, St. Louis, Missouri, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- Department of Psychiatry, Washington University, St. Louis, Missouri, USA
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, JAPAN
| | | | | | - Jonathan Vöglein
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - John C. Morris
- Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, USA
- The Dominantly Inherited Alzheimer Network, Washington University, St. Louis, Missouri, USA
- Department of Neurology, Washington University, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA
- Department of Physical Therapy, Washington University, St. Louis, Missouri, USA
- Department of Occupational Therapy, Washington University, St. Louis, Missouri, USA
| | | |
Collapse
|
44
|
Zavecz Z, Shah VD, Murillo OG, Vallat R, Mander BA, Winer JR, Jagust WJ, Walker MP. NREM sleep as a novel protective cognitive reserve factor in the face of Alzheimer's disease pathology. BMC Med 2023; 21:156. [PMID: 37138290 PMCID: PMC10155344 DOI: 10.1186/s12916-023-02811-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/28/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) pathology impairs cognitive function. Yet some individuals with high amounts of AD pathology suffer marked memory impairment, while others with the same degree of pathology burden show little impairment. Why is this? One proposed explanation is cognitive reserve i.e., factors that confer resilience against, or compensation for the effects of AD pathology. Deep NREM slow wave sleep (SWS) is recognized to enhance functions of learning and memory in healthy older adults. However, that the quality of NREM SWS (NREM slow wave activity, SWA) represents a novel cognitive reserve factor in older adults with AD pathology, thereby providing compensation against memory dysfunction otherwise caused by high AD pathology burden, remains unknown. METHODS Here, we tested this hypothesis in cognitively normal older adults (N = 62) by combining 11C-PiB (Pittsburgh compound B) positron emission tomography (PET) scanning for the quantification of β-amyloid (Aβ) with sleep electroencephalography (EEG) recordings to quantify NREM SWA and a hippocampal-dependent face-name learning task. RESULTS We demonstrated that NREM SWA significantly moderates the effect of Aβ status on memory function. Specifically, NREM SWA selectively supported superior memory function in individuals suffering high Aβ burden, i.e., those most in need of cognitive reserve (B = 2.694, p = 0.019). In contrast, those without significant Aβ pathological burden, and thus without the same need for cognitive reserve, did not similarly benefit from the presence of NREM SWA (B = -0.115, p = 0.876). This interaction between NREM SWA and Aβ status predicting memory function was significant after correcting for age, sex, Body Mass Index, gray matter atrophy, and previously identified cognitive reserve factors, such as education and physical activity (p = 0.042). CONCLUSIONS These findings indicate that NREM SWA is a novel cognitive reserve factor providing resilience against the memory impairment otherwise caused by high AD pathology burden. Furthermore, this cognitive reserve function of NREM SWA remained significant when accounting both for covariates, and factors previously linked to resilience, suggesting that sleep might be an independent cognitive reserve resource. Beyond such mechanistic insights are potential therapeutic implications. Unlike many other cognitive reserve factors (e.g., years of education, prior job complexity), sleep is a modifiable factor. As such, it represents an intervention possibility that may aid the preservation of cognitive function in the face of AD pathology, both present moment and longitudinally.
Collapse
Affiliation(s)
- Zsófia Zavecz
- Department of Psychology, Center for Human Sleep Science, University of California Berkeley, Berkeley, CA, 94720, USA.
| | - Vyoma D Shah
- Department of Psychology, Center for Human Sleep Science, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Olivia G Murillo
- Department of Psychology, Center for Human Sleep Science, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Raphael Vallat
- Department of Psychology, Center for Human Sleep Science, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, 92617, USA
| | - Joseph R Winer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Matthew P Walker
- Department of Psychology, Center for Human Sleep Science, University of California Berkeley, Berkeley, CA, 94720, USA.
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
45
|
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: 17] [Impact Index Per Article: 8.5] [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
|
46
|
Li JQ, Song JH, Suckling J, Wang YJ, Zuo CT, Zhang C, Gao J, Song YQ, Xie AM, Tan L, Yu JT. Disease trajectories in elders with suspected non-Alzheimer's pathophysiology and its comparison with Alzheimer's disease pathophysiology: a longitudinal study. RESEARCH SQUARE 2023:rs.3.rs-2744271. [PMID: 37034751 PMCID: PMC10081361 DOI: 10.21203/rs.3.rs-2744271/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Background According to the new 'AT(N)' system, those with a normal amyloid biomarker but with abnormal tauopathy or biomarkers of neurodegeneration or neuronal injury, have been labeled suspected non-Alzheimer's pathophysiology (SNAP). We aimed to estimate the long-term clinical and cognitive trajectories of SNAP individuals in non-demented elders and its comparison with individual in the Alzheimer's disease (AD) pathophysiology using 'AT(N)' system. Methods We included individuals with available baseline cerebrospinal fluid (CSF) Aβ (A), CSF phosphorylated tau examination (T) and 18F-uorodeoxyglucose PET or volumetric magnetic resonance imaging (N) from the Alzheimer's Disease Neuroimaging Initiative database. Longitudinal change in clinical outcomes are assessed using linear mixed effects models. Conversion risk from cognitively normal (CN) to cognitively impairment, and conversion from mild cognitive impairment (MCI) to dementia are assessed using multivariate Cox proportional hazard models. Results Totally, 366 SNAP individuals were included (114 A-T-N-, 154 A-T + N-, 54 A-T-N + and 44 A-T + N+) of whom 178 were CN and 188 were MCI. Compared with A-T-N-, CN elders with A-T + N-, A-T-N + and A-T + N + had a faster rate of ADNI-MEM score decline. Moreover, CN older individuals with A-T + N + also had a faster rate of decline in ADNI-MEM score than those with A-T + N- individuals. MCI patients with A-T + N + had a faster rate of ADNI-MEM and ADNI-EF decline and hippocampal volume loss compared with A-T-N- and A-T + N- profiles. CN older individuals with A-T + N + had an increased risk of conversion to cognitive impairment (CDR-GS ≥ 0.5) compared with A-T + N- and A-T-N-. In MCI patients, A-T + N + also had an increased risk of conversion to dementia compared with A-T + N- and A-T-N-. Compared with A-T + N-, CN elders and MCI patients with A + T + N- and A + T + N + had a faster rate of ADNI-MEM score, ADNI-EF score decline, and hippocampal volume loss. CN individuals with A + T + N + had a faster rate of ADNI-EF score decline compare with A-T + N + individuals. Moreover, MCI patients with A + T + N + also had a faster rate of decline in ADNI-MEM score, ADNI-EF score and hippocampal volume loss than those with A-T + N + individuals. Conclusions The findings from clinical, imaging and biomarker studies on SNAP, and its comparison with AD pathophysiology offered an important foundation for future studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Can Zhang
- Massachusetts General Hospital, Harvard Medical School
| | | | | | | | | | | |
Collapse
|
47
|
Liu KY, Thambisetty M, Howard R. How can secondary dementia prevention trials of Alzheimer's disease be clinically meaningful? Alzheimers Dement 2023; 19:1073-1085. [PMID: 36161763 PMCID: PMC10039957 DOI: 10.1002/alz.12788] [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: 04/21/2022] [Revised: 07/06/2022] [Accepted: 08/09/2022] [Indexed: 11/08/2022]
Abstract
After clinical trial failures in symptomatic Alzheimer's disease (AD), our field has moved to earlier intervention in cognitively normal individuals with biomarker evidence of AD. This offers potential for dementia prevention, but mainly low and variable rates of progression to AD dementia reduce the usefulness of trials' data in decision making by potential prescribers. With results from several Phase 3 secondary prevention studies anticipated within the next few years and the Food and Drug Administration's recent endorsement of amyloid beta as a surrogate outcome biomarker for AD clinical trials, it is time to question the clinical significance of changes in biomarkers, adequacy of current trial durations, and criteria for treatment success if cognitively unimpaired patients and their doctors are to meaningfully evaluate the potential value of new agents. We argue for a change of direction toward trial designs that can unambiguously inform clinical decision making about dementia risk and progression.
Collapse
Affiliation(s)
- Kathy Y. Liu
- Division of Psychiatry, University College London, London, UK
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, USA
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
48
|
Delvenne A, Gobom J, Tijms B, Bos I, Reus LM, Dobricic V, Kate MT, Verhey F, Ramakers I, Scheltens P, Teunissen CE, Vandenberghe R, Schaeverbeke J, Gabel S, Popp J, Peyratout G, Martinez-Lage P, Tainta M, Tsolaki M, Freund-Levi Y, Lovestone S, Streffer J, Barkhof F, Bertram L, Blennow K, Zetterberg H, Visser PJ, Vos SJB. Cerebrospinal fluid proteomic profiling of individuals with mild cognitive impairment and suspected non-Alzheimer's disease pathophysiology. Alzheimers Dement 2023; 19:807-820. [PMID: 35698882 DOI: 10.1002/alz.12713] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 04/06/2022] [Accepted: 05/12/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Suspected non-Alzheimer's disease pathophysiology (SNAP) is a biomarker concept that encompasses individuals with neuronal injury but without amyloidosis. We aim to investigate the pathophysiology of SNAP, defined as abnormal tau without amyloidosis, in individuals with mild cognitive impairment (MCI) by cerebrospinal fluid (CSF) proteomics. METHODS Individuals were classified based on CSF amyloid beta (Aβ)1-42 (A) and phosphorylated tau (T), as cognitively normal A-T- (CN), MCI A-T+ (MCI-SNAP), and MCI A+T+ (MCI-AD). Proteomics analyses, Gene Ontology (GO), brain cell expression, and gene expression analyses in brain regions of interest were performed. RESULTS A total of 96 proteins were decreased in MCI-SNAP compared to CN and MCI-AD. These proteins were enriched for extracellular matrix (ECM), hemostasis, immune system, protein processing/degradation, lipids, and synapse. Fifty-one percent were enriched for expression in the choroid plexus. CONCLUSION The pathophysiology of MCI-SNAP (A-T+) is distinct from that of MCI-AD. Our findings highlight the need for a different treatment in MCI-SNAP compared to MCI-AD.
Collapse
Affiliation(s)
- Aurore Delvenne
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Johan Gobom
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Betty Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands
| | - Isabelle Bos
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands
| | - Lianne M Reus
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
| | - Mara Ten Kate
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Frans Verhey
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Inez Ramakers
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Centers (AUMC), Amsterdam Neuroscience, the Netherlands
| | - Rik Vandenberghe
- Neurology Service, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Jolien Schaeverbeke
- Neurology Service, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Silvy Gabel
- Neurology Service, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Julius Popp
- Old Age Psychiatry, University Hospital Lausanne, Lausanne, Switzerland
- Department of Geriatric Psychiatry, Psychiatry University Hospital Zürich, Zürich, Switzerland
| | | | | | - Mikel Tainta
- Fundación CITA-Alzhéimer Fundazioa, San Sebastian, Spain
| | - Magda Tsolaki
- 1st Department of Neurology, AHEPA University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Makedonia, Thessaloniki, Greece
| | - Yvonne Freund-Levi
- Department of Neurobiology, Caring Sciences and Society (NVS), Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry in Region Örebro County and School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Old Age Psychiatry, Psychology & Neuroscience, King's College, London, UK
| | - Simon Lovestone
- University of Oxford, Oxford, United Kingdom (currently at Johnson and Johnson Medical Ltd.), London, UK
| | - Johannes Streffer
- Institute Born-Bunge, Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Belgium
- UCB Biopharma SPRL, Brain-l'Alleud, Belgium
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Institutes of Neurology & Healthcare Engineering, UCL London, London, UK
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
49
|
Zhao YL, Ou YN, Ma YH, Tan L, Yu JT. Characteristics of Subjective Cognitive Decline Associated with Alzheimer's Disease Amyloid Pathology: Findings from The CABLE Study. J Alzheimers Dis 2023; 92:581-590. [PMID: 36776070 DOI: 10.3233/jad-221154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
BACKGROUND Subjective cognitive decline (SCD) is considered as a preclinical hallmark of Alzheimer's disease (AD). However, the characteristics of SCD associated with amyloid pathology remain unclear. OBJECTIVE We aimed to explore the associations between SCD characteristics with amyloid pathology. METHODS Using logistic regression analyses, we analyzed the associations between cerebrospinal fluid (CSF) amyloid pathology with AD risk factors, SCD-specific characteristics (onset of SCD within the last five years, age at onset ≥60 years, feelings of worse performance, informant confirmation of complaints, worries, other domains of cognition complaints), as well as subthreshold depressive and anxiety symptoms among individuals with SCD. RESULTS A total of 535 SCD individuals with available CSF Aβ 42 information from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) study (mean age of 63.5 years, range 40 to 88 years; 47.10% female) were enrolled. The characteristics of informant confirmation of complaints (OR, 95% CI = 2.00, 1.19-3.36), subthreshold depressive symptoms (OR, 95% CI = 2.31, 1.05-5.09), and subthreshold anxiety symptoms (OR, 95% CI = 2.22, 1.09-4.51) were found to be significantly associated with pathological amyloid in multivariate analyses when adjusting for age, sex, education, and APOE ɛ4. Besides, age and females were observed risks for amyloid pathology in subscale analyses. Nonetheless, we did not find any associations of other SCD-specific characteristics with amyloid pathology in this study. CONCLUSION Our study suggested that informant confirmed complaints and subthreshold psychiatric symptoms might be critical for discriminating AD-related SCD from non-AD related SCD.
Collapse
Affiliation(s)
- Yong-Li Zhao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Rådestig MA, Skoog J, Zetterberg H, Skillbäck T, Zettergren A, Sterner TR, Fässberg MM, Sacuiu S, Waern M, Wetterberg H, Blennow K, Skoog I, Kern S. Subtle Differences in Cognition in 70-Year-Olds with Elevated Cerebrospinal Fluid Neurofilament Light and Neurogranin: A H70 Cross-Sectional Study. J Alzheimers Dis 2023; 91:291-303. [PMID: 36617786 PMCID: PMC9881027 DOI: 10.3233/jad-220452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Most research on cerebrospinal fluid (CSF) neurofilament light protein (NfL) as a marker for neurodegeneration and neurogranin (Ng) for synaptic dysfunction has largely focused on clinical cohorts rather than population-based samples. OBJECTIVE We hypothesized that increased CSF levels of NfL and Ng are associated with subtle cognitive deficits in cognitively unimpaired (CU) older adults. METHODS The sample was derived from the Gothenburg H70 Birth Cohort Studies and comprised 258 CU 70-year-olds, with a Clinical Dementia Rating score of zero. All participants underwent extensive cognitive testing. CSF levels of NfL and Ng, as well as amyloid β1 - 42, total tau, and phosphorylated tau, were measured. RESULTS Participants with high CSF NfL performed worse in one memory-based test (Immediate recall, p = 0.013) and a language test (FAS, p = 0.016). Individuals with high CSF Ng performed worse on the memory-based test Supra Span (p = 0.035). When stratified according to CSF tau and Aβ42 concentrations, participants with high NfL and increased tau performed worse on a memory test than participants normal tau concentrations (Delayed recall, p = 0.003). In participants with high NfL, those with pathologic Aβ42 concentrations performed worse on the Delayed recall memory (p = 0.044). In the high Ng group, participants with pathological Aβ42 concentrations had lower MMSE scores (p = 0.027). However, in regression analysis we found no linear correlations between CSF NfL or CSF Ng in relation to cognitive tests when controlled for important co-variates. CONCLUSION Markers of neurodegeneration and synaptic pathology might be associated with subtle signs of cognitive decline in a population-based sample of 70-year-olds.
Collapse
Affiliation(s)
- Maya Arvidsson Rådestig
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Johan Skoog
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychology, University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UCL Institute of Neurology, Queen Square, London, UK
- The UK Dementia Research Institute, UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Tobias Skillbäck
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Anna Zettergren
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Therese Rydberg Sterner
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Madeleine Mellqvist Fässberg
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Simona Sacuiu
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Memory Disorders Clinic, Theme Inflammation and Aging, Karolinska University Hospital, Region Stockholm, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society (NVS), Clinical Geriatric, Karolinska Institute, Stockholm, Sweden
| | - Margda Waern
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychosis Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Hanna Wetterberg
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ingmar Skoog
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Silke Kern
- Center for Ageing and Health (AgeCap), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Psychiatry/Cognition and Old Age Psychiatry Clinic, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| |
Collapse
|